51
|
Olivotto S, Duse A, Bova SM, Leonardi V, Biganzoli E, Milanese A, Cereda C, Bertoli S, Previtali R, Veggiotti P. Glut1 deficiency syndrome throughout life: clinical phenotypes, intelligence, life achievements and quality of life in familial cases. Orphanet J Rare Dis 2022; 17:365. [PMID: 36153584 PMCID: PMC9509642 DOI: 10.1186/s13023-022-02513-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/04/2022] [Indexed: 11/28/2022] Open
Abstract
Background Glut1 deficiency syndrome (Glut1-DS) is a rare metabolic encephalopathy. Familial forms are poorly investigated, and no previous studies have explored aspects of Glut1-DS over the course of life: clinical pictures, intelligence, life achievements, and quality of life in adulthood. Clinical, biochemical and genetic data in a cohort of familial Glut1-DS cases were collected from medical records. Intelligence was assessed using Raven’s Standard Progressive Matrices and Raven’s Colored Progressive Matrices in adults and children, respectively. An ad hoc interview focusing on life achievements and the World Health Organization Quality of Life Questionnaire were administered to adult subjects. Results The clinical picture in adults was characterized by paroxysmal exercise-induced dyskinesia (PED) (80%), fatigue (60%), low intelligence (60%), epilepsy (50%), and migraine (50%). However, 20% of the adults had higher-than-average intelligence. Quality of Life (QoL) seemed unrelated to the presence of PED or fatigue in adulthood. An association of potential clinical relevance, albeit not statistically significant, was found between intelligence and QoL. The phenotype of familial Glut1-DS in children was characterized by epilepsy (83.3%), intellectual disability (50%), and PED (33%). Conclusion The phenotype of familial Glut1-DS shows age-related differences: epilepsy predominates in childhood; PED and fatigue, followed by epilepsy and migraine, characterize the condition in adulthood. Some adults with familial Glut1-DS may lead regular and fulfilling lives, enjoying the same QoL as unaffected individuals. The disorder tends to worsen from generation to generation, with new and more severe symptoms arising within the same family. Epigenetic studies might be useful to assess the phenotypic variability in Glut1-DS. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02513-4.
Collapse
|
52
|
Abstract
To maintain energy supply to the brain, a direct energy source called adenosine triphosphate (ATP) is produced by oxidative phosphorylation and aerobic glycolysis of glucose in the mitochondria and cytoplasm. Brain glucose metabolism is reduced in many neurodegenerative diseases, including Alzheimer's disease (AD), where it appears presymptomatically in a progressive and region-specific manner. Following dysregulation of energy metabolism in AD, many cellular repair/regenerative processes are activated to conserve the energy required for cell viability. Glucose metabolism plays an important role in the pathology of AD and is closely associated with the tricarboxylic acid cycle, type 2 diabetes mellitus, and insulin resistance. The glucose intake in neurons is from endothelial cells, astrocytes, and microglia. Damage to neurocentric glucose also damages the energy transport systems in AD. Gut microbiota is necessary to modulate bidirectional communication between the gastrointestinal tract and brain. Gut microbiota may influence the process of AD by regulating the immune system and maintaining the integrity of the intestinal barrier. Furthermore, some therapeutic strategies have shown promising therapeutic effects in the treatment of AD at different stages, including the use of antidiabetic drugs, rescuing mitochondrial dysfunction, and epigenetic and dietary intervention. This review discusses the underlying mechanisms of alterations in energy metabolism in AD and provides potential therapeutic strategies in the treatment of AD.
