1
|
Kalra V, Passi GR. Hypoglycorrhachia: Unravelling the Pathogenetic Mechanisms. Indian Pediatr 2025:10.1007/s13312-025-00057-x. [PMID: 40244533 DOI: 10.1007/s13312-025-00057-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 04/18/2025]
Affiliation(s)
- Veena Kalra
- Department of Pediatrics, All India Institute of Medical Sciences, 101, Jorbhagh, New Delhi, India.
- Pediatric Neurology, Indraprastha Apollo, New Delhi, India.
- The Child Centre, Delhi, India.
| | - Gouri Rao Passi
- Pediatric Neurology, Choithram Hospital & Research Centre, Indore, Madhya Pradesh, 452014, India
| |
Collapse
|
2
|
Tang M, Teng S, Kim AY, Peng Y, Monani UR. A transgene harboring the human Glucose Transporter1 (GLUT1) gene locus ameliorates disease in GLUT1 deficiency syndrome model mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.05.647372. [PMID: 40236204 PMCID: PMC11996456 DOI: 10.1101/2025.04.05.647372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Proper brain function relies on an adequate supply of energy - mainly glucose - to power neuronal activity. Delivery of this nutrient to the neuropil is mediated by the Glucose Transporter1 (GLUT1) protein. Perturbing glucose supply to the brain is profoundly damaging and exemplified by the neurodevelopmental disorder, GLUT1 deficiency syndrome (GLUT1DS). Resulting from haploinsufficiency of the SLC2A1 (GLUT1) gene, GLUT1DS is characterized by intractable infantile-onset seizures and a disabling movement disorder. Ketogenic diets, which supply the brain with an alternate energy source, ketone bodies, are currently the preferred therapeutic option for Glut1DS patients but do not address the underlying cause - low brain glucose - of the disease. One intuitively appealing therapeutic strategy that does, involves restoring GLUT1 levels to the patient brain. Here, we demonstrate that transgenic expression of the human GLUT1 genomic locus in a mouse model of GLUT1DS raises brain GLUT1 levels and reduces disease burden. Augmenting GLUT1 levels in mutants correspondingly raised cerebrospinal fluid (CSF) glucose levels, improved motor performance and reduced the frequency of seizures characteristically observed in GLUT1DS. Interestingly, the increased GLUT1 in mutants harboring the human GLUT1 locus was at least partly the result of an increase in murine Slc2a1 (Glut1) activity, most likely the effect of a long non-coding RNA (lncRNA) embedded in the human transgene. Collectively, our work has not only shown that repleting human GLUT1 mitigates GLUT1DS but also has yielded transgenic mice that constitute a useful tool to test and optimize clinically promising agents designed to stimulate this gene for therapeutic purposes.
Collapse
|
3
|
Lehner‐Gulotta D, Blackford R, Bessone S, Lowman W, Haggerty N, Huang C, Axeen ET. Practical experience and challenges in nutritional management of glucose transporter 1 deficiency syndrome: Provider survey results. Epilepsia Open 2025; 10:628-634. [PMID: 39918171 PMCID: PMC12014909 DOI: 10.1002/epi4.13135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 04/24/2025] Open
Abstract
Ketogenic diets (KDs) are recommended as precision therapy for glucose transporter 1 deficiency syndrome (GLUT-1 DS) but there are no recommendations for optimal implementation in this population. We administered an online survey targeting clinicians with experience implementing KDs in GLUT-1 DS, focusing on diet selection, initiation, and management. Respondents were primarily experienced registered dietitian nutritionists (RDNs) from 34 centers in 10 countries. Most reported a preference for carbohydrate counting or ratios under 3:1. KD-related laboratory monitoring (including blood ketones) and vitamin/mineral supplementation did not differ for GLUT-1 DS patients compared to the epilepsy population. Routine use of exogenous ketones was not endorsed for improved ketosis, whereas MCT oil is a commonly used supplement. Respondents overwhelmingly endorsed discussing and implementing gastrostomy feedings to support continued dietary therapy when medically indicated. Most but not all providers (80%) were familiar with the 2020 consensus guidelines. Our survey demonstrates practice variability among experienced dietitians, particularly in diet type and ratio. Identified challenges carry clinical significance, as the diet is a precision therapy in GLUT1-DS. Further research is needed to examine the outcomes of different approaches to KDs in GLUT-1 DS before consensus about the most effective interventions can be reached. PLAIN LANGUAGE SUMMARY: Glucose transporter deficiency syndrome is a genetic condition caused by an inability to move sugar (glucose) into the brain, which is needed for proper brain function. Ketogenic diets (low in carbohydrate and high in fat) are the established treatment to help control symptoms. Although the diet is effective, it can be challenging. To understand these challenges, we surveyed experts in ketogenic diet management and found significant variability regarding specifics of how the diet is managed. More research is needed before one approach can be endorsed as most effective.
Collapse
Affiliation(s)
- Diana Lehner‐Gulotta
- Department of Clinical NutritionUniversity of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Robyn Blackford
- Ann & Robert H. Lurie Children's Hospital of Chicago, Clinical NutritionChicagoIllinoisUSA
| | - Stacey Bessone
- Johns Hopkins All Children's Hospital, Nutritional ServicesSt. PetersburgFloridaUSA
| | - Wesley Lowman
- Ann & Robert H. Lurie Children's Hospital of Chicago, Clinical NutritionChicagoIllinoisUSA
| | - Nicole Haggerty
- Ann & Robert H. Lurie Children's Hospital of Chicago, Clinical NutritionChicagoIllinoisUSA
| | - Chu‐Yi Huang
- Ann & Robert H. Lurie Children's Hospital of Chicago, Clinical NutritionChicagoIllinoisUSA
| | - Erika T. Axeen
- Department of Neurology, Division of Child NeurologyUniversity of Virginia Health SystemCharlottesvilleVirginiaUSA
| | | |
Collapse
|
4
|
Coggan JS, Shichkova P, Markram H, Keller D. Seizure and redox rescue in a model of glucose transport deficiency. PLoS Comput Biol 2025; 21:e1012959. [PMID: 40184423 PMCID: PMC12002639 DOI: 10.1371/journal.pcbi.1012959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 04/16/2025] [Accepted: 03/12/2025] [Indexed: 04/06/2025] Open
Abstract
Disruptions of energy supply to the brain are associated with many neurodegenerative pathologies and are difficult to study due to numerous interlinked metabolic pathways. We explored the effects of diminished energy supply on brain metabolism using a computational model of the neuro-glia-vasculature ensemble, in the form of a neuron, an astrocyte and local blood supply. As a case study, we investigated the glucose transporter type-1 deficiency syndrome (GLUT1-DS), a childhood affliction characterized by impaired glucose utilization and associated with phenotypes including seizures. Compared to neurons, astrocytes exhibited markedly higher metabolite concentration variabilities for all but a few redox species. This effect could signal a role for astrocytes in absorbing the shock of blood nutrient fluctuations. Redox balances were disrupted in GLUT1-DS with lower levels of reducing equivalent carriers NADH and ATP. The best non-glucose nutrient or pharmacotherapies for re-establishing redox normalcy involved lactate, the keto-diet (β-hydroxybutyrate), NAD and Q10 supplementation, suggesting a possible glucose sparing mechanism. GLUT1-DS seizures resulted from after-discharge neuronal firing caused by post-stimulus ATP reductions and impaired Na+/K+-ATPase, which can be rescued by restoring either normal glucose or by relatively small increases in neuronal ATP.
Collapse
Affiliation(s)
- Jay S. Coggan
- Blue Brain Project, EPFL: École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | - Polina Shichkova
- Blue Brain Project, EPFL: École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
- Biognosys AG, Schlieren, Switzerland
| | - Henry Markram
- Blue Brain Project, EPFL: École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| | - Daniel Keller
- Blue Brain Project, EPFL: École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
| |
Collapse
|
5
|
Tang M, Teng S, Peng Y, Kim AY, Canoll P, Bruce JN, Faust PL, Adhikari K, De Vivo DC, Monani UR. A therapeutic role for a regulatory glucose transporter1 (Glut1)-associated natural antisense transcript. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.644647. [PMID: 40196663 PMCID: PMC11974780 DOI: 10.1101/2025.03.26.644647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The mammalian brain relies primarily on glucose for its energy needs. Delivery of this nutrient to the brain is mediated by the glucose transporter-1 (Glut1) protein. Low Glut1 thwarts glucose entry into the brain, causing an energy crisis and, triggering, in one instance, the debilitating neurodevelopmental condition - Glut1 deficiency syndrome (Glut1DS). Current treatments for Glut1DS are sub-optimal, as none address the root cause - low Glut1 - of the condition. Levels of this transporter must respond rapidly to the brain's changing energy requirements. This necessitates fine-tuning its expression. Here we describe a long-noncoding RNA (lncRNA) antisense to Glut1 and show that it is involved in such regulation. Raising levels of the lncRNA had a concordant effect on Glut1 in cultured human cells and transgenic mice; reducing levels elicited the opposite effect. Delivering the lncRNA to Glut1DS model mice via viral vectors induced Glut1 expression, enhancing brain glucose levels to mitigate disease. Direct delivery of such a lncRNA to combat disease has not been reported previously and constitutes a unique therapeutic paradigm. Moreover, considering the importance of maintaining homeostatic Glut1 levels, calibrating transporter expression via the lncRNA could become broadly relevant to the myriad conditions, including Alzheimer's disease, wherein Glut1 concentrations are perturbed.
Collapse
|
6
|
Zhang MJ, Zhang SM, Zhang QP, Wen YX, Wang JP, Jiang YW, Bao XH. Clinical and genetic characteristics of glucose transporter 1 deficiency syndrome in a large cohort of Chinese patients. World J Pediatr 2025; 21:274-283. [PMID: 40048124 PMCID: PMC11958367 DOI: 10.1007/s12519-025-00884-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Mutations in the SLC2A1 gene cause glucose transporter type 1 deficiency syndrome (Glut1DS). This study aimed to investigate the clinical and molecular genetics characteristics of Chinese patients with Glut1DS. METHODS The clinical data of patients with Glut1DS were analyzed retrospectively. SLC2A1 mutation analysis was performed using Sanger sequencing or next-generation sequencing (NGS). Multiplex ligation-dependent probe amplification (MLPA) was conducted in patients with negative results. RESULTS A total of 90 patients were diagnosed with Glut1DS, including 63 (70%) classic type and 27 (30%) non-classic type. Seizures occurred in 69 patients (77%), movement disorders were observed in 58 (68%), and episodic eye-head movements were noted in 17 (19%). Cerebrospinal fluid (CSF) glucose levels were available for 73 patients (81%), ranging from 1.0 to 2.6 mmol/L (median 1.9 mmol/L), with 90% (66/73) of patients showing levels below 2.2 mmol/L. Additionally, CSF-to-blood glucose ratios measured in 71 patients (79%) ranged from 0.20 to 0.63 (median 0.37), with 87% (62/71) of patients having ratios below 0.45. Genetic analysis identified 69 variants of the SLC2A1 gene including 39 previously reported and 30 unreported variants. The two most common variants were c.997C > T (p.Arg333Trp) and c.988C > T (p.Arg330*). Following ketogenic diet therapy, seizures were controlled in 47 of 57 patients (82%), movement disorders resolved in 18 of 47 patients (38%), and improved in 26 of 47 patients (55%). CONCLUSIONS The clinical manifestations of Glut1DS primarily include seizures, movement disorders, and developmental delay. Most affected children had CSF glucose levels below 2.2 mmol/L, with CSF-to-blood glucose ratios under 0.45. Two of the most common SLC2A1 variants were identified in our cohort. Ketogenic diet therapy was effective in controlling seizures, improving movement disorders, and was well tolerated.
Collapse
Affiliation(s)
- Mei-Jiao Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Shi-Min Zhang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | - Qing-Ping Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yong-Xin Wen
- Department of Pediatric Neurology, Guangdong Women and Children Hospital, Guangdong, China
| | - Jia-Ping Wang
- Department of Neurology, Beijing Children'S Hospital, Capital Medical University, Beijing, China
| | - Yu-Wu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xin-Hua Bao
- Department of Pediatrics, Peking University First Hospital, Beijing, China.
| |
Collapse
|
7
|
Zhang MJ, Wu D, Yu LF, Li H, Sun D, Liang JM, Lu XP, Luo R, Guo QH, Jin RF, Zhang HW, Lei GF, Sun RP, Wang M, Zhou YF, Wang YY, Tang JH, Hua Y, Shi XL, Liu XM, Shi XY, Yang G, Wang H, Gao F, Jia TM, Wang JW, Liao JX, Bao XH. Diagnosis and treatment recommendations for glucose transporter 1 deficiency syndrome. World J Pediatr 2025; 21:149-158. [PMID: 39745620 PMCID: PMC11885374 DOI: 10.1007/s12519-024-00864-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Glucose transporter 1 deficiency syndrome (Glut1DS) was initially reported by De Vivo and colleagues in 1991. This disease arises from mutations in the SLC2A1 and presents with a broad clinical spectrum. It is a treatable neuro-metabolic condition, where prompt diagnosis and initiation of ketogenic dietary therapy can markedly enhance the prognosis. However, due to its rarity, Glut1DS is susceptible to misdiagnosis or missed diagnosis, which can lead to delayed treatment and irreversible dysfunction of the central nervous system. To promote diagnostic awareness and effective treatments, the recommendations for diagnosis and treatment have been developed. METHODS The panel on Glut1DS included 28 participants from the members of the Ketogenic Diet Professional Committee of the Chinese Epilepsy Association and Chinese experts with extensive experience in managing Glut1DS. All authors extensively reviewed the literature, and the survey results were discussed in detail over several online meetings. Following multiple deliberative sessions, all participants approved the final manuscript for submission. RESULTS Early diagnosis and timely treatment of Glut1DS are crucial for improving prognosis. Physicians should be alert to suspiction of this disease if the following clinical manifestations appear: seizures, episodic or persistent movement disorders (often triggered by fasting, fatigue, or exercise), delayed motor and cognitive development. Characteristic clinical presentations may include seizures combined with movement disorders, episodic eye-head movements, and paroxysmal exercise-induced dyskinesia (PED). In these cases, genetic testing should be promptly completed, and a lumbar puncture should be performed if necessary. The ketogenic diet is internationally recognized as the first-line treatment; the earlier it is started, the better the prognosis. It can effectively control seizures and improve motor disorders. Antiepileptic drug treatment is generally ineffective or provides limited symptom improvement before starting the ketogenic diet. CONCLUSION The recommendations provide clinicians with a relatively systematic guide for the rapid identification, diagnosis, and timely treatment of Glut1DS.