Collapse
|
53
|
Zhao X, Ning H, Wang Y, Zhao G, Mei S, Liu N, Wang C, Cai A, Wei E, Kong X. Genetic analysis and identification of novel variations in Chinese patients with pediatric epilepsy by whole-exome sequencing. Neurol Sci 2022; 43:4439-4451. [DOI: 10.1007/s10072-022-05953-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/12/2022] [Indexed: 11/28/2022]
|
54
|
One Molecule for Mental Nourishment and More: Glucose Transporter Type 1—Biology and Deficiency Syndrome. Biomedicines 2022; 10:biomedicines10061249. [PMID: 35740271 PMCID: PMC9219734 DOI: 10.3390/biomedicines10061249] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/17/2022] [Accepted: 05/23/2022] [Indexed: 01/27/2023] Open
Abstract
Glucose transporter type 1 (Glut1) is the main transporter involved in the cellular uptake of glucose into many tissues, and is highly expressed in the brain and in erythrocytes. Glut1 deficiency syndrome is caused mainly by mutations of the SLC2A1 gene, impairing passive glucose transport across the blood–brain barrier. All age groups, from infants to adults, may be affected, with age-specific symptoms. In its classic form, the syndrome presents as an early-onset drug-resistant metabolic epileptic encephalopathy with a complex movement disorder and developmental delay. In later-onset forms, complex motor disorder predominates, with dystonia, ataxia, chorea or spasticity, often triggered by fasting. Diagnosis is confirmed by hypoglycorrhachia (below 45 mg/dL) with normal blood glucose, 18F-fluorodeoxyglucose positron emission tomography, and genetic analysis showing pathogenic SLC2A1 variants. There are also ongoing positive studies on erythrocytes’ Glut1 surface expression using flow cytometry. The standard treatment still consists of ketogenic therapies supplying ketones as alternative brain fuel. Anaplerotic substances may provide alternative energy sources. Understanding the complex interactions of Glut1 with other tissues, its signaling function for brain angiogenesis and gliosis, and the complex regulation of glucose transportation, including compensatory mechanisms in different tissues, will hopefully advance therapy. Ongoing research for future interventions is focusing on small molecules to restore Glut1, metabolic stimulation, and SLC2A1 transfer strategies. Newborn screening, early identification and treatment could minimize the neurodevelopmental disease consequences. Furthermore, understanding Glut1 relative deficiency or inhibition in inflammation, neurodegenerative disorders, and viral infections including COVID-19 and other settings could provide clues for future therapeutic approaches.
Collapse
|
55
|
Zuberi SM, Wirrell E, Yozawitz E, Wilmshurst JM, Specchio N, Riney K, Pressler R, Auvin S, Samia P, Hirsch E, Galicchio S, Triki C, Snead OC, Wiebe S, Cross JH, Tinuper P, Scheffer IE, Perucca E, Moshé SL, Nabbout R. ILAE classification and definition of epilepsy syndromes with onset in neonates and infants: Position statement by the ILAE Task Force on Nosology and Definitions. Epilepsia 2022; 63:1349-1397. [PMID: 35503712 DOI: 10.1111/epi.17239] [Citation(s) in RCA: 397] [Impact Index Per Article: 132.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 12/20/2022]
Abstract
The International League Against Epilepsy (ILAE) Task Force on Nosology and Definitions proposes a classification and definition of epilepsy syndromes in the neonate and infant with seizure onset up to 2 years of age. The incidence of epilepsy is high in this age group and epilepsy is frequently associated with significant comorbidities and mortality. The licensing of syndrome specific antiseizure medications following randomized controlled trials and the development of precision, gene-related therapies are two of the drivers defining the electroclinical phenotypes of syndromes with onset in infancy. The principal aim of this proposal, consistent with the 2017 ILAE Classification of the Epilepsies, is to support epilepsy diagnosis and emphasize the importance of classifying epilepsy in an individual both by syndrome and etiology. For each syndrome, we report epidemiology, clinical course, seizure types, electroencephalography (EEG), neuroimaging, genetics, and differential diagnosis. Syndromes are separated into self-limited syndromes, where there is likely to be spontaneous remission and developmental and epileptic encephalopathies, diseases where there is developmental impairment related to both the underlying etiology independent of epileptiform activity and the epileptic encephalopathy. The emerging class of etiology-specific epilepsy syndromes, where there is a specific etiology for the epilepsy that is associated with a clearly defined, relatively uniform, and distinct clinical phenotype in most affected individuals as well as consistent EEG, neuroimaging, and/or genetic correlates, is presented. The number of etiology-defined syndromes will continue to increase, and these newly described syndromes will in time be incorporated into this classification. The tables summarize mandatory features, cautionary alerts, and exclusionary features for the common syndromes. Guidance is given on the criteria for syndrome diagnosis in resource-limited regions where laboratory confirmation, including EEG, MRI, and genetic testing, might not be available.