Collapse
Affiliation(s)
| | - De Wu
- The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Li-Fei Yu
- Children's Hospital of Fudan University, Shanghai, China
| | - Hua Li
- Guangdong Sanjiu Brain Hospital, Guangdong, China
| | - Dan Sun
- Huazhong University of Science and Technology Tongji Medical College Affiliated Wuhan Children's Hospital, Wuhan, China
| | - Jian-Min Liang
- Department of Pediatric Neurology, Children's Medical Center, The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Xiao-Peng Lu
- Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Luo
- Second Hospital of West China of Sichuan University, Sichuan, China
| | - Qing-Hui Guo
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong, China
| | - Rui-Feng Jin
- Children's Hospital Affiliated to Shandong University, Shandong, China
| | - Hong-Wei Zhang
- Children's Hospital Affiliated to Shandong University, Shandong, China
| | - Ge-Fei Lei
- Qilu Hospital of Shandong University, Shandong, China
| | - Ruo-Peng Sun
- Qilu Hospital of Shandong University, Shandong, China
| | - Man Wang
- Shanghai Deji Hospital, Shanghai, China
| | - You-Feng Zhou
- Fujian Children's Hospital (Fujian Branch of Shanghai Children's Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China
| | - Ying-Yan Wang
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ji-Hong Tang
- Children's Hospital of Soochow University, Jiangsu, China
| | - Ying Hua
- Wuxi Children's Hospital, Jiangsu, China
| | - Xu-Lai Shi
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Zhejiang, China
| | | | - Xiu-Yu Shi
- General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Guang Yang
- General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Hua Wang
- Shengjing Hospital of China Medical University, Liaoning, China
| | - Feng Gao
- Children's Hospital affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-Ming Jia
- The Third Affiliated Hospital of Zhengzhou University, Henan, China
| | - Ji-Wen Wang
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | | | - Xin-Hua Bao
- The First Hospital of Peking University, Beijing, China.
| |
Collapse
|
8
|
Abend NS, Wusthoff CJ, Jensen FE, Inder TE, Volpe JJ. Neonatal Seizures. VOLPE'S NEUROLOGY OF THE NEWBORN 2025:381-448.e17. [DOI: 10.1016/b978-0-443-10513-5.00015-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Elitt CM, Volpe JJ. Degenerative Disorders of the Newborn. VOLPE'S NEUROLOGY OF THE NEWBORN 2025:967-1007.e17. [DOI: 10.1016/b978-0-443-10513-5.00033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
10
|
Amer A, Murrell K, Edmonds L, Bernhardt I, Akroyd R, Ryder B, Wilson C, Glamuzina E. D,L-3-hydroxybutyrate in the treatment of glucose transporter 1 deficiency syndrome (Glut1DS). JIMD Rep 2025; 66:e12461. [PMID: 39830115 PMCID: PMC11739118 DOI: 10.1002/jmd2.12461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 01/22/2025] Open
Abstract
Background Deficiency of the Glut1 transporter due to mono-allelic variants in SLC2A1 causes hypoglycorrhachia, resulting in a neurological spectrum from neonatal epilepsy to adult-onset paroxysmal movement disorders (PMD). The brain utilises ketone bodies as an alternative energy source to glucose. Thus, early initiation of the ketogenic diet (KD) is standard care for Glut1 deficiency syndrome (Glut1DS). Commencement and adherence in older Glut1DS patients is difficult to achieve, leaving few treatment options. Oral D,L-3-hydroxybutyrate (D,L-3-HB) crosses the blood-brain barrier, making it a potential treatment for Glut1DS. Methods A retrospective case review of patients with Glut1DS under the Adult and Paediatric National Metabolic Service (APNMS) of New Zealand, treated with D,L-3-HB between 2012 and 2023 was performed. Clinical notes, standardised, neuropsychological assessments and subjective data on and off D,L-3-HB were obtained. The best on and off D,L-3-HB measures of working memory (WMI) and processing speed (PSI) were compared to assess the efficacy. Results D,L-3-HB was offered to 12 patients with Glut1DS (age 10-52 years). Compliance-dependent improvements in subjective, cognitive and adaptive function were reported by those who were reassessed on-treatment (9/12). Four reported improved PMD. Objective improvements were found in WM (9/9) and PS (6/9). Subjective improvements were reported in patients' health, wellbeing and independence. Conclusions KD remains standard of care for Glut1DS, but effective alternatives are lacking for those who do not tolerate this. D,L-3-HB was associated with improved WM, PS and perceived life quality in this small group of patients with Glut1DS, thus providing a potential treatment for this distinct group.
Collapse
Affiliation(s)
- Aya Amer
- Dunedin HospitalTe Whatu Ora Health New Zealand, SouthernDunedinNew Zealand
| | - Kathryn Murrell
- Adult and Paediatric National Metabolic ServiceStarship Children's Hospital, Te Toka Tumai, Te Whatu Ora Health New Zealand Tāmaki MakaurauAucklandNew Zealand
| | - Liza Edmonds
- Te Tātai Hauora o Hine Victoria UniversityWellingtonNew Zealand
| | - Isaac Bernhardt
- Adult and Paediatric National Metabolic ServiceStarship Children's Hospital, Te Toka Tumai, Te Whatu Ora Health New Zealand Tāmaki MakaurauAucklandNew Zealand
| | - Rhonda Akroyd
- Adult and Paediatric National Metabolic ServiceStarship Children's Hospital, Te Toka Tumai, Te Whatu Ora Health New Zealand Tāmaki MakaurauAucklandNew Zealand
| | - Bryony Ryder
- Adult and Paediatric National Metabolic ServiceStarship Children's Hospital, Te Toka Tumai, Te Whatu Ora Health New Zealand Tāmaki MakaurauAucklandNew Zealand
| | - Callum Wilson
- Adult and Paediatric National Metabolic ServiceStarship Children's Hospital, Te Toka Tumai, Te Whatu Ora Health New Zealand Tāmaki MakaurauAucklandNew Zealand
| | - Emma Glamuzina
- Adult and Paediatric National Metabolic ServiceStarship Children's Hospital, Te Toka Tumai, Te Whatu Ora Health New Zealand Tāmaki MakaurauAucklandNew Zealand
| |
Collapse
|
11
|
Pervaiz I, Mehta Y, Al-Ahmad AJ. Glucose Transporter 1 Deficiency Impairs Glucose Metabolism and Barrier Induction in Human Induced Pluripotent Stem Cell-Derived Astrocytes. J Cell Physiol 2025; 240:e31523. [PMID: 39807611 DOI: 10.1002/jcp.31523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025]
Abstract
Glucose is a major source of energy for the brain. At the blood-brain barrier (BBB), glucose uptake is facilitated by glucose transporter 1 (GLUT1). GLUT1 Deficiency Syndrome (GLUT1DS), a haploinsufficiency affecting SLC2A1, reduces glucose brain uptake. A lot of effort has been made to characterize GLUT1DS at the BBB, but the impact on astrocytes remains unclear. In this study, we investigated the impact of GLUT1DS on astrocyte differentiation and function in vitro, using human induced pluripotent stem cells GLUT1DS (GLUT1DS-iPSCs) differentiated into astrocyte-like cells (iAstros). GLUT1 expression is decreased during the differentiation of iPSCs into astrocytes, with neural progenitor cells showing the lowest expression. The presence of a truncated GLUT1 did not compromise the differentiation of iPSCs into iAstros, as these cells could express several key markers representative of the astrocyte lineage. GLUT1DS-iAstros failed to express full-length GLUT1 at protein levels while showing no signs of impaired GLUT4 expression. However, GLUT1DS-iAstros showed decreased glucose uptake and lactate production compared to control-iAstros, reduced glycolysis, and mitochondrial activity as well as ATP deficit. In addition to reduced energy production, astrocytes displayed a reduced extracellular glutamate release. As previously observed, one iAstros clone (C7) showed the most severe phenotype from all groups. Our study provides an insightful view of the contribution of GLUT1 in astrocytes' energetic metabolism and raises the possible contribution of these cells in the astrocyte-neuron metabolic coupling. Our future direction is to understand better how GLUT1DS impacts astrocytes and neurons within their metabolic coupling.
Collapse
Affiliation(s)
- Iqra Pervaiz
- Department of Pharmaceutical Sciences and Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, USA
| | - Yash Mehta
- Department of Pharmaceutical Sciences and Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, USA
| | - Abraham Jacob Al-Ahmad
- Department of Pharmaceutical Sciences and Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, USA
| |
Collapse
|
12
|
Furtado J, Geraldo LH, Leser FS, Bartkowiak B, Poulet M, Park H, Robinson M, Pibouin-Fragner L, Eichmann A, Boyé K. Interplay between Netrin-1 and Norrin controls arteriovenous zonation of blood-retina barrier integrity. Proc Natl Acad Sci U S A 2024; 121:e2408674121. [PMID: 39693351 DOI: 10.1073/pnas.2408674121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/26/2024] [Indexed: 12/20/2024] Open
Abstract
The integrity of the blood-retina barrier (BRB) is crucial for phototransduction and vision, by tightly restricting transport of molecules between the blood and surrounding neuronal cells. Breakdown of the BRB leads to the development of retinal diseases. Here, we show that Netrin-1/Unc5b and Norrin/Lrp5 signaling establish a zonated endothelial cell gene expression program that controls BRB integrity. Using single-cell RNA sequencing (scRNA-seq) of postnatal BRB-competent mouse retina endothelial cells (ECs), we identify >100 BRB genes encoding Wnt signaling components, tight junction proteins, and ion and nutrient transporters. We find that BRB gene expression is zonated across arteries, capillaries, and veins and regulated by opposing gradients of the Netrin-1 receptor Unc5b and Lrp5-β-catenin signaling between retinal arterioles and venules. Mice deficient for Ntn1 or Unc5b display more BRB leakage at the arterial end of the vasculature, while Lrp5 loss of function causes predominantly venular BRB leakage. ScRNA-seq of Ntn1 and Unc5b mutant ECs reveals down-regulated β-catenin signaling and BRB gene expression that is rescued by Ctnnb1 overactivation, along with BRB integrity. Mechanistically, we demonstrate that Netrin-1 and Norrin additively enhance β-catenin transcriptional activity and Lrp5 phosphorylation via the Discs large homologue 1 (Dlg1) scaffolding protein, and endothelial Lrp5-Unc5b function converges in protection of capillary BRB integrity. These findings explain how arteriovenous zonation is established and maintained in the BRB and reveal that BRB gene expression is regulated at the level of endothelial subtypes.
Collapse
Affiliation(s)
- Jessica Furtado
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT 06511
| | - Luiz Henrique Geraldo
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT 06511
| | - Felipe Saceanu Leser
- Paris Cardiovascular Research Center, Université Paris Cité, Inserm U970, Paris F-75015, France
| | - Bartlomiej Bartkowiak
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT 06511
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06511
| | - Mathilde Poulet
- Paris Cardiovascular Research Center, Université Paris Cité, Inserm U970, Paris F-75015, France
| | - Hyojin Park
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT 06511
| | - Mark Robinson
- Center of Molecular and Cellular Oncology, Department of Internal Medicine, Yale University, School of Medicine, New Haven CT 06511
| | | | - Anne Eichmann
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT 06511
- Paris Cardiovascular Research Center, Université Paris Cité, Inserm U970, Paris F-75015, France
| | - Kevin Boyé
- Paris Cardiovascular Research Center, Université Paris Cité, Inserm U970, Paris F-75015, France
| |
Collapse
|
13
|
Li R, Tsuboi H, Ito H, Takagi D, Chang YH, Shimizu T, Arai Y, Matsuo-Takasaki M, Noguchi M, Nakamura Y, Ohnuma K, Takahashi S, Hayashi Y. Generation of human induced pluripotent stem cell lines derived from two glucose transporter 1 deficiency syndrome patients. Stem Cell Res 2024; 81:103584. [PMID: 39490212 DOI: 10.1016/j.scr.2024.103584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 11/05/2024] Open
Abstract
Glucose transporter 1 deficiency syndrome (GLUT1DS), caused by impaired glucose transport at the blood-brain barriers, leads to various central nervous system dysfunctions. A comprehensive understanding of the underlying disease pathogenesis is still lacking. In this study, we have generated GLUT1DS-specific human induced pluripotent stem cells (hiPSCs) derived from two patients. These established GLUT1DS-specific hiPSC lines showed self-renewal and pluripotency and carried heterozygous frameshift or missense mutations in the responsible SLC2A1 gene. These novel cell resources provide new avenues for understanding disease mechanisms and developing new therapies for GLUT1DS.
Collapse
Affiliation(s)
- Rui Li
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan; School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Hazuki Tsuboi
- Department of Materials Science and Bioengineering, Nagaoka University of Technology, 1603-1 Kami-Tomioka, Nagaoka, Niigata 940-2188, Japan
| | - Hidenori Ito
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Daigo Takagi
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Yun-Hsuan Chang
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Tomoya Shimizu
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Yutaka Arai
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Mami Matsuo-Takasaki
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Michiya Noguchi
- Cell Engineering Division, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Yukio Nakamura
- Cell Engineering Division, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Kiyoshi Ohnuma
- Department of Materials Science and Bioengineering, Nagaoka University of Technology, 1603-1 Kami-Tomioka, Nagaoka, Niigata 940-2188, Japan; Department of Science of Technology Innovation, Nagaoka University of Technology, 1603-1 Kami-Tomioka, Nagaoka, Niigata 940-2188, Japan
| | - Satoru Takahashi
- Department of Pediatrics, Asahikawa Medical University, 2-1-1-1 Midorigaoka Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Yohei Hayashi
- iPS Cell Advanced Characterization and Development Team, BioResource Research Center, RIKEN, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan; School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan.
| |
Collapse
|
14
|
Sobrinho LMF, Silva TO, Refosco LF, Poloni S, Poswar FO, de Souza CFM, Sperb-Ludwig F, Schwartz IVD. A novel frameshift variant in the SLC2A1 gene causing a mild phenotype of GLUT1 deficiency syndrome: case report. Mol Genet Metab Rep 2024; 41:101164. [PMID: 39629096 PMCID: PMC11612342 DOI: 10.1016/j.ymgmr.2024.101164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 12/06/2024] Open
Abstract
Glucose transporter type 1 deficiency syndrome (GLUT1) is a genetic condition, most often of autosomal dominant inheritance, and corresponds to a broad spectrum of signs and symptoms due to hypoglycorrhachia, which include seizures, delay in neuropsychomotor development, intellectual disability, movement disorders, dysarthria and postnatal microcephaly. The severity of symptoms are variable. Symptomatic treatment consists of the ketogenic diet, which allows energy supply to the brain through sustained and continuous ketosis. In this study, we report a novel heterozygous frameshift variant (c.855_856insTT; p.Gly286Leufs*55) in the SLC2A1 gene in a preschool Brazilian child with atypical phenotype of GLUT1 deficiency syndrome, characterized by ataxia and mild speech delay. Our study enriches the SLC2A1 gene mutation spectrum and emphasizes the importance of molecular genetic studies for screening patients with neuropsychomotor developmental delay. Sentence take-home message (synopsis) of the article: The study enriches the SLC2A1 gene mutation spectrum and emphasizes the importance of molecular genetic studies for screening patients with neuropsychomotor developmental delay.
Collapse
Affiliation(s)
- Lívia Maria Ferreira Sobrinho
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
- Department of Pediatrics, School of Medicine, Federal University of Minas Gerais, Minas Gerais, Brazil
| | - Thiago Oliveira Silva
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Lilia Farret Refosco
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Soraia Poloni
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Fabiano Oliveira Poswar
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Fernanda Sperb-Ludwig
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
- Clinical Research Center, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
- BRAIN Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
| | - Ida Vanessa Doederlein Schwartz
- Postgraduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
- BRAIN Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Rio Grande do Sul, Brazil
- InRaras – Instituto Nacional de Ciencia e Tecnologia em Doenças Raras, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
15
|
Banks WA, Rhea EM, Reed MJ, Erickson MA. The penetration of therapeutics across the blood-brain barrier: Classic case studies and clinical implications. Cell Rep Med 2024; 5:101760. [PMID: 39383873 PMCID: PMC11604479 DOI: 10.1016/j.xcrm.2024.101760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/20/2024] [Accepted: 09/11/2024] [Indexed: 10/11/2024]
Abstract
The blood-brain barrier (BBB) plays central roles in the maintenance and health of the brain. Its mechanisms to safeguard the brain against xenobiotics and endogenous toxins also make the BBB the primary obstacle to the development of drugs for the central nervous system (CNS). Here, we review classic examples of the intersection of clinical medicine, drug delivery, and the BBB. We highlight the role of lipid solubility (heroin), saturable brain-to-blood (efflux: opiates) and blood-to-brain (influx: nutrients, vitamins, and minerals) transport systems, and adsorptive transcytosis (viruses and incretin receptor agonists). We examine how the disruption of the BBB that occurs in certain diseases (tumors) can also be modulated (osmotic agents and microbubbles) and used to deliver treatments, and the role of extracellular pathways in gaining access to the CNS (albumin and antibodies). In summary, this review provides a historical perspective of the key role of the BBB in delivery of drugs to the brain in health and disease.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA.
| | - Elizabeth M Rhea
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - May J Reed
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| | - Michelle A Erickson
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98104, USA
| |
Collapse
|
16
|
Qian H, Ying G, Xu H, Wang S, Wu B, Wang X, Qi H, He M, Ud Din MJ, Huang T, Wu Y, Zhang G. Clinical and genetic analysis of children with glucose transporter type 1 deficiency syndrome. MEDICINE INTERNATIONAL 2024; 4:57. [PMID: 39092009 PMCID: PMC11289861 DOI: 10.3892/mi.2024.181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024]
Abstract
Glucose transporter type 1 deficiency syndrome (GLUT1-DS) is a rare metabolic encephalopathy with a wide variety of clinical phenotypes. In the present study, 15 patients diagnosed with GLUT1-DS were selected, all of whom had obvious clinical manifestations and complete genetic testing. Their clinical data and genetic reports were collated. All patients were provided with a ketogenic diet (KD) and an improvement in their symptoms was observed during a follow-up period of up to 1 year. The results revealed that the 15 cases had clinical symptoms, such as convulsions or dyskinesia. Although none had a cerebrospinal fluid/glucose ratio <0.4, the genetic report revealed that all had the solute carrier family 2 member 1 gene variant, and their clinical symptoms basically improved following the use of the KD. GLUT1-DS is a genetic metabolic disease that causes a series of neurological symptoms due to glucose metabolism disorders in the brain. Low glucose levels in cerebrospinal fluid and genetic testing are key diagnostic criteria, and the KD is a highly effective treatment option. By summarizing and analyzing patients with GLUT1-DS, summarizing clinical characteristics and expanding their gene profile, the findings of the present study may be of clinical significance for the early recognition and diagnosis of the disease, so as to conduct early treatment and shorten the duration of brain energy deficiency. This is of utmost importance for improving the prognosis and quality of life of affected children.
Collapse
Affiliation(s)
- Hao Qian
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Guohuan Ying
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Haifeng Xu
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Shangyu Wang
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Bing Wu
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Xin Wang
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Hongdan Qi
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Mingying He
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - M. Jalal Ud Din
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Tingting Huang
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yimei Wu
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Gang Zhang
- Department of Neurology, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| |
Collapse
|
17
|
Ito Y, Nakatsukasa H, Toyoma Y, Nagata S, Oguni H. Differentiating non-epileptic seizures from epileptic seizures in Glut1 deficiency syndrome. Dev Med Child Neurol 2024; 66:1466-1475. [PMID: 38655597 DOI: 10.1111/dmcn.15942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024]
Abstract
AIM To investigate the clinical characteristics of non-epileptic seizures due to transient brain dysfunction caused by energy deficiency after prolonged fasting or exercise in individuals with glucose transporter type 1 deficiency syndrome (Glut1DS), and then elucidate further the seizure features to distinguish non-epileptic seizures from epileptic seizures. METHOD This retrospective case-control study included 57 non-epileptic seizures and 23 epileptic seizures (control group) in 14 individuals (11 males, three females; aged 5-44 years, median = 20 years) with Glut1DS, all with a heterozygous pathogenic SLC2A1 mutation. RESULTS Non-epileptic seizures were classified as paroxysmal altered consciousness (n = 8), movement disorders (n = 35) (eye-head movements, ataxia, spasticity, weakness, involuntary movement), dysaesthesia (n = 8), and vomiting (n = 6) at the peak ages at onset of 5 to 10 years. Ketogenic diet therapy was effective in 33 of 43 (77%) non-epileptic seizures. Providing supplementary food before high-impact exercise or during attacks prevented or mitigated non-epileptic seizures in some individuals. Glut1DS-associated non-epileptic seizures are fundamentally situation-related seizures with specific provoking and ameliorating factors. Non-epileptic seizures can be distinguished from epileptic seizures by the absence of complete consciousness loss and rapid postictal recovery despite prolonged seizures. INTERPRETATION Non-epileptic seizures are not well recognized but require different therapeutic approaches compared to epileptic seizures. Awareness of the differentiation of non-epileptic seizures from epileptic seizures is essential when performing preventive or therapeutic decision-making for acute exacerbation seizures. WHAT THIS PAPER ADDS Non-epileptic seizures are invariably situation-related seizures. Non-epileptic seizures were classified as altered consciousness, movement disorders, dysaesthesia, and vomiting. Non-epileptic seizures were characterized by the absence of complete consciousness loss and were accompanied by rapid recovery. Non-epileptic seizures can occur simultaneously or consecutively with another. Supplementary food can be effective in preventing the development of sustained exercise-induced movement disorders.
Collapse
Affiliation(s)
- Yasushi Ito
- Department of Pediatrics, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
- Research Department of Pediatric and Maternal Health, Aiiku Research Institute, Maternal & Child Health Center, Imperial Gift Foundation Boshi-Aiiku-Kai, Tokyo, Japan
| | - Hidetsugu Nakatsukasa
- Department of Pediatrics, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Yuriko Toyoma
- Department of Pediatrics, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Satoru Nagata
- Department of Pediatrics, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Hirokazu Oguni
- Department of Pediatrics, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
- Epilepsy Center, TMG Asaka Medical Center, Saitama, Japan
| |
Collapse
|
18
|
Giugno A, Falcone E, Fortunato F, Sammarra I, Procopio R, Gagliardi M, Bauleo A, de Stefano L, Martino I, Gambardella A. Glucose transporter-1 deficiency syndrome with extreme phenotypic variability in a five-generation family carrying a novel SLC2A1 variant. Eur J Neurol 2024; 31:e16325. [PMID: 38803061 PMCID: PMC11235872 DOI: 10.1111/ene.16325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND AND PURPOSE Glucose transporter-1 (GLUT1) deficiency syndrome (GLUT1-DS) is a metabolic disorder due to reduced expression of GLUT1, a glucose transporter of the central nervous system. GLUT1-DS is caused by heterozygous SLC2A1 variants that mostly arise de novo. Here, we report a large family with heterogeneous phenotypes related to a novel SLC2A1 variant. METHODS We present clinical and genetic features of a five-generation family with GLUT1-DS. RESULTS The 14 (nine living) affected members had heterogeneous phenotypes, including seizures (11/14), behavioral disturbances (5/14), mild intellectual disability (3/14), and/or gait disabilities (2/14). Brain magnetic resonance imaging revealed hippocampal sclerosis in the 8-year-old proband, who also had drug-responsive absences associated with attention-deficit/hyperactivity disorder. His 52-year-old father, who had focal epilepsy since childhood, developed paraparesis related to a reversible myelitis associated with hypoglycorrhachia. Molecular study detected a novel heterozygous missense variant (c.446C>T) in exon 4 of SLC2A1 (NM: 006516.2) that cosegregated with the illness. This variant causes an amino acid replacement (p.Pro149Leu) at the fourth transmembrane segment of GLUT1, an important domain located at its catalytic core. CONCLUSIONS Our study illustrates the extremely heterogenous phenotypes in familial GLUT1-DS, ranging from milder classic phenotypes to more subtle neurological disorder including paraparesis. This novel SLC2A1 variant (c.446C>T) provides new insight into the pathophysiology of GLUT1-DS.
Collapse
Affiliation(s)
- Alessia Giugno
- Department of Medical and Surgical Sciences, Institute of NeurologyUniversity Magna GræciaCatanzaroItaly
| | - Elena Falcone
- BIOGENET–Medical and Forensic Genetics LaboratoryCosenzaItaly
| | - Francesco Fortunato
- Department of Medical and Surgical Sciences, Institute of NeurologyUniversity Magna GræciaCatanzaroItaly
| | - Ilaria Sammarra
- Department of Medical and Surgical Sciences, Institute of NeurologyUniversity Magna GræciaCatanzaroItaly
| | - Radha Procopio
- Department of Medical and Surgical Sciences, Neuroscience Research CenterMagna Graecia UniversityCatanzaroItaly
| | - Monica Gagliardi
- Department of Medical and Surgical Sciences, Neuroscience Research CenterMagna Graecia UniversityCatanzaroItaly
| | - Alessia Bauleo
- BIOGENET–Medical and Forensic Genetics LaboratoryCosenzaItaly
| | | | - Iolanda Martino
- Department of Medical and Surgical Sciences, Institute of NeurologyUniversity Magna GræciaCatanzaroItaly
| | - Antonio Gambardella
- Department of Medical and Surgical Sciences, Institute of NeurologyUniversity Magna GræciaCatanzaroItaly
- Department of Medical and Surgical Sciences, Neuroscience Research CenterMagna Graecia UniversityCatanzaroItaly
| |
Collapse
|
19
|
De Giorgis V, Bhatia KP, Boespflug-Tanguy O, Gras D, Marina AD, Desurkar A, Toledo M, Miller I, Rotstein M, Schneider SA, Tarquinio DC, Weber Y, Brandabur M, Mayhew J, Koutsoukos T, De Vivo DC. Triheptanoin Did Not Show Benefit versus Placebo for the Treatment of Paroxysmal Movement Disorders in Glut1 Deficiency Syndrome: Results of a Randomized Phase 3 Study. Mov Disord 2024; 39:1386-1396. [PMID: 38725190 DOI: 10.1002/mds.29822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Paroxysmal movement disorders are common in Glut1 deficiency syndrome (Glut1DS). Not all patients respond to or tolerate ketogenic diets. OBJECTIVES The objective was to evaluate the effectiveness and safety of triheptanoin in reducing the frequency of disabling movement disorders in patients with Glut1DS not receiving a ketogenic diet. METHODS UX007G-CL301 was a randomized, double-blind, placebo-controlled, phase 3 crossover study. After a 6-week run-in, eligible patients were randomized 1:1 to the first sequence (triheptanoin/placebo or placebo/triheptanoin) titration plus maintenance, followed by washout and the opposite sequence titration plus maintenance. The placebo (safflower oil) matched the appearance, taste, and smell of triheptanoin. Open-label triheptanoin was administered in the extension. The frequency of disabling paroxysmal movement disorder events per 4 weeks (recorded by diary during maintenance; primary endpoint) was assessed by Wilcoxon rank-sum test. RESULTS Forty-three patients (children, n = 16; adults, n = 27) were randomized and treated. There was no difference between triheptanoin and placebo in the mean (interquartile range) number of disabling paroxysmal movement disorder events (14.3 [4.7-38.3] vs. 11.8; [3.2-28.7]; Hodges-Lehmann estimated median difference: 1.46; 95% confidence interval, -1.12 to 4.36; P = 0.2684). Treatment-emergent adverse events were mild/moderate in severity and included diarrhea, vomiting, upper abdominal pain, headache, and nausea. Two patients discontinued the study because of non-serious adverse events that were predominantly gastrointestinal. The study was closed early during the open-label extension because of lack of effectiveness. Seven patients continued to receive triheptanoin compassionately. CONCLUSION There were no significant differences between the triheptanoin and placebo groups in the frequency of disabling movement disorder events during the double-blind maintenance period. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Kailash P Bhatia
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| | - Odile Boespflug-Tanguy
- Service de Neurologie Pédiatrique, Centre de Référence Leucodystrophies et Leucoencephalopathies de Cause Rare (LEUKOFRANCE), APHP Robert-Debré, Paris, France
| | - Domitille Gras
- Service de Neurologie Pédiatrique, Centre de Référence Leucodystrophies et Leucoencephalopathies de Cause Rare (LEUKOFRANCE), APHP Robert-Debré, Paris, France
| | - Adela Della Marina
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Archana Desurkar
- Neurology Department, Sheffield Children's National Health Service Foundation Trust, Sheffield, United Kingdom
| | - Manuel Toledo
- Epilepsy Unit, Neurology Department, Hospital Vall d'Hebron, Barcelona, Spain
| | - Ian Miller
- Department of Neurology and Comprehensive Epilepsy Program, Brain Institute, Miami Children's Hospital, Miami, Florida, USA
| | - Michael Rotstein
- Pediatric Movement Disorders Service, The Pediatric Neurology Unite and Child Development Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Susanne A Schneider
- Department of Neurology, Ludwig-Maximilians-University of München, Munich, Germany
| | | | - Yvonne Weber
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Section of Epileptology, Department of Neurology, University Hospital RWTH Aachen, Aachen, Germany
| | | | - Jill Mayhew
- Ultragenyx Pharmaceutical Inc., Novato, California, USA
| | | | - Darryl C De Vivo
- Department of Neurology and Pediatrics, Columbia University, New York, New York, USA
| |
Collapse
|
20
|
Rana AK, Bhatt B, Kumar M. β-Hydroxybutyrate Improves the Redox Status, Cytokine Production and Phagocytic Potency of Glucose-Deprived HMC3 Human Microglia-like Cells. J Neuroimmune Pharmacol 2024; 19:35. [PMID: 39042253 DOI: 10.1007/s11481-024-10139-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 07/06/2024] [Indexed: 07/24/2024]
Abstract
Brain glucose deprivation is a component of the pathophysiology of ischemia, glucose transporter1 (GLUT1) deficiency, neurological disorders and occurs transiently in diabetes. Microglia, the neuroimmune cells must function effectively to offer immune defence and debris removal in low-energy settings. Brain glucose deprivation may compromise microglial functions further escalating the disease pathology and deteriorating the overall mental health. In the current study, HMC3 human microglia-like cells were cultured in vitro and exposed to glucose deprivation to investigate the effects of glucose deprivation on phenotypic state, redox status, secretion of cytokines and phagocytic capabilities of HMC3 cells. However, HMC3 cells were able to proliferate in the absence of glucose but showed signs of redox imbalance and mitochondrial dysfunction, as demonstrated by decreased MTT reduction and Mito Tracker™ staining of cells, along with a concomitant reduction in NOX2 protein, superoxide, and nitrite levels. Reduced levels of secreted TNF and IL-1β were the signs of compromised cytokine secretion by glucose-deprived HMC3 microglia-like cells. Moreover, glucose-deprived HMC3 cells also showed reduced phagocytic activity as assessed by fluorescently labelled latex beads-based functional phagocytosis assay. β-hydroxybutyrate (BHB) supplementation restored the redox status, mitochondrial health, cytokine secretion, and phagocytic activity of glucose-deprived HMC3 microglia-like cells. Overall, impaired brain glucose metabolism may hinder microglia's capacity to release diffusible immune factors and perform phagocytosis. This could escalate the mental health issues in neurological diseases where brain glucose metabolism is compromised. Moreover, nutritional ketosis or exogenous ketone supplementation such as BHB may be utilized as a potential metabolic therapies for these conditions.
Collapse
Affiliation(s)
- Anil Kumar Rana
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, 140306, India
| | - Babita Bhatt
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, 140306, India
| | - Mohit Kumar
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, 140306, India.
- Adjunct faculty, Regional Centre for Biotechnology, Faridabad, 121001, India.
| |
Collapse
|
21
|
Groulx-Boivin E, Bouchet T, Myers KA. Understanding of Consciousness in Absence Seizures: A Literature Review. Neuropsychiatr Dis Treat 2024; 20:1345-1353. [PMID: 38947367 PMCID: PMC11212660 DOI: 10.2147/ndt.s391052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/14/2024] [Indexed: 07/02/2024] Open
Abstract
Absence seizures are classically associated with behavioral arrest and transient deficits in consciousness, yet substantial variability exists in the severity of the impairment. Despite several decades of research on the topic, the pathophysiology of absence seizures and the mechanisms underlying behavioral impairment remain unclear. Several rationales have been proposed including widespread cortical deactivation, reduced perception of external stimuli, and transient suspension of the default mode network, among others. This review aims to summarize the current knowledge on the neural correlates of impaired consciousness in absence seizures. We review evidence from studies using animal models of absence epilepsy, electroencephalography, functional magnetic resonance imaging, magnetoencephalography, positron emission tomography, and single photon emission computed tomography.