Collapse
Affiliation(s)
- Sameer M Zuberi
- Paediatric Neurosciences Research Group, Royal Hospital for Children, Institute of Health & Wellbeing, Collaborating Centre of European Reference Network EpiCARE, University of Glasgow, Glasgow, UK
| | - Elaine Wirrell
- Divisions of Child and Adolescent Neurology and Epilepsy, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elissa Yozawitz
- Isabelle Rapin Division of Child Neurology, Saul R. Korey Department of Neurology, Montefiore Medical Center, Bronx, New York, USA
| | - Jo M Wilmshurst
- Department of Paediatric Neurology, Red Cross War Memorial Children's Hospital, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Nicola Specchio
- Rare and Complex Epilepsy Unit, Department of Neuroscience, Bambino Gesu' Children's Hospital, IRCCS, Member of European Reference Network EpiCARE, Rome, Italy
| | - Kate Riney
- Neurosciences Unit, Queensland Children's Hospital, South Brisbane, Queensland, Australia.,Faculty of Medicine, University of Queensland, St Lucia, Queensland, Australia
| | - Ronit Pressler
- Clinical Neuroscience, UCL- Great Ormond Street Institute of Child Health, London, UK.,Department of Clinical Neurophysiology, Great Ormond Street Hospital for Children NHS Foundation Trust, Member of European Reference Network EpiCARE, London, UK
| | - Stephane Auvin
- AP-HP, Hôpital Robert-Debré, INSERM NeuroDiderot, DMU Innov-RDB, Neurologie Pédiatrique, Member of European Reference Network EpiCARE, Université de Paris, Paris, France
| | - Pauline Samia
- Department of Paediatrics and Child Health, Aga Khan University, Nairobi, Kenya
| | - Edouard Hirsch
- Neurology Epilepsy Unit "Francis Rohmer", INSERM 1258, FMTS, Strasbourg University, Strasbourg, France
| | - Santiago Galicchio
- Child Neurology Department, Victor J Vilela Child Hospital of Rosario, Santa Fe, Argentina
| | - Chahnez Triki
- Child Neurology Department, LR19ES15 Neuropédiatrie, Sfax Medical School, University of Sfax, Sfax, Tunisia
| | - O Carter Snead
- Pediatric Neurology, Hospital for Sick Children, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Samuel Wiebe
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - J Helen Cross
- Programme of Developmental Neurosciences, UCL NIHR BRC Great Ormond Street Institute of Child Health, Great Ormond Street Hospital for Children, Member of European Reference Network EpiCARE, London, UK.,Young Epilepsy, Lingfield, UK
| | - Paolo Tinuper
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,IRCCS Istituto delle Scienze Neurologiche, Bologna, Italy
| | - Ingrid E Scheffer
- Austin Health and Royal Children's Hospital, Florey Institute, Murdoch Children's Research Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Emilio Perucca
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Solomon L Moshé
- Isabelle Rapin Division of Child Neurology, Saul R. Korey Department of Neurology, Bronx, New York, USA.,Departments of Neuroscience and Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA.,Montefiore Medical Center, Bronx, New York, USA
| | - Rima Nabbout
- Reference Centre for Rare Epilepsies, Department of Pediatric Neurology, Necker-Enfants Malades University Hospital, APHP, Member of European Reference Network EpiCARE, Institut Imagine, INSERM, UMR 1163, Université Paris cité, Paris, France
| |
Collapse
|
56
|
Lee H, Xu Y, Zhu X, Jang C, Choi W, Bae H, Wang W, He L, Jin S, Arany Z, Simons M. Endothelium-derived lactate is required for pericyte function and blood-brain barrier maintenance. EMBO J 2022; 41:e109890. [PMID: 35243676 PMCID: PMC9058541 DOI: 10.15252/embj.2021109890] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 01/05/2023] Open
Abstract
Endothelial cells differ from other cell types responsible for the formation of the vascular wall in their unusual reliance on glycolysis for most energy needs, which results in extensive production of lactate. We find that endothelium-derived lactate is taken up by pericytes, and contributes substantially to pericyte metabolism including energy generation and amino acid biosynthesis. Endothelial-pericyte proximity is required to facilitate the transport of endothelium-derived lactate into pericytes. Inhibition of lactate production in the endothelium by deletion of the glucose transporter-1 (GLUT1) in mice results in loss of pericyte coverage in the retina and brain vasculatures, leading to the blood-brain barrier breakdown and increased permeability. These abnormalities can be largely restored by oral lactate administration. Our studies demonstrate an unexpected link between endothelial and pericyte metabolisms and the role of endothelial lactate production in the maintenance of the blood-brain barrier integrity. In addition, our observations indicate that lactate supplementation could be a useful therapeutic approach for GLUT1 deficiency metabolic syndrome patients.