Collapse
Affiliation(s)
- Emilie Groulx-Boivin
- Department of Neurology and Neurosurgery, Montreal Children’s Hospital, McGill University, Montreal, Quebec, Canada
- Department of Pediatrics, Montreal Children’s Hospital, McGill University, Montreal, Quebec, Canada
| | - Tasha Bouchet
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Kenneth A Myers
- Department of Neurology and Neurosurgery, Montreal Children’s Hospital, McGill University, Montreal, Quebec, Canada
- Department of Pediatrics, Montreal Children’s Hospital, McGill University, Montreal, Quebec, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
22
|
Chen Y, Long J, Wu S, Wei Y, Yan F, Li Q, Yan J, Zhang N, Xu W. Disruption of a DNA G-quadruplex causes a gain-of-function SCL45A1 variant relevant to developmental disorders. Acta Biochim Biophys Sin (Shanghai) 2024; 56:709-716. [PMID: 38655615 PMCID: PMC11177108 DOI: 10.3724/abbs.2024053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/04/2024] [Indexed: 04/26/2024] Open
Abstract
SLC45A1 encodes a glucose transporter protein highly expressed in the brain. Mutations in SLC45A1 may lead to neurological diseases and developmental disorders, but its exact role is poorly understood. DNA G-quadruplexes (DNA G4s) are stable structures formed by four guanine bases and play a role in gene regulation and genomic stability. Changes in DNA G4s may affect brain development and function. The mechanism linking alterations in DNA G-quadruplex structures to SLC45A1 pathogenicity remains unknown. In this study, we identify a functional DNA G-quadruplex and its key binding site on SLC45A1 (NM_001080397.3: exon 2: c.449 G>A: p.R150K). This variant results in the upregulation of mRNA and protein expression, which may lead to intellectual developmental disorder with neuropsychiatric features. Mechanistically, the mutation is found to disrupt DNA G-quadruplex structures on SLC45A1, leading to transcriptional enhancement and a gain-of-function mutation, which further causes increased expression and function of the SLC45A1 protein. The identification of the functional DNA G-quadruplex and its effects on DNA G4s may provide new insights into the genetic basis of SLC45A1 pathogenicity and highlight the importance of DNA G4s of SLC45A1 in regulating gene expression and brain development.
Collapse
Affiliation(s)
- Yuxi Chen
- Joint Laboratory of Reproductive MedicineGynaecology and Paediatric Diseases and Birth Defects of Ministry of EducationWest China Second University HospitalSichuan UniversityChengdu610041China
- West China School of PharmacySichuan UniversityChengdu610041China
| | - Jiang Long
- The Mental Health Centre and the Psychiatric LaboratoryWest China HospitalSichuan UniversityChengdu610041China
| | - Sixian Wu
- Joint Laboratory of Reproductive MedicineGynaecology and Paediatric Diseases and Birth Defects of Ministry of EducationWest China Second University HospitalSichuan UniversityChengdu610041China
| | - Yazhen Wei
- West China School of PharmacySichuan UniversityChengdu610041China
| | - Fei Yan
- Joint Laboratory of Reproductive MedicineGynaecology and Paediatric Diseases and Birth Defects of Ministry of EducationWest China Second University HospitalSichuan UniversityChengdu610041China
| | - Qing Li
- Joint Laboratory of Reproductive MedicineGynaecology and Paediatric Diseases and Birth Defects of Ministry of EducationWest China Second University HospitalSichuan UniversityChengdu610041China
| | - Jierui Yan
- Joint Laboratory of Reproductive MedicineGynaecology and Paediatric Diseases and Birth Defects of Ministry of EducationWest China Second University HospitalSichuan UniversityChengdu610041China
| | - Nannan Zhang
- National Centre for Birth Defect MonitoringKey Laboratory of Birth Defects and Related Diseases of Women and ChildrenMinistry of EducationWest China Second University HospitalSichuan UniversityChengdu610041China
| | - Wenming Xu
- Joint Laboratory of Reproductive MedicineGynaecology and Paediatric Diseases and Birth Defects of Ministry of EducationWest China Second University HospitalSichuan UniversityChengdu610041China
| |
Collapse
|
23
|
Hayden AN, Brandel KL, Merlau PR, Vijayakumar P, Leptich EJ, Pietryk EW, Gaytan ES, Ni CW, Chao HT, Rosenfeld JA, Arey RN. Behavioral screening of conserved RNA-binding proteins reveals CEY-1/YBX RNA-binding protein dysfunction leads to impairments in memory and cognition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574402. [PMID: 38260399 PMCID: PMC10802296 DOI: 10.1101/2024.01.05.574402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
RNA-binding proteins (RBPs) regulate translation and plasticity which are required for memory. RBP dysfunction has been linked to a range of neurological disorders where cognitive impairments are a key symptom. However, of the 2,000 RBPs in the human genome, many are uncharacterized with regards to neurological phenotypes. To address this, we used the model organism C. elegans to assess the role of 20 conserved RBPs in memory. We identified eight previously uncharacterized memory regulators, three of which are in the C. elegans Y-Box (CEY) RBP family. Of these, we determined that cey-1 is the closest ortholog to the mammalian Y-Box (YBX) RBPs. We found that CEY-1 is both necessary in the nervous system for memory ability and sufficient to increase memory. Leveraging human datasets, we found both copy number variation losses and single nucleotide variants in YBX1 and YBX3 in individuals with neurological symptoms. We identified one predicted deleterious YBX3 variant of unknown significance, p.Asn127Tyr, in two individuals with neurological symptoms. Introducing this variant into endogenous cey-1 locus caused memory deficits in the worm. We further generated two humanized worm lines expressing human YBX3 or YBX1 at the cey-1 locus to test evolutionary conservation of YBXs in memory and the potential functional significance of the p.Asn127Tyr variant. Both YBX1/3 can functionally replace cey-1, and introduction of p.Asn127Tyr into the humanized YBX3 locus caused memory deficits. Our study highlights the worm as a model to reveal memory regulators and identifies YBX dysfunction as a potential new source of rare neurological disease.
Collapse
Affiliation(s)
- Ashley N Hayden
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Katie L Brandel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Paul R Merlau
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | | | - Emily J Leptich
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
| | - Edward W Pietryk
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
| | - Elizabeth S Gaytan
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Postbaccalaureate Research Education Program, Baylor College of Medicine, Houston, TX, 77030
| | - Connie W Ni
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Neuroscience, Rice University, Houston, TX 77005
| | - Hsiao-Tuan Chao
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Department of Pediatrics, Division of Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX, 77030
- Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, 77030
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, TX, 77030
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030
- Baylor Genetics Laboratories, Houston, TX 77021
| | - Rachel N Arey
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, 77030
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|
24
|
Peng Q, Zeng W. The protective role of endothelial GLUT1 in ischemic stroke. Brain Behav 2024; 14:e3536. [PMID: 38747733 PMCID: PMC11095318 DOI: 10.1002/brb3.3536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
OBJECTIVE To provide thorough insight on the protective role of endothelial glucose transporter 1 (GLUT1) in ischemic stroke. METHODS We comprehensively review the role of endothelial GLUT1 in ischemic stroke by narrating the findings concerning biological characteristics of GLUT1 in brain in depth, summarizing the changes of endothelial GLUT1 expression and activity during ischemic stroke, discussing how GLUT1 achieves its neuroprotective effect via maintaining endothelial function, and identifying some outstanding blind spots in current studies. RESULTS Endothelial GLUT1 maintains persistent high glucose and energy requirements of the brain by transporting glucose through the blood-brain barrier, which preserves endothelial function and is beneficial to stroke prognosis. CONCLUSION This review underscores the potential involvement of GLUT1 trafficking, activity modulation, and degradation, and we look forward to more clinical and animal studies to illuminate these mechanisms.
Collapse
Affiliation(s)
- Qiwei Peng
- Department of Critical Care Medicine, Union HospitalTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology)Ministry of EducationWuhanChina
| | - Weiqi Zeng
- Department of NeurologyThe First People's Hospital of FoshanFoshanChina
| |
Collapse
|
25
|
Pedrón Giner CC. [Fourteenth Jesús Culebras Lecture. Ketogenic diet, a half-discovered treatment]. NUTR HOSP 2024; 41:477-488. [PMID: 38450481 DOI: 10.20960/nh.05171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024] Open
Abstract
Introduction The ketogenic diet was an amazing approach to treating epilepsy from its beginning. The body undergoes a change in obtaining energy, going from depending on carbohydrates to depending on fats, and then a whole series of biochemical routes are launched that, independently but also complementary, give rise to a set of effects that benefit the patient. This search for its mechanism of action, of devising how to improve compliance and take advantage of it for other diseases has marked its trajectory. This article briefly reviews these aspects, emphasizing the importance of continuing to carry out basic and clinical research so that this treatment can be applied with solid scientific bases.
Collapse
Affiliation(s)
- Consuelo Carmen Pedrón Giner
- Sección de Gastroenterología y Nutrición. Servicio de Pediatría. Hospital Infantil Universitario Niño Jesús. Departamento de Pediatría. Universidad Autónoma de Madrid
| |
Collapse
|
26
|
Wortmann SB, Feichtinger RG, Abela L, van Gemert LA, Aubart M, Dufeu-Berat CM, Boddaert N, de Coo R, Stühn L, Hebbink J, Heinritz W, Hildebrandt J, Himmelreich N, Korenke C, Lehman A, Leyland T, Makowski C, Martinez Marin RJ, Marzin P, Mühlhausen C, Rio M, Rotig A, Roux CJ, Schiff M, Haack TB, Syrbe S, Zylicz SA, Thiel C, Veiga da Cunha M, van Schaftingen E, Wagner M, Mayr JA, Wevers RA, Boltshauser E, Willemsen MA. Clinical, Neuroimaging, and Metabolic Footprint of the Neurodevelopmental Disorder Caused by Monoallelic HK1 Variants. Neurol Genet 2024; 10:e200146. [PMID: 38617198 PMCID: PMC11010246 DOI: 10.1212/nxg.0000000000200146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/13/2024] [Indexed: 04/16/2024]
Abstract
Background and Objectives Hexokinase 1 (encoded by HK1) catalyzes the first step of glycolysis, the adenosine triphosphate-dependent phosphorylation of glucose to glucose-6-phosphate. Monoallelic HK1 variants causing a neurodevelopmental disorder (NDD) have been reported in 12 individuals. Methods We investigated clinical phenotypes, brain MRIs, and the CSF of 15 previously unpublished individuals with monoallelic HK1 variants and an NDD phenotype. Results All individuals had recurrent variants likely causing gain-of-function, representing mutational hot spots. Eight individuals (c.1370C>T) had a developmental and epileptic encephalopathy with infantile onset and virtually no development. Of the other 7 individuals (n = 6: c.1334C>T; n = 1: c.1240G>A), 3 adults showed a biphasic course of disease with a mild static encephalopathy since early childhood and an unanticipated progressive deterioration with, e.g., movement disorder, psychiatric disease, and stroke-like episodes, epilepsy, starting in adulthood. Individuals who clinically presented in the first months of life had (near)-normal initial neuroimaging and severe cerebral atrophy during follow-up. In older children and adults, we noted progressive involvement of basal ganglia including Leigh-like MRI patterns and cerebellar atrophy, with remarkable intraindividual variability. The CSF glucose and the CSF/blood glucose ratio were below the 5th percentile of normal in almost all CSF samples, while blood glucose was unremarkable. This biomarker profile resembles glucose transporter type 1 deficiency syndrome; however, in HK1-related NDD, CSF lactate was significantly increased in all patients resulting in a substantially different biomarker profile. Discussion Genotype-phenotype correlations appear to exist for HK1 variants and can aid in counseling. A CSF biomarker profile with low glucose, low CSF/blood glucose, and high CSF lactate may point toward monoallelic HK1 variants causing an NDD. This can help in variant interpretation and may aid in understanding the pathomechanism. We hypothesize that progressive intoxication and/or ongoing energy deficiency lead to the clinical phenotypes and progressive neuroimaging findings.
Collapse
Affiliation(s)
- Saskia B Wortmann
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rene G Feichtinger
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lucia Abela
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Loes A van Gemert
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mélodie Aubart
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Claire-Marine Dufeu-Berat
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nathalie Boddaert
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rene de Coo
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lara Stühn
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jasmijn Hebbink
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Wolfram Heinritz
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Julia Hildebrandt
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nastassja Himmelreich
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christoph Korenke
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anna Lehman
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas Leyland
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christine Makowski
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rafael Jenaro Martinez Marin
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pauline Marzin
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chris Mühlhausen
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marlène Rio
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Agnes Rotig
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Charles-Joris Roux
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Manuel Schiff
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tobias B Haack
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Steffen Syrbe
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stas A Zylicz
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christian Thiel
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maria Veiga da Cunha
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Emile van Schaftingen
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Matias Wagner
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Johannes A Mayr
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ron A Wevers
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Eugen Boltshauser
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michel A Willemsen
- From the University Children's Hospital Salzburg (S.B.W., R.G.F., J.A.M.), Austria; Amalia Children's Hospital (S.B.W., L.A.G., J. Hebbink, M.A.W.), Department of Pediatrics (Pediatric Neurology), Nijmegen, The Netherlands; Division of Child Neurology (L.A., E.B.), University Children's Hospital Zurich, Switzerland; Pediatric Neurology Department (M.A.), Necker-Enfants Malades University Hospital, Paris Cité University, APHP; Reference Centre for Mitochondrial Disorders (CARAMMEL) (C.-M.D.-B., M.S.), Hôpital Necker-Enfants-Malades, APHP, Université Paris Cité, Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; 6Paediatric Radiology Department (N.B.), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163France; Department of Toxicogenomics (R.C.), Research School of Mental Health and Neuroscience, Maastricht University, The Netherlands; Institute of Medical Genetics and Applied Genomics (L.S., T.B.H.), University of Tübingen; Praxis für Humangenetik (W.H.); Carl-Thiem-Klinikum Cottbus (W.H.); Center for Human Genetics Tübingen (J. Hildebrandt, N.H.); CeGaT GmbH (J. Hildebrandt, N.H.), Tübingen; Department Pediatrics (N.H., C.T.), Centre for Child and Adolescent Medicine, University of Heidelberg; Department of Neuropediatrics (C.K.), University Children's Hospital, Klinikum Oldenburg, Germany; University of British Columbia (A.L.), Vancouver, Canada; Royal Belfast Hospital for Sick Children (T.L.), Belfast, Northern Ireland; University Hospital (C. Makowski), LMU Munich, Division of Pediatric Neurology, Developmental Medicine and Social Pediatrics, Department of Pediatrics, Dr. von Hauner Children's Hospital, Munich, Germany; Department of Neurology (R.J.M.M.), Hospital Universitario La Paz, Madrid, Spain; Reference Center for Intellectual Disabilities of Rare causes (P.M., M.R.), Federation de médecine Génomique des maladies Rares, APHP, Hôpital Necker-Enfants Malades, Paris, France; University Medical Centre Göttingen (C. Mühlhausen), Department of Pediatrics and Adolescent Medicine, Göttingen, Germany; Université Paris Cité (A.R.), Imagine Institute, Genetics of Mitochondrial Disorders, INSERM UMR 1163; Paediatric Radiology Department (C.-J.R), AP-HP, Hôpital Necker Enfants Malades, Université Paris Cité, Institut Imagine INSERM U1163, Paris France; Division of Pediatric Epileptology (S.S.), Centre for Child and Adolescent Medicine, University of Heidelberg, Germany; Department of Neurology (S.A.Z.), LangeLand Hospital, Zoetermeer, The Netherlands; Metabolic Research Group (M.V.C., E.S.), de Duve Institute and UCLouvain, Brussels, Belgium; Technical University of Munich (M. Wagner), School of Medicine, Institute of Human Genetics, Munich, Germany; and Department of Human Genetics (R.A.W.), Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
27
|
Falsaperla R, Sortino V, Vitaliti G, Privitera GF, Ruggieri M, Fusto G, Pappalardo XG. GLUT-1DS resistant to ketogenic diet: from clinical feature to in silico analysis. An exemplificative case report with a literature review. Neurogenetics 2024; 25:69-78. [PMID: 38190079 DOI: 10.1007/s10048-023-00742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/31/2023] [Indexed: 01/09/2024]
Abstract
Glucose transporter type 1 deficiency syndrome (GLUT-1DS) is characterized by alterations in glucose translocation through the blood-brain barrier (BBB) due to mutation involving the GLUT-1 transporter. The fundamental therapy is ketogenic diet (KD) that provide an alternative energetic substrate - ketone bodies that across the BBB via MCT-1 - for the brain. Symptoms are various and include intractable seizure, acquired microcephalia, abnormal ocular movement, movement disorder, and neurodevelopment delay secondary to an energetic crisis for persistent neuroglycopenia. KD is extremely effective in controlling epileptic seizures and has a positive impact on movement disorders and cognitive impairment. Cases of KD resistance are rare, and only a few of them are reported in the literature, all regarding seizure. Our study describes a peculiar case of GLUT-1DS due to a new deletion involving the first codon of SLC2A1 gene determining a loss of function with a resistance to KD admitted to hospital due to intractable episodes of dystonia. This patient presented a worsening of symptomatology at higher ketonemia values but without hyperketosis and showed a complete resolution of symptomatology while maintaining low ketonemia values. Our study proposes an in-silico genomic and proteomic analysis aimed at explaining the atypical response to KD exhibited by our patient. In this way, we propose a new clinical and research approach based on precision medicine and molecular modelling to be applied to patients with GLUT-1DS resistant to first-line treatment with ketogenic diet by in silico study of genetic and altered protein product.
Collapse
Affiliation(s)
- Raffaele Falsaperla
- Neonatal Intensive Care Unit and Neonatal Accompaniment Unit, Azienda Ospedaliero-Universitaria Policlinico "Rodolico-San Marco," San Marco Hospital, University of Catania, Catania, Italy.
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico, "Rodolico-San Marco," San Marco Hospital, Catania, Italy.
| | - Vincenzo Sortino
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico, "Rodolico-San Marco," San Marco Hospital, Catania, Italy
| | - Giovanna Vitaliti
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico, "Rodolico-San Marco," San Marco Hospital, Catania, Italy
| | | | - Martino Ruggieri
- Unit of Clinical Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, AOU "Policlinico," PO "G. Rodolico", Via S. Sofia, 78, 95124, Catania, Italy
| | - Gaia Fusto
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy
| | - Xena Giada Pappalardo
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy
- National Council of Research, Institute for Research and Biomedical Innovation (IRIB), Unit of Catania, Catania, Italy
| |
Collapse
|
28
|
Zhang C, Gu L, Xie H, Liu Y, Huang P, Zhang J, Luo D, Zhang J. Glucose transport, transporters and metabolism in diabetic retinopathy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166995. [PMID: 38142757 DOI: 10.1016/j.bbadis.2023.166995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Diabetic retinopathy (DR) is the most common reason for blindness in working-age individuals globally. Prolonged high blood glucose is a main causative factor for DR development, and glucose transport is prerequisite for the disturbances in DR caused by hyperglycemia. Glucose transport is mediated by its transporters, including the facilitated transporters (glucose transporter, GLUTs), the "active" glucose transporters (sodium-dependent glucose transporters, SGLTs), and the SLC50 family of uniporters (sugars will eventually be exported transporters, SWEETs). Glucose transport across the blood-retinal barrier (BRB) is crucial for nourishing the neuronal retina in the context of retinal physiology. This physiological process primarily relies on GLUTs and SGLTs, which mediate the glucose transportation across both the cell membrane of retinal capillary endothelial cells and the retinal pigment epithelium (RPE). Under diabetic conditions, increased accumulation of extracellular glucose enhances the retinal cellular glucose uptake and metabolism via both glycolysis and glycolytic side branches, which activates several biochemical pathways, including the protein kinase C (PKC), advanced glycation end-products (AGEs), polyol pathway and hexosamine biosynthetic pathway (HBP). These activated biochemical pathways further increase the production of reactive oxygen species (ROS), leading to oxidative stress and activation of Poly (ADP-ribose) polymerase (PARP). The activated PARP further affects all the cellular components in the retina, and finally resulting in microangiopathy, neurodegeneration and low-to-moderate grade inflammation in DR. This review aims to discuss the changes of glucose transport, glucose transporters, as well as its metabolism in DR, which influences the retinal neurovascular unit (NVU) and implies the possible therapeutic strategies for treating DR.
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Limin Gu
- Department of Ophthalmology, Shanghai Aier Eye Hospital, Shanghai, China.
| | - Hai Xie
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Yan Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Jingting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Dawei Luo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| |
Collapse
|
29
|
Falsaperla R, Sortino V, Striano P, Kluger G, Ramantani G, Ruggieri M. Is ketogenic diet a 'precision medicine'? Recent developments and future challenges. Eur J Paediatr Neurol 2024; 48:13-16. [PMID: 37984007 DOI: 10.1016/j.ejpn.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/22/2023]
Abstract
Recently, precision medicine has attracted much attention in the management of epilepsies, but it remains unclear if the increasingly utilized ketogenic diet approaches can truly be considered precision medicine in all epilepsy treatment. Currently, it is the standard treatment for patients with GLUT1 deficiency and the latest NICE guidelines highlight ketogenic diet as a therapeutic option for multi-drug resistant epilepsy patients. Ketogenic diet is presumed to be a precision medicine tool when applied to the treatment of seizures secondary to GLUT1 transporter deficiency. In contrast, the genetic and epigenetic mechanisms modulated by ketogenic diet and underlying its efficacy in other epilepsy types can only be hypothesized to relate to mechanisms of neuroprotection, neuromodulation, and reduction of neuroinflammation. Early ketogenic diet initiation in well-selected patients, would allow immediate action in the direction of neuroprotection and modulation of neuroinflammation, ensuring higher success rates and lower "cost" to the patient in terms of quality of life and comorbidities. These considerations have fueled an increasing interest in investigating the efficacy, side effects, and adherence to long-term use of the ketogenic diet in epilepsy treatment in large contemporary cohorts, available within the scope of multicentric collaborations, such as the European Network for Therapy in Rare Epilepsies (NETRE). Future directions should involve the use of precision medicine, applied to each patient with the help of "omics", whose use should be expanded and inclusive.
Collapse
Affiliation(s)
- Raffaele Falsaperla
- Neonatal Intensive Care Unit and Neonatal Accompaniment Unit, Azienda Ospedaliero-Universitaria Policlinico "Rodolico-San Marco", San Marco Hospital, University of Catania, Catania, Italy; Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico, "Rodolico-San Marco", San Marco Hospital, Catania, Italy.
| | - Vincenzo Sortino
- Unit of Pediatrics and Pediatric Emergency, Azienda Ospedaliero-Universitaria Policlinico, "Rodolico-San Marco", San Marco Hospital, Catania, Italy; Postgraduate Training Program in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Pasquale Striano
- IRCCS 'G. Gaslini Institute', Genoa, Italy; Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.
| | - Gerhard Kluger
- Research Institute for Rehabilitation, Transition, and Palliation, PMU Salzburg, Salzburg, Austria; Epilepsy Center for Children and Adolescents, Schön Clinic Vogtareuth, Vogtareuth, Germany
| | - Georgia Ramantani
- Department of Neuropediatrics, University Children's Hospital Zurich, and University of Zurich, Zurich, Switzerland.
| | - Martino Ruggieri
- Unit of Clinical Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, AOU "Policlinico", PO "G. Rodolico", via S. Sofia, 78, 95124, Catania, Italy
| |
Collapse
|
30
|
van Gemert LA, van Alfen N, van Gaal L, Wortmann S, Willemsen MA. Effects of Sodium Lactate Infusion in Two Girls with Glucose Transporter 1 Deficiency Syndrome. Neuropediatrics 2023; 54:365-370. [PMID: 37478891 PMCID: PMC10643022 DOI: 10.1055/a-2134-8766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/30/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Glucose is an important fuel for the brain. In glucose transporter 1 deficiency syndrome (GLUT1DS), the transport of glucose across the blood-brain barrier is limited. Most individuals with GLUT1DS present with developmental problems, epilepsy, and (paroxysmal) movement disorders, and respond favorably to the ketogenic diet. Similar to ketones, lactate is an alternative energy source for the brain. The aim of this study is to investigate whether intravenous infusion of sodium lactate in children with GLUT1DS has beneficial effects on their epilepsy. METHODS We performed a proof of principle study with two subjects with GLUT1DS who were not on a ketogenic diet and suffered from absence epilepsy. After overnight fasting, sodium lactate (600 mmol/L) was infused during 120 minutes, under video electroencephalographic (EEG) recording and monitoring of serum lactate, glucose, electrolytes, and pH. Furthermore, the EEGs were compared with pre-/postprandial EEGs of both subjects, obtained shortly before the study. RESULTS Fasting EEGs of both subjects showed frequent bilateral, frontocentral polyspike and wave complexes. In one subject, no more epileptic discharges were seen postprandially and after the start of lactate infusion. The EEG of the other subject did not change, neither postprandially nor after lactate infusion. Serum pH, lactate, and sodium changed temporarily during the study. CONCLUSION This study suggests that sodium lactate infusion is possible in individuals with GLUT1DS, and may have potential therapeutic effects. Cellular abnormalities, beyond neuronal energy failure, may contribute to the underlying disease mechanisms of GLUT1DS, explaining why not all individuals respond to the supplementation of alternative energy sources.
Collapse
Affiliation(s)
- Loes A. van Gemert
- Department of Pediatric Neurology, Amalia Children's Hospital, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nens van Alfen
- Department of Neurology and Clinical Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lizzy van Gaal
- Department of Neurology and Clinical Neurophysiology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Saskia Wortmann
- University Childrens Hospital, Paracelsus Medical University Salzburg, Salzburg, Austria
- Department of Metabolic Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michèl A. Willemsen
- Department of Pediatric Neurology, Amalia Children's Hospital, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
31
|
Al-Bishri WM. Glucose transporter 1 deficiency, AMP-activated protein kinase activation and immune dysregulation in autism spectrum disorder: Novel biomarker sources for clinical diagnosis. Saudi J Biol Sci 2023; 30:103849. [PMID: 38020228 PMCID: PMC10654234 DOI: 10.1016/j.sjbs.2023.103849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
The neurophysiological basis of autism spectrum disorder (ASD) is still uncertain. Nevertheless, studies support the hypotheses that oxidative stress, neuroinflammation, immune dysregulation, and metabolic stress are contributors. In this study, the serum levels of 3-nitrotyrosine (3-NT), hypoxia-inducible factor 1 α (HIF-1 α), heat shock protein 70 (HSP-70), interleukin-17A (IL-17A), IL-35, vitamin D3 (VITD), glucose transporter-1 (GUT1), and AMP-activated protein kinase (AMPK) were estimated in Saudi ASD children versus age-matched neurotypical controls, aiming to investigate whether these parameters have potential roles in the pathophysiologic mechanisms of ASD and hoping to find a reliable marker for early ASD diagnosis. This study included 25 ASD children and 25 typically developing children (3-11 years old). The diagnosis of ASD cases was made based on the Autism Diagnostic Observation Schedule (ADOS) and the Statistical Manual of Mental Disorders (DSM-5). ASD subjects were commonly male and revealed an intelligence quotient (IQ) < 70.The results detected that ASD children have remarkable greater serum levels of nitrosative stress (3-NT), hypoxia (HIF-1 α), inflammatory (HSP-70, IL-17A, and AMPK) biomarkers and lower serum levels of anti-inflammatory (IL-35 and VITD) and metabolic stress (GUT-1) biomarkers versus age-matched controls (P ≤ 0.0001). Pearson's correlation study revealed that 3-NT was positively associated with HIF-1 α and HSP-70. HIF-1 α was also positively correlated with HSP-70. AMPK was positively associated with GUT-1, however, IL-17A was negatively correlated with IL-35 and VITD.Limitation:No specific therapeuticdrugs were administered in this study, and further studies are required to confirm the role of the selected biomarkers in ASD managements. Conclusion Changes in concentrations of different biomarkers indicate that they are involved in oxidative stress, metabolic stress, immune dysregulation and ASD pathogenesis. Hence, these parameters can prove to be promising biomarkers as well as therapeutic targets for the timely diagnosis and treatment of ASD patients.
Collapse
Affiliation(s)
- Widad M. Al-Bishri
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 80327, Saudi Arabia
| |
Collapse
|
32
|
Wakid M, Almeida D, Aouabed Z, Rahimian R, Davoli MA, Yerko V, Leonova-Erko E, Richard V, Zahedi R, Borchers C, Turecki G, Mechawar N. Universal method for the isolation of microvessels from frozen brain tissue: A proof-of-concept multiomic investigation of the neurovasculature. Brain Behav Immun Health 2023; 34:100684. [PMID: 37822873 PMCID: PMC10562768 DOI: 10.1016/j.bbih.2023.100684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/29/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
The neurovascular unit, comprised of vascular cell types that collectively regulate cerebral blood flow to meet the needs of coupled neurons, is paramount for the proper function of the central nervous system. The neurovascular unit gatekeeps blood-brain barrier properties, which experiences impairment in several central nervous system diseases associated with neuroinflammation and contributes to pathogenesis. To better understand function and dysfunction at the neurovascular unit and how it may confer inflammatory processes within the brain, isolation and characterization of the neurovascular unit is needed. Here, we describe a singular, standardized protocol to enrich and isolate microvessels from archived snap-frozen human and frozen mouse cerebral cortex using mechanical homogenization and centrifugation-separation that preserves the structural integrity and multicellular composition of microvessel fragments. For the first time, microvessels are isolated from postmortem ventromedial prefrontal cortex tissue and are comprehensively investigated as a structural unit using both RNA sequencing and Liquid Chromatography with tandem mass spectrometry (LC-MS/MS). Both the transcriptome and proteome are obtained and compared, demonstrating that the isolated brain microvessel is a robust model for the NVU and can be used to generate highly informative datasets in both physiological and disease contexts.
Collapse
Affiliation(s)
- Marina Wakid
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Daniel Almeida
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Zahia Aouabed
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | | | - Volodymyr Yerko
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | - Elena Leonova-Erko
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
| | - Vincent Richard
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, Quebec, Canada
| | - René Zahedi
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, Quebec, Canada
| | - Christoph Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montréal, Quebec, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Research Centre, Montréal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
- Department of Psychiatry, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
33
|
Tramutola A, Bakels HS, Perrone F, Di Nottia M, Mazza T, Abruzzese MP, Zoccola M, Pagnotta S, Carrozzo R, de Bot ST, Perluigi M, van Roon-Mom WMC, Squitieri F. GLUT-1 changes in paediatric Huntington disease brain cortex and fibroblasts: an observational case-control study. EBioMedicine 2023; 97:104849. [PMID: 37898095 PMCID: PMC10630613 DOI: 10.1016/j.ebiom.2023.104849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Paediatric Huntington disease with highly expanded mutations (HE-PHD; >80 CAG repeats) presents atypically, compared to adult-onset Huntington disease (AOHD), with neurodevelopmental delay, epilepsy, abnormal brain glucose metabolism, early striatal damage, and reduced lifespan. Since genetic GLUT-1 deficiency syndrome shows a symptom spectrum similar to HE-PHD, we investigated the potential role of the two main glucose transporters, GLUT-1 and GLUT-3, in HE-PHD. METHODS We compared GLUT-1 and GLUT-3 protein expression in HE-PHD, juvenile-onset (JOHD), and AOHD brains (n = 2; n = 3; n = 6) and periphery (n = 3; n = 2; n = 2) versus healthy adult controls (n = 6; n = 6). We also investigated mitochondrial complexes and hexokinase-II protein expression. FINDINGS GLUT-1 and GLUT-3 expression were significantly lower in HE-PHD frontal cortex (p = 0.009, 95% [CI 13.4, 14.7]; p = 0.017, 95% [CI 14.2, 14.5]) versus controls. In fibroblasts, GLUT-1 and GLUT-3 expression were lower compared to controls (p < 0.0001, 95% [CI 0.91, 1.09]; p = 0.046, 95% [CI 0.93, 1.07]). In the frontal cortex, this occurred without evidence of extensive neuronal degeneration. Patients with HE-PHD had deregulated mitochondrial complex expression, particularly complexes II-III, levels of which were lower in frontal cortex versus controls (p = 0.027, 95% [CI 17.1, 17.6]; p = 0.002, 95% CI [16.6, 16.9]) and patients with AOHD (p = 0.052, 95% [CI 17.0, 17.6]; p = 0.002, 95% [CI 16.6, 16.7]). Hexokinase-II expression was also lower in HE-PHD frontal cortex and striatum versus controls (p = 0.010, 95% [CI 17.8, 18.2]; p = 0.045, 95% [CI 18.6, 18.7]) and in frontal cortex versus patients with AOHD (p = 0.013, 95% [CI 17.7, 18.1]). Expression JOHD levels were consistently different to those of HE-PHD but similar to those of AOHD. INTERPRETATION Our data suggest a dysfunctional hypometabolic state occurring specifically in paediatric Huntington disease brains. FUNDING '5 × 1000' Personal Income Tax donation to LIRH Foundation; Italian Ministry of HealthRC2301MH04 and RF-2016-02364123 to CSS.
Collapse
Affiliation(s)
- Antonella Tramutola
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Hannah S Bakels
- Department of Neurology, Leiden University Medical Centre, ZA Leiden 2311, the Netherlands
| | - Federica Perrone
- Huntington and Rare Diseases Unit, IRCCS Casa Sollievo della Sofferenza (CSS) Research Hospital, San Giovanni Rotondo 71013, Italy
| | - Michela Di Nottia
- Unit of Cellular Biology and Mitochondrial Diseases, IRCCS Bambino Gesú Children's Hospital, Rome 00146, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, IRCCS Casa Sollievo della Sofferenza (CSS) Research Hospital, San Giovanni Rotondo 71013, Italy
| | - Maria Pia Abruzzese
- Huntington and Rare Diseases Unit, IRCCS Casa Sollievo della Sofferenza (CSS) Research Hospital, San Giovanni Rotondo 71013, Italy
| | - Martina Zoccola
- Unit of Cellular Biology and Mitochondrial Diseases, IRCCS Bambino Gesú Children's Hospital, Rome 00146, Italy
| | - Sara Pagnotta
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | - Rosalba Carrozzo
- Unit of Cellular Biology and Mitochondrial Diseases, IRCCS Bambino Gesú Children's Hospital, Rome 00146, Italy
| | - Susanne T de Bot
- Department of Neurology, Leiden University Medical Centre, ZA Leiden 2311, the Netherlands
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Rome 00185, Italy
| | | | - Ferdinando Squitieri
- Huntington and Rare Diseases Unit, IRCCS Casa Sollievo della Sofferenza (CSS) Research Hospital, San Giovanni Rotondo 71013, Italy; Centre for Rare Neurological Diseases (CMRN), Italian League for Research on Huntington (LIRH) Foundation, Viale di Villa Massimo 4, Rome 00161, Italy.
| |
Collapse
|
34
|
Mylvaganam S, Freeman SA. The resolution of phagosomes. Immunol Rev 2023; 319:45-64. [PMID: 37551912 DOI: 10.1111/imr.13260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 07/18/2023] [Indexed: 08/09/2023]
Abstract
Phagocytosis is a fundamental immunobiological process responsible for the removal of harmful particulates. While the number of phagocytic events achieved by a single phagocyte can be remarkable, exceeding hundreds per day, the same phagocytic cells are relatively long-lived. It should therefore be obvious that phagocytic meals must be resolved in order to maintain the responsiveness of the phagocyte and to avoid storage defects. In this article, we discuss the mechanisms involved in the resolution process, including solute transport pathways and membrane traffic. We describe how products liberated in phagolysosomes support phagocyte metabolism and the immune response. We also speculate on mechanisms involved in the redistribution of phagosomal metabolites back to circulation. Finally, we highlight the pathologies owed to impaired phagosome resolution, which range from storage disorders to neurodegenerative diseases.
Collapse
Affiliation(s)
- Sivakami Mylvaganam
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Spencer A Freeman
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Rastin C, Schenkel LC, Sadikovic B. Complexity in Genetic Epilepsies: A Comprehensive Review. Int J Mol Sci 2023; 24:14606. [PMID: 37834053 PMCID: PMC10572646 DOI: 10.3390/ijms241914606] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Epilepsy is a highly prevalent neurological disorder, affecting between 5-8 per 1000 individuals and is associated with a lifetime risk of up to 3%. In addition to high incidence, epilepsy is a highly heterogeneous disorder, with variation including, but not limited to the following: severity, age of onset, type of seizure, developmental delay, drug responsiveness, and other comorbidities. Variable phenotypes are reflected in a range of etiologies including genetic, infectious, metabolic, immune, acquired/structural (resulting from, for example, a severe head injury or stroke), or idiopathic. This review will focus specifically on epilepsies with a genetic cause, genetic testing, and biomarkers in epilepsy.
Collapse
Affiliation(s)
- Cassandra Rastin
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada
| | - Laila C. Schenkel
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada
| | - Bekim Sadikovic
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, ON N6A 5W9, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada
| |
Collapse
|
36
|
Little JN, Intagliata V, Garris JF. Microcephaly, Dystonia, and Periodic Ataxia in a 10-year-old Boy. Pediatr Rev 2023; 44:408-411. [PMID: 37391632 DOI: 10.1542/pir.2021-005322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/02/2023]
Affiliation(s)
| | | | - Jordan F Garris
- Department of Pediatrics
- Department of Neurology, University of Virginia, Charlottesville, VA
| |
Collapse
|
37
|
Mochel F, Gras D, Luton MP, Nizou M, Giovannini D, Delattre C, Aubart M, Barth M, De Saint-Martin A, Doummar D, Essid N, Garros A, Le Camus CH, Hoebeke C, The Tich SN, Perivier M, Rivera S, Rolland A, Roubertie A, Sarret C, Sevin C, Ville D, Sitbon M, Costa JM, Pons R, Garcia-Cazorla A, Vuillaumier S, Petit V, Boespflug-Tanguy O, De Vivo DC. Prospective Multicenter Validation of a Simple Blood Test for the Diagnosis of Glut1 Deficiency Syndrome. Neurology 2023; 100:e2360-e2373. [PMID: 37076312 PMCID: PMC10256121 DOI: 10.1212/wnl.0000000000207296] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 03/02/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND AND OBJECTIVE GLUT1 deficiency syndrome (Glut1DS) is a treatable neurometabolic disease that causes a wide range of neurologic symptoms in children and adults. However, its diagnosis relies on an invasive test, that is, a lumbar puncture (LP) to measure glycorrhachia, and sometimes complex molecular analyses of the SLC2A1 gene. This procedure limits the number of patients able to receive the standard of care. We wished to validate the diagnostic performance of METAglut1, a simple blood test that quantifies GLUT1 on the erythrocyte surface. METHODS We performed a multicenter validation study in France, involving 33 centers. We studied 2 patient cohorts: a prospective cohort consisting of patients with a clinical suspicion of Glut1DS explored through the reference strategy, that is, LP and analyses of the SLC2A1 gene, and a retrospective cohort that included patients previously diagnosed with Glut1DS. All patients were blind-tested with METAglut1. RESULTS We analyzed 428 patients in the prospective cohort, including 15 patients newly diagnosed with Glut1DS, and 67 patients in the retrospective cohort. METAglut1 was 80% sensitive and >99% specific for the diagnosis of Glut1DS. Concordance analyses showed a substantial agreement between METAglut1 and glycorrhachia. In the prospective cohort, the positive predictive value of METAglut1 was slightly higher than that of glycorrhachia. METAglut1 succeeded to identify patients with Glut1DS with SCL2A1 mosaicism and variants of unknown significance. DISCUSSION METAglut1 is an easily performed, robust, and noninvasive diagnostic test for the diagnosis of Glut1DS, which allows wide screening of children and adults, including those with atypical forms of this treatable condition. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that a positive METAglut1 test accurately distinguishes patients with suspected GLUT1 deficiency syndrome from other neurologic syndromes as compared with invasive and genetic testing.
Collapse
Affiliation(s)
- Fanny Mochel
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York.
| | - Domitille Gras
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Marie-Pierre Luton
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Manon Nizou
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Donatella Giovannini
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Caroline Delattre
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Mélodie Aubart
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Magalie Barth
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Anne De Saint-Martin
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Diane Doummar
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Nouha Essid
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Alexa Garros
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Caroline Hachon Le Camus
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Celia Hoebeke
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Sylvie Nguyen The Tich
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Maximilien Perivier
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Serge Rivera
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Anne Rolland
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Agathe Roubertie
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Catherine Sarret
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Caroline Sevin
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Dorothee Ville
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Marc Sitbon
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Jean-Marc Costa
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Roser Pons
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Angels Garcia-Cazorla
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Sandrine Vuillaumier
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Vincent Petit
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Odile Boespflug-Tanguy
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| | - Darryl C De Vivo
- From the Hôpital La Pitié-Salpêtrière (F.M., M-P.L., C.D.), Assistance-Publique Hôpitaux de Paris, Inserm U1127; Sorbonne University (D.Gras), Inserm U1127, CNRS UMR7225, Paris Brain Institute; U1141 Neurodiderot (D.Gras), équipe 5 InDev, Inserm, CEA, UP UNIACT, Neurospin, Joliot, DRF, CEA-Saclay; Metafora Biosystems (M.N., V.P.); Institut de Génétique Moléculaire de Montpellier (D.Giovannini), Univ. Montpellier, CNRS; Service de Neurologie Pédiatrique (M.A.), Hôpital Necker-Enfants Malades, Assistance-Publique Hôpitaux de Paris; Service de Génétique (M.B.), Centre Hospitalier Universitaire Angers; Service de Neuropédiatrie (A.D.S-M.), Hôpital de Hautepierre, Strasbourg; Sorbonne Université (D.D.), Service de Neuropédiatrie-Pathologie du développement, Hôpital Trousseau AP-HP.SU, FHU I2D2, Paris; Service de Neurologie et Réanimation Pédiatrique (N.E.), APHP, Hôpital Raymond Poincaré, Garches; Service de Neurologie Pédiatrique (A.G.), Hôpital Nord, Grenoble; Service de Neurologie Pédiatrique (C.H.L.C.), et Unité de recherche clinique, module plurithématique sous axe pédiatrique CI 1436, Hôpital des enfants, CHU Toulouse; Service de Neurologie Pédiatrique (C.H.), Hôpital La Timone, Assistance-Publique Hôpitaux de Marseille; Service de Neuropédiatrie (S.N.T.T.), CRMR Epilepsies Rares, Hôpital Roger Salengro, Lille; Service de Neuropédiatrie et handicaps (M.P.), Hôpital pédiatrique Gatien de Clocheville, CHU de Tours; Pole Mère Enfant (S.R.), Centre Hospitalier de la Côte Basque, Bayonne; Service de Neuropédiatrie (A.R.), Hôpital Mère-Enfant, Nantes; Département de Neuropédiatrie, Centre d'investigation Clinique Inserm (A.R.), INM, Univ Montpellier, INSERM U 1298, CHU Montpellier, CIC1411; Service de Pédiatrie (C.Sarret), Hôpital Estaing, CHU Clermont-Ferrand; Service de Neurologie Pédiatrique (C.Sevin), Hôpital Bicêtre, Assistance-Publique Hôpitaux de Paris; Service de Neurologie Pédiatrique (D.V.), Hôpital Femme Mère Enfant, Centre Hospitalier de Lyon; Institut de Génétique Moléculaire de Montpellier (M.S.), Univ. Montpellier, CNRS; Laboratoire Cerba (J-M.C.), Saint-Ouen l'Aumône, France; Department of Pediatrics (R.P.), Aghia Sofia Hospital, University of Athens, Greece; Department of Neurology (A.G-C.), Hospital Sant Joan de Déu, Barcelona, Spain; Service de Biochimie et Génétique (S.V.), Hôpital Bichat Claude Bernard, APHP, Paris; CRMR Leukofrance Service de neuropediatrie (O.B-T.), Hôpital Robert Debré, AP-HP, Paris; UMR1141, Neurodiderot Université de Paris, France; Department of Pediatrics (D.C.D.V.), Columbia University Irving Medical Center, New York
| |
Collapse
|
38
|
De Amicis R, Leone A, Pellizzari M, Foppiani A, Battezzati A, Lessa C, Tagliabue A, Ferraris C, De Giorgis V, Olivotto S, Previtali R, Veggiotti P, Bertoli S. Long-term follow-up of nutritional status in children with GLUT1 Deficiency Syndrome treated with classic ketogenic diet: a 5-year prospective study. Front Nutr 2023; 10:1148960. [PMID: 37293674 PMCID: PMC10244766 DOI: 10.3389/fnut.2023.1148960] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction The classic ketogenic diet (cKD) is an isocaloric, high fat, low-carbohydrate diet that induces the production of ketone bodies. High consumption of dietary fatty acids, particularly long-chain saturated fatty acids, could impair nutritional status and increase cardiovascular risk. The purpose of this study was to evaluate the long-term effects of a 5-year cKD on body composition, resting energy expenditure, and biochemical parameters in children affected by Glucose Transporter 1 Deficiency Syndrome (GLUT1DS). Methods This was a prospective, multicenter, 5-year longitudinal study of children with GLUT1DS treated with a cKD. The primary outcome was to assess the change in nutritional status compared with pre-intervention, considering anthropometric measurements, body composition, resting energy expenditure, and biochemical parameters such as glucose and lipid profiles, liver enzymes, uric acid, creatinine, and ketonemia. Assessments were conducted at pre-intervention and every 12 months of cKD interventions. Results Ketone bodies increased significantly in children and adolescents, and remained stable at 5 years, depending on the diet. No significant differences were reported in anthropometric and body composition standards, as well as in resting energy expenditure and biochemical parameters. Bone mineral density increased significantly over time according to increasing age. Body fat percentage significantly and gradually decreased in line with the increase in body weight and the consequent growth in lean mass. As expected, we observed a negative trend in respiratory quotient, while fasting insulin and insulin resistance were found to decrease significantly after cKD initiation. Conclusion Long-term adherence to cKD showed a good safety profile on anthropometric measurements, body composition, resting energy expenditure, and biochemical parameters, and we found no evidence of potential adverse effects on the nutritional status of children and adolescents.
Collapse
Affiliation(s)
- Ramona De Amicis
- ICANS-DIS, Department of Food Environmental and Nutritional Sciences, University of Milan, Milan, Italy
- Obesity Unit and Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Alessandro Leone
- ICANS-DIS, Department of Food Environmental and Nutritional Sciences, University of Milan, Milan, Italy
| | - Marta Pellizzari
- ICANS-DIS, Department of Food Environmental and Nutritional Sciences, University of Milan, Milan, Italy
- Obesity Unit and Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Andrea Foppiani
- ICANS-DIS, Department of Food Environmental and Nutritional Sciences, University of Milan, Milan, Italy
| | - Alberto Battezzati
- ICANS-DIS, Department of Food Environmental and Nutritional Sciences, University of Milan, Milan, Italy
- Clinical Nutrition Unit, Department of Endocrine and Metabolic Medicine, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Chiara Lessa
- ICANS-DIS, Department of Food Environmental and Nutritional Sciences, University of Milan, Milan, Italy
| | - Anna Tagliabue
- Human Nutrition and Eating Disorder Centre, University of Pavia, Pavia, Italy
- Ketogenic Metabolic Therapy Laboratory, Department of Public Health, Experimental and Forensic Medicine University of Pavia, Pavia, Italy
| | - Cinzia Ferraris
- Human Nutrition and Eating Disorder Centre, University of Pavia, Pavia, Italy
- Ketogenic Metabolic Therapy Laboratory, Department of Public Health, Experimental and Forensic Medicine University of Pavia, Pavia, Italy
| | - Valentina De Giorgis
- Department of Child Neurology and Psychiatry, IRCCS “C. Mondino” National Neurological Institute, Pavia, Italy
| | - Sara Olivotto
- Pediatric Neurology Unit, “V. Buzzi” Hospital, Milan, Italy
| | - Roberto Previtali
- Pediatric Neurology Unit, “V. Buzzi” Hospital, Milan, Italy
- Biomedical and Clinical Sciences Department, University of Milan, Milan, Italy
| | - Pierangelo Veggiotti
- Pediatric Neurology Unit, “V. Buzzi” Hospital, Milan, Italy
- Biomedical and Clinical Sciences Department, University of Milan, Milan, Italy
| | - Simona Bertoli
- ICANS-DIS, Department of Food Environmental and Nutritional Sciences, University of Milan, Milan, Italy
- Obesity Unit and Laboratory of Nutrition and Obesity Research, Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| |
Collapse
|
39
|
Skinner OS, Blanco-Fernández J, Goodman RP, Kawakami A, Shen H, Kemény LV, Joesch-Cohen L, Rees MG, Roth JA, Fisher DE, Mootha VK, Jourdain AA. Salvage of ribose from uridine or RNA supports glycolysis in nutrient-limited conditions. Nat Metab 2023; 5:765-776. [PMID: 37198474 PMCID: PMC10229423 DOI: 10.1038/s42255-023-00774-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/03/2023] [Indexed: 05/19/2023]
Abstract
Glucose is vital for life, serving as both a source of energy and carbon building block for growth. When glucose is limiting, alternative nutrients must be harnessed. To identify mechanisms by which cells can tolerate complete loss of glucose, we performed nutrient-sensitized genome-wide genetic screens and a PRISM growth assay across 482 cancer cell lines. We report that catabolism of uridine from the medium enables the growth of cells in the complete absence of glucose. While previous studies have shown that uridine can be salvaged to support pyrimidine synthesis in the setting of mitochondrial oxidative phosphorylation deficiency1, our work demonstrates that the ribose moiety of uridine or RNA can be salvaged to fulfil energy requirements via a pathway based on: (1) the phosphorylytic cleavage of uridine by uridine phosphorylase UPP1/UPP2 into uracil and ribose-1-phosphate (R1P), (2) the conversion of uridine-derived R1P into fructose-6-P and glyceraldehyde-3-P by the non-oxidative branch of the pentose phosphate pathway and (3) their glycolytic utilization to fuel ATP production, biosynthesis and gluconeogenesis. Capacity for glycolysis from uridine-derived ribose appears widespread, and we confirm its activity in cancer lineages, primary macrophages and mice in vivo. An interesting property of this pathway is that R1P enters downstream of the initial, highly regulated steps of glucose transport and upper glycolysis. We anticipate that 'uridine bypass' of upper glycolysis could be important in the context of disease and even exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Owen S Skinner
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | | | - Russell P Goodman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Akinori Kawakami
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Hongying Shen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
- Yale Systems Biology Institute, Yale West Campus, West Haven, CT, USA
| | - Lajos V Kemény
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | | | | | | | - David E Fisher
- Cutaneous Biology Research Center, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Vamsi K Mootha
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Molecular Biology and Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Alexis A Jourdain
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
40
|
Tayebi N, Leon‐Ricardo B, McCall K, Mehinovic E, Engelstad K, Huynh V, Turner TN, Weisenberg J, Thio LL, Hruz P, Williams RSB, De Vivo DC, Petit V, Haller G, Gurnett CA. Quantitative determination of SLC2A1 variant functional effects in GLUT1 deficiency syndrome. Ann Clin Transl Neurol 2023; 10:787-801. [PMID: 37000947 PMCID: PMC10187726 DOI: 10.1002/acn3.51767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 04/03/2023] Open
Abstract
OBJECTIVE The goal of this study is to demonstrate the utility of a growth assay to quantify the functional impact of single nucleotide variants (SNVs) in SLC2A1, the gene responsible for Glut1DS. METHODS The functional impact of 40 SNVs in SLC2A1 was quantitatively determined in HAP1 cells in which SLC2A1 is required for growth. Donor libraries were introduced into the endogenous SLC2A1 gene in HAP1-Lig4KO cells using CRISPR/Cas9. Cell populations were harvested and sequenced to quantify the effect of variants on growth and generate a functional score. Quantitative functional scores were compared to 3-OMG uptake, SLC2A1 cell surface expression, CADD score, and clinical data, including CSF/blood glucose ratio. RESULTS Nonsense variants (N = 3) were reduced in cell culture over time resulting in negative scores (mean score: -1.15 ± 0.17), whereas synonymous variants (N = 10) were not depleted (mean score: 0.25 ± 0.12) (P < 2e-16). Missense variants (N = 27) yielded a range of functional scores including slightly negative scores, supporting a partial function and intermediate phenotype. Several variants with normal results on either cell surface expression (p.N34S and p.W65R) or 3-OMG uptake (p.W65R) had negative functional scores. There is a moderate but significant correlation between our functional scores and CADD scores. INTERPRETATION Cell growth is useful to quantitatively determine the functional effects of SLC2A1 variants. Nonsense variants were reliably distinguished from benign variants in this in vitro functional assay. For facilitating early diagnosis and therapeutic intervention, future work is needed to determine the functional effect of every possible variant in SLC2A1.
Collapse
Affiliation(s)
- Naeimeh Tayebi
- Department of NeurologyWashington University in St LouisSt LouisMissouriUSA
| | - Brian Leon‐Ricardo
- Department of NeurologyWashington University in St LouisSt LouisMissouriUSA
| | - Kevin McCall
- Department of NeurologyWashington University in St LouisSt LouisMissouriUSA
| | - Elvisa Mehinovic
- Department of GeneticsWashington University in St LouisSt LouisMissouriUSA
| | - Kristin Engelstad
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Vincent Huynh
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Tychele N. Turner
- Department of GeneticsWashington University in St LouisSt LouisMissouriUSA
| | - Judy Weisenberg
- Department of NeurologyWashington University in St LouisSt LouisMissouriUSA
| | - Liu L. Thio
- Department of NeurologyWashington University in St LouisSt LouisMissouriUSA
| | - Paul Hruz
- Department of PediatricsWashington University in St LouisSt LouisMissouriUSA
| | - Robin S. B. Williams
- Centre for Biomedical Sciences, Department of Biological SciencesRoyal Holloway University of LondonEghamUK
| | - Darryl C. De Vivo
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | | | - Gabe Haller
- Department of NeurologyWashington University in St LouisSt LouisMissouriUSA
- Department of GeneticsWashington University in St LouisSt LouisMissouriUSA
- Department of Neurological SurgeryWashington University in St LouisSt LouisMissouriUSA
| | | |
Collapse
|
41
|
Bartolini E, Ferrari AR, Fiori S, Della Vecchia S. Glycaemic Imbalances in Seizures and Epilepsy of Paediatric Age: A Literature Review. J Clin Med 2023; 12:jcm12072580. [PMID: 37048663 PMCID: PMC10095009 DOI: 10.3390/jcm12072580] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/20/2023] [Accepted: 03/26/2023] [Indexed: 04/01/2023] Open
Abstract
Cerebral excitability and systemic metabolic balance are closely interconnected. Energy supply to neurons depends critically on glucose, whose fluctuations can promote immediate hyperexcitability resulting in acute symptomatic seizures. On the other hand, chronic disorders of sugar metabolism (e.g., diabetes mellitus) are often associated with long-term epilepsy. In this paper, we aim to review the existing knowledge on the association between acute and chronic glycaemic imbalances (hyper- and hypoglycaemia) with seizures and epilepsy, especially in the developing brain, focusing on clinical and instrumental features in order to optimize the care of children and adolescents and prevent the development of chronic neurological conditions in young patients.
Collapse
Affiliation(s)
- Emanuele Bartolini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy (A.R.F.)
- Tuscany PhD Programme in Neurosciences, 50139 Florence, Italy
| | - Anna Rita Ferrari
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy (A.R.F.)
| | - Simona Fiori
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy (A.R.F.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56128 Pisa, Italy
| | - Stefania Della Vecchia
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy (A.R.F.)
- Department of Molecular Medicine and Neurogenetics, IRCCS Stella Maris Foundation, 56128 Pisa, Italy
- Correspondence: ; Tel.: +39-050-886-332
| |
Collapse
|
42
|
Nabatame S, Tanigawa J, Tominaga K, Kagitani-Shimono K, Yanagihara K, Imai K, Ando T, Tsuyusaki Y, Araya N, Matsufuji M, Natsume J, Yuge K, Bratkovic D, Arai H, Okinaga T, Matsushige T, Azuma Y, Ishihara N, Miyatake S, Kato M, Matsumoto N, Okamoto N, Takahashi S, Hattori S, Ozono K. Association between cerebrospinal fluid parameters and developmental and neurological status in glucose transporter 1 deficiency syndrome. J Neurol Sci 2023; 447:120597. [PMID: 36965413 DOI: 10.1016/j.jns.2023.120597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 01/30/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023]
Abstract
OBJECTIVE In glucose transporter 1 deficiency syndrome (Glut1DS), cerebrospinal fluid glucose (CSFG) and CSFG to blood glucose ratio (CBGR) show significant differences among groups classified by phenotype or genotype. The purpose of this study was to investigate the association between these biochemical parameters and Glut1DS severity. METHODS The medical records of 45 patients who visited Osaka University Hospital between March 2004 and December 2021 were retrospectively examined. Neurological status was determined using the developmental quotient (DQ), assessed using the Kyoto Scale of Psychological Development 2001, and the Scale for the Assessment and Rating of Ataxia (SARA). CSF parameters included CSFG, CBGR, and CSF lactate (CSFL). RESULTS CSF was collected from 41 patients, and DQ and SARA were assessed in 24 and 27 patients, respectively. Simple regression analysis showed moderate associations between neurological status and biochemical parameters. CSFG resulted in a higher R2 than CBGR in these analyses. CSF parameters acquired during the first year of life were not comparable to those acquired later. CSFL was measured in 16 patients (DQ and SARA in 11 and 14 patients, respectively). Although simple regression analysis also showed moderate associations between neurological status and CSFG and CSFL, the multiple regression analysis for DQ and SARA resulted in strong associations through the use of a combination of CSFG and CSFL as explanatory variables. CONCLUSION The severity of Glut1DS can be predicted from CSF parameters. Glucose and lactate are independent contributors to the developmental and neurological status in Glut1DS.
Collapse
Affiliation(s)
- Shin Nabatame
- Department of Pediatrics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Junpei Tanigawa
- Department of Pediatrics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Koji Tominaga
- Department of Pediatrics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Kuriko Kagitani-Shimono
- Department of Pediatrics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Child Development, United Graduate School of Child Development, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Keiko Yanagihara
- Department of Pediatric Neurology, Osaka Women's and Children's Hospital, 840 Murodocho, Izumi, Osaka 594-1101, Japan.
| | - Katsumi Imai
- Department of Clinical Research, National Epilepsy Center, NHO Shizuoka Institute of Epilepsy and Neurological Disorders, 886 Urushiyama, Aoi, Shizuoka, Shizuoka 420-8688, Japan.
| | - Toru Ando
- Department of Pediatric Medicine, Municipal Tsuruga Hospital, 1-6-60, Mishimacho, Tsuruga, Fukui 914-8502, Japan.
| | - Yu Tsuyusaki
- Division of Neurology, Kanagawa Children's Medical Center, 2-138-4 Mutsukawa, Minami, Yokohama, Kanagawa 232-8555, Japan.
| | - Nami Araya
- Department of Pediatrics, School of Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba, Shiwa, Iwate 028-3695, Japan; Epilepsy Clinic Bethel Satellite Sendai-Station, Comfort Hotel Sendai-Higashiguchi #1F, 205-5 Nakakecho, Miyagino, Sendai, Miyagi 983-0864, Japan.
| | - Mayumi Matsufuji
- Department of Pediatrics, Kagoshima City Hospital, 37-1 Uearatacho, Kagoshima, Kagoshima 890-8760, Japan.
| | - Jun Natsume
- Department of Developmental Disability Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumaicho, Showa, Nagoya, Aichi 466-8550, Japan.
| | - Kotaro Yuge
- Department of Pediatrics and Child Health, Kurume University School of Medicine, 67 Asahimachi, Kurume, Fukuoka 830-0011, Japan.
| | - Drago Bratkovic
- Metabolic Clinic, Women's and Children's Hospital, 72 King William Rd, North Adelaide 5006, SA, Australia.
| | - Hiroshi Arai
- Department of Pediatric Neurology, Bobath Memorial Hospital, 1-6-5 Higashinakahama, Joto, Osaka, Osaka 536-0023, Japan.
| | - Takeshi Okinaga
- Department of Pediatrics, Bell Land General Hospital, 500-3 Higashiyama, Naka, Sakai, Osaka, 599-8247, Japan.
| | - Takeshi Matsushige
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan.
| | - Yoshiteru Azuma
- Department of Pediatrics, Aichi Medical University, 1-1, Yazakokarimata, Nagakute, Aichi 480-1195, Japan; Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa, Yokohama, Kanagawa 236-0004, Japan.
| | - Naoko Ishihara
- Department of Pediatrics, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukakecho, Toyoake, Aichi 470-1192, Japan.
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa, Yokohama, Kanagawa 236-0004, Japan; Clinical Genetics Department, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa, Yokohama, Kanagawa 236-0004, Japan.
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan.
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa, Yokohama, Kanagawa 236-0004, Japan.
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, 840 Murodocho, Izumi, Osaka 594-1101, Japan.
| | - Satoru Takahashi
- Department of Pediatrics, Asahikawa Medical University, 2-1-1-1 Midorigaoka-higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Satoshi Hattori
- Department of Biomedical Statistics, Graduate School of Medicine and Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Keiichi Ozono
- Department of Pediatrics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
43
|
Wu WZ, Bai YP. Endothelial GLUTs and vascular biology. Biomed Pharmacother 2023; 158:114151. [PMID: 36565587 DOI: 10.1016/j.biopha.2022.114151] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/15/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022] Open
Abstract
Endothelial metabolism is a promising target for vascular functional regulation and disease therapy. Glucose is the primary fuel for endothelial metabolism, supporting ATP generation and endothelial cell survival. Multiple studies have discussed the role of endothelial glucose catabolism, such as glycolysis and oxidative phosphorylation, in vascular functional remodeling. However, the role of the first gatekeepers of endothelial glucose utilization, glucose transporters, in the vasculature has long been neglected. Here, this review summarizes glucose transporter studies in vascular research. We mainly focus on GLUT1 and GLUT3 because they are the most critical glucose transporters responsible for most endothelial glucose uptake. Some interesting topics are also discussed, intending to provide directions for endothelial glucose transporter research in the future.
Collapse
Affiliation(s)
- Wan-Zhou Wu
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Center for Vascular Disease and Translational Medicine, Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yong-Ping Bai
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
44
|
Furtado J, Geraldo LH, Leser FS, Poulet M, Park H, Pibouin-Fragner L, Eichmann A, Boyé K. Netrin-1 binding to Unc5B regulates Blood-Retina Barrier integrity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.21.525006. [PMID: 36711611 PMCID: PMC9882365 DOI: 10.1101/2023.01.21.525006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Background The blood brain barrier (BBB) preserves neuronal function in the central nervous system (CNS) by tightly controlling metabolite exchanges with the blood. In the eye, the retina is likewise protected by the blood-retina barrier (BRB) to maintain phototransduction. We showed that the secreted guidance cue Netrin-1 regulated BBB integrity, by binding to endothelial Unc5B and regulating canonical β-catenin dependent expression of BBB gene expression. Objective Here, we investigated if Netrin-1-binding to endothelial Unc5B also controlled BRB integrity, and if this process involved Norrin/β-catenin signaling, which is the major known driver of BRB development and maintenance. Methods We analyzed Tamoxifen-inducible loss- and gain- of-function alleles of Unc5B, Ntn1 and Ctnnb1 in conjunction with tracer injections and biochemical signaling studies. Results Inducible endothelial Unc5B deletion, and inducible global Ntn1 deletion in postnatal mice reduced phosphorylation of the Norrin receptor LRP5, leading to reduced β-catenin and LEF1 expression, conversion of retina endothelial cells from a barrier-competent Claudin-5+/PLVAP- state to a Claudin-5-/PLVAP+ leaky phenotype, and extravasation of injected low molecular weight tracers. Inducible Ctnnb1 gain of function rescued vascular leak in Unc5B mutants, and Ntn1 overexpression induced BRB tightening. Unc5B expression in pericytes contributed to BRB permeability, via regulation of endothelial Unc5B. Mechanistically, Netrin-1-Unc5B signaling promoted β-catenin dependent BRB signaling by enhancing phosphorylation of the Norrin receptor LRP5 via the Discs large homologue 1 (Dlg1) intracellular scaffolding protein. Conclusions The data identify Netrin1-Unc5B as novel regulators of BRB integrity, with implications for diseases associated with BRB disruption.
Collapse
Affiliation(s)
- Jessica Furtado
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven CT, USA
| | - Luiz Henrique Geraldo
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven CT, USA
| | | | - Mathilde Poulet
- Paris Cardiovascular Research Center, Inserm U970, Université Paris, France
| | - Hyojin Park
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven CT, USA
| | | | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven CT, USA
- Paris Cardiovascular Research Center, Inserm U970, Université Paris, France
| | - Kevin Boyé
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven CT, USA
- Paris Cardiovascular Research Center, Inserm U970, Université Paris, France
| |
Collapse
|
45
|
Caraballo R. Ketogenic Diet and Drug-Resistant Epilepsy. PHARMACORESISTANCE IN EPILEPSY 2023:479-498. [DOI: 10.1007/978-3-031-36526-3_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
46
|
Peters TMA, Merx J, Kooijman PC, Noga M, de Boer S, van Gemert LA, Salden G, Engelke UFH, Lefeber DJ, van Outersterp RE, Berden G, Boltje TJ, Artuch R, Pías-Peleteiro L, García-Cazorla Á, Barić I, Thöny B, Oomens J, Martens J, Wevers RA, Verbeek MM, Coene KLM, Willemsen MAAP. Novel cerebrospinal fluid biomarkers of glucose transporter type 1 deficiency syndrome: Implications beyond the brain's energy deficit. J Inherit Metab Dis 2023; 46:66-75. [PMID: 36088537 PMCID: PMC10091941 DOI: 10.1002/jimd.12554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/26/2022] [Accepted: 09/07/2022] [Indexed: 01/19/2023]
Abstract
We used next-generation metabolic screening to identify new biomarkers for improved diagnosis and pathophysiological understanding of glucose transporter type 1 deficiency syndrome (GLUT1DS), comparing metabolic cerebrospinal fluid (CSF) profiles from 12 patients to those of 116 controls. This confirmed decreased CSF glucose and lactate levels in patients with GLUT1DS and increased glutamine at group level. We identified three novel biomarkers significantly decreased in patients, namely gluconic + galactonic acid, xylose-α1-3-glucose, and xylose-α1-3-xylose-α1-3-glucose, of which the latter two have not previously been identified in body fluids. CSF concentrations of gluconic + galactonic acid may be reduced as these metabolites could serve as alternative substrates for the pentose phosphate pathway. Xylose-α1-3-glucose and xylose-α1-3-xylose-α1-3-glucose may originate from glycosylated proteins; their decreased levels are hypothetically the consequence of insufficient glucose, one of two substrates for O-glucosylation. Since many proteins are O-glucosylated, this deficiency may affect cellular processes and thus contribute to GLUT1DS pathophysiology. The novel CSF biomarkers have the potential to improve the biochemical diagnosis of GLUT1DS. Our findings imply that brain glucose deficiency in GLUT1DS may cause disruptions at the cellular level that go beyond energy metabolism, underlining the importance of developing treatment strategies that directly target cerebral glucose uptake.
Collapse
Affiliation(s)
- Tessa M A Peters
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jona Merx
- Institute for Molecules and Materials, Synthetic Organic Chemistry, Radboud University, Nijmegen, The Netherlands
| | - Pieter C Kooijman
- Institute for Molecules and Materials, FELIX Laboratory, Radboud University, Nijmegen, The Netherlands
| | - Marek Noga
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Siebolt de Boer
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Loes A van Gemert
- Amalia Children's Hospital, Department of Pediatric Neurology & Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Guido Salden
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Udo F H Engelke
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dirk J Lefeber
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rianne E van Outersterp
- Institute for Molecules and Materials, FELIX Laboratory, Radboud University, Nijmegen, The Netherlands
| | - Giel Berden
- Institute for Molecules and Materials, FELIX Laboratory, Radboud University, Nijmegen, The Netherlands
| | - Thomas J Boltje
- Institute for Molecules and Materials, Synthetic Organic Chemistry, Radboud University, Nijmegen, The Netherlands
| | - Rafael Artuch
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, CIBERER and MetabERN Hospital Sant Joan de Déu, Barcelona, Spain
| | - Leticia Pías-Peleteiro
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, CIBERER and MetabERN Hospital Sant Joan de Déu, Barcelona, Spain
| | - Ángeles García-Cazorla
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, CIBERER and MetabERN Hospital Sant Joan de Déu, Barcelona, Spain
| | - Ivo Barić
- Department of Pediatrics, University Hospital Center Zagreb & University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Beat Thöny
- Division of Metabolism, University Children's Hospital Zürich, Zürich, Switzerland
| | - Jos Oomens
- Institute for Molecules and Materials, FELIX Laboratory, Radboud University, Nijmegen, The Netherlands
| | - Jonathan Martens
- Institute for Molecules and Materials, FELIX Laboratory, Radboud University, Nijmegen, The Netherlands
| | - Ron A Wevers
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel M Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Karlien L M Coene
- Department of Laboratory Medicine, Translational Metabolic Laboratory (TML), Radboud University Medical Center, Nijmegen, The Netherlands
- Laboratory of Clinical Chemistry and Hematology, Elisabeth TweeSteden Hospital, Tilburg, The Netherlands
| | - Michèl A A P Willemsen
- Amalia Children's Hospital, Department of Pediatric Neurology & Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
47
|
Kyoko N, Masakazu H, Shin N, Hidetaka S, Narikazu U. Perioperative management of surgical orthodontic treatment in a patient with glucose transporter 1 deficiency: report of a case and review of the literature. Perioper Med (Lond) 2022; 11:55. [PMID: 36527119 PMCID: PMC9756684 DOI: 10.1186/s13741-022-00287-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Glucose transporter 1 (GLUT1) deficiency is a rare cerebral metabolic disorder caused by the shortage of glucose supply to the brain. For this disease, ketogenic diet therapy is essential. In addition, perioperative management requires not only the continuation of ketogenic diet therapy but also the management of nausea/vomiting, diarrhea, seizures, and infection. However, there have been few reports regarding oral and maxillofacial surgery. CASE PRESENTATION We describe a patient with GLUT1 deficiency who underwent orthognathic surgery. An 18-year-old man was referred to our hospital with the chief complaint of mandibular regression. Surgical tolerance was assessed by a fasting test and tooth extraction under general anesthesia, and orthognathic surgery was then performed. For orthognathic surgery, the mandibular dentition had scissor-like occlusion, and it was difficult to arrange the mandible. Therefore, we decided to perform maxillary osteotomy first. After the mandibular dentition was arranged by maxillary osteotomy, sagittal split ramus osteotomy (SSRO) was performed. Intermaxillary fixation (IMF) was necessary for SSRO, and caution was needed to prevent suffocation. The orthognathic surgery was successful, although complications, such as vomiting, diarrhea, and seizures, developed. CONCLUSION Surgical orthodontic treatment in GLUT1 deficiency can be performed relatively safely by maintaining the diet, taking measures against epilepsy and vomiting, and using antimicrobial agents in close collaboration with pediatricians, anesthesiologists, pharmacists, and nutritionists.
Collapse
Affiliation(s)
- Nishiyama Kyoko
- grid.136593.b0000 0004 0373 3971Department of Oral and Maxillofacial Surgery II, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka, 565-0871 Japan
| | - Hamada Masakazu
- grid.136593.b0000 0004 0373 3971Department of Oral and Maxillofacial Surgery II, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka, 565-0871 Japan
| | - Nabatame Shin
- grid.136593.b0000 0004 0373 3971Department of Pediatrics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871 Japan
| | - Shimizu Hidetaka
- grid.136593.b0000 0004 0373 3971Department of Oral and Maxillofacial Surgery II, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka, 565-0871 Japan ,grid.416633.5Department of Oral and Maxillofacial Surgery, Saiseikai Suita Hospital, 1-2 Kawazonocho, Suita, Osaka, 564-0013 Japan
| | - Uzawa Narikazu
- grid.136593.b0000 0004 0373 3971Department of Oral and Maxillofacial Surgery II, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka, 565-0871 Japan
| |
Collapse
|
48
|
Porthukaran A, Zak M, Moharir M, Mamak E, Sinopoli KJ. Neuropsychological Outcome of Glucose Transporter-1 Deficiency Syndrome: a Case Study of Identical Twin Boys Without Intellectual Disability. JOURNAL OF PEDIATRIC NEUROPSYCHOLOGY 2022. [DOI: 10.1007/s40817-022-00130-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
49
|
Mauri A, Duse A, Palm G, Previtali R, Bova SM, Olivotto S, Benedetti S, Coscia F, Veggiotti P, Cereda C. Molecular Genetics of GLUT1DS Italian Pediatric Cohort: 10 Novel Disease-Related Variants and Structural Analysis. Int J Mol Sci 2022; 23:ijms232113560. [PMID: 36362347 PMCID: PMC9654628 DOI: 10.3390/ijms232113560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022] Open
Abstract
GLUT1 deficiency syndrome (GLUT1DS1; OMIM #606777) is a rare genetic metabolic disease, characterized by infantile-onset epileptic encephalopathy, global developmental delay, progressive microcephaly, and movement disorders (e.g., spasticity and dystonia). It is caused by heterozygous mutations in the SLC2A1 gene, which encodes the GLUT1 protein, a glucose transporter across the blood-brain barrier (BBB). Most commonly, these variants arise de novo resulting in sporadic cases, although several familial cases with AD inheritance pattern have been described. Twenty-seven Italian pediatric patients, clinically suspect of GLUT1DS from both sporadic and familial cases, have been enrolled. We detected by trios sequencing analysis 25 different variants causing GLUT1DS. Of these, 40% of the identified variants (10 out of 25) had never been reported before, including missense, frameshift, and splice site variants. Their structural mapping on the X-ray structure of GLUT1 strongly suggested the potential pathogenic effects of these novel disease-related mutations, broadening the genotypic spectrum heterogeneity found in the SLC2A1 gene. Moreover, 24% is located in a vulnerable region of the GLUT1 protein that involves transmembrane 4 and 5 helices encoded by exon 4, confirming a mutational hotspot in the SLC2A1 gene. Lastly, we investigated possible correlations between mutation type and clinical and biochemical data observed in our GLUT1DS cohort, revealing that splice site and frameshift variants are related to a more severe phenotype and low CSF parameters.
Collapse
Affiliation(s)
- Alessia Mauri
- Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy
- Newborn Screening and Genetic Metabolic Diseases Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Alessandra Duse
- Pediatric Neurology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Giacomo Palm
- Structural Biology Center, Human Technopole, 20157 Milan, Italy
| | - Roberto Previtali
- Pediatric Neurology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | | | - Sara Olivotto
- Pediatric Neurology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Sara Benedetti
- Newborn Screening and Genetic Metabolic Diseases Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | | | - Pierangelo Veggiotti
- Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy
- Pediatric Neurology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Cristina Cereda
- Newborn Screening and Genetic Metabolic Diseases Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy
- Correspondence:
| |
Collapse
|
50
|
Yazdani S, Bilan PJ, Jaldin-Fincati JR, Pang J, Ceban F, Saran E, Brumell JH, Freeman SA, Klip A. Dynamic glucose uptake, storage, and release by human microvascular endothelial cells. Mol Biol Cell 2022; 33:ar106. [PMID: 35921166 DOI: 10.1091/mbc.e22-04-0146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Endothelia determine blood-to-tissue solute delivery, yet glucose transit is poorly understood. To illuminate mechanisms, we tracked [3H]-2-deoxyglucose (2-DG) in human adipose-tissue microvascular endothelial cells. 2-DG uptake was largely facilitated by the glucose transporters GLUT1 and GLUT3. Once in the cytosol, >80% of 2-DG became phosphorylated and ∼20% incorporated into glycogen, suggesting that transported glucose is readily accessible to cytosolic enzymes. Interestingly, a fraction of intracellular 2-DG was released over time (15-20% over 30 min) with slower kinetics than for uptake, involving GLUT3. In contrast to intracellular 2-DG, the released 2-DG was largely unphosphorylated. Glucose release involved endoplasmic reticulum-resident translocases/phosphatases and was stimulated by adrenaline, consistent with participation of glycogenolysis and glucose dephosphorylation. Surprisingly, the fluorescent glucose derivative 2-NBD-glucose (2-NBDG) entered cells largely via fluid phase endocytosis and exited by recycling. 2-NBDG uptake was insensitive to GLUT1/GLUT3 inhibition, suggesting poor influx across membranes. 2-NBDG recycling, but not 2-DG efflux, was sensitive to N-ethyl maleimide. In sum, by utilizing radioactive and fluorescent glucose derivatives, we identified two parallel routes of entry: uptake into the cytosol through dedicated glucose transporters and endocytosis. This reveals the complex glucose handling by endothelial cells that may contribute to glucose delivery to tissues.
Collapse
Affiliation(s)
- Samaneh Yazdani
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada, M5G 0A4
| | - Philip J Bilan
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada, M5G 0A4
| | | | - Janice Pang
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada, M5G 0A4
| | - Felicia Ceban
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada, M5G 0A4
| | - Ekambir Saran
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada, M5G 0A4
| | - John H Brumell
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada, M5G 0A4.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada, M5S 1A1.,SickKids IBD Centre, Hospital for Sick Children, Toronto, ON, Canada, M5G 0A4
| | - Spencer A Freeman
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada, M5G 0A4.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada, M5S 1A1
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada, M5G 0A4.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Department of Paediatrics, University of Toronto, Toronto, ON, Canada, M5S 1A1.,Department of Physiology, University of Toronto, Toronto, ON, Canada, M5S 1A1
| |
Collapse
|