Collapse
Affiliation(s)
- Heon‐Woo Lee
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Yanying Xu
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- Department of Cardiovascular MedicineXiangya HospitalCentral South UniversityChangshaChina
| | - Xiaolong Zhu
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
| | - Cholsoon Jang
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Woosoung Choi
- School of Life Sciences and Cell Logistics Research CenterGwangju Institute of Science and Technology (GIST)GwangjuKorea
| | - Hosung Bae
- Department of Biological ChemistryUniversity of California IrvineIrvineCAUSA
| | - Weiwei Wang
- W. M. Keck Biotechnology Resource LaboratoryYale University School of MedicineNew HavenCTUSA
| | - Liqun He
- Department of Immunology, Genetics and PathologyRudbeck LaboratoryUppsala UniversityUppsalaSweden
| | - Suk‐Won Jin
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- School of Life Sciences and Cell Logistics Research CenterGwangju Institute of Science and Technology (GIST)GwangjuKorea
| | - Zoltan Arany
- Cardiovascular InstitutePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Michael Simons
- Yale Cardiovascular Research CenterSection of Cardiovascular MedicineDepartment of Internal MedicineYale University School of MedicineNew HavenCTUSA
- Department of Cell BiologyYale University School of MedicineNew HavenCTUSA
| |
Collapse
|
57
|
van Gemert LA, Leen WG, Draaisma JM, Roeleveld N, Willemsen MA. Head circumference in glucose transporter 1 deficiency syndrome: Normal for individuals, abnormal as a group. Eur J Paediatr Neurol 2022; 38:73-76. [PMID: 35500464 DOI: 10.1016/j.ejpn.2022.04.005] [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: 12/23/2021] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 10/18/2022]
Abstract
In the literature, microcephaly is considered as part of the classical phenotype of glucose transporter 1 deficiency syndrome (GLUT1DS), and previous cohort studies reported a prevalence of microcephaly of around 50%. In our clinical experience, however, only very few patients with GLUT1DS appear to have microcephaly. Therefore, we conducted an observational study among a large cohort of Dutch patients with GLUT1DS to investigate the prevalence of microcephaly, defined as < 2 standard deviations (SD) below the mean. We analysed the head circumference of 54 patients and found a prevalence of microcephaly at last known measurement of 6.5%. Notably, none of the patients had a head circumference < -3 SD. However, we learned that 75.9% of the patients had a head circumference below 0 SD. This study shows that microcephaly occurs less often than previously thought in patients with GLUT1DS, and that primary or secondary microcephaly does not seem to be a sign for clinicians to suspect GLUT1DS. As a group, however, patients with GLUT1DS seem to have decreased head circumference compared to healthy individuals and as such, our study suggests that early brain development and brain growth may be compromised in GLUT1DS.
Collapse
Affiliation(s)
- Loes A van Gemert
- Department of Pediatric Neurology, Amalia Children's Hospital, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, the Netherlands.
| | - Wilhelmina G Leen
- Department of Neurology, Canisius Wilhelmina Hospital, Weg Door Jonkerbos 100, 6532, SZ, Nijmegen, the Netherlands.
| | - Jos M Draaisma
- Department of General Pediatrics, Amalia Children's Hospital, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, the Netherlands.
| | - Nel Roeleveld
- Department for Health Evidence, Radboud Institute for Health Sciences, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, the Netherlands.
| | - Michèl A Willemsen
- Department of Pediatric Neurology, Amalia Children's Hospital, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Montessorilaan 3, 6525, HR, Nijmegen, the Netherlands.
| |
Collapse
|
58
|
Li W, Zou J, Shang J, Gao C, Sun R, Liu R, Cao H, Wang Y, Zhang J. Both the Complexity of Tight Junctions and Endothelial Transcytosis Are Increased During BBB Postnatal Development in Rats. Front Neurosci 2022; 16:850857. [PMID: 35573303 PMCID: PMC9095945 DOI: 10.3389/fnins.2022.850857] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/14/2022] [Indexed: 12/01/2022] Open
Abstract
The blood-brain barrier (BBB) comprises a single layer of endothelial cells and maintains a safe and homeostatic environment for proper neuronal function and synaptic transmission. BBB is not a discrete physical barrier, but a complex, dynamic, and adaptable interface. BBB continues to mature under the influence of the neural environment within a short period of time after birth. However, the basic mechanism of BBB formation and maintenance remains a mystery. Early studies have identified two structural characteristics of microvascular endothelium: special tight junctions (TJs) and a very low transcellular vesicle transport rate. Previous studies believed that BBB damage was mainly due to the destruction of tight junctions, and the role of vesicle transcytosis was neglected, so there was a lack of research on its impact on blood-brain barrier. It is urgent to get a better clarification of the unique structural and functional characteristics of the BBB endothelium to explain the role of BBB injury in neurological diseases. RNA sequencing was used to study the molecular characterization of cerebral cortex vascular endothelium by isolating them from neonatal, adolescent and adult rats. For investigation the maintenance mechanism of the BBB, we focused on the cellular and molecular regulation of barrier formation and the two characteristics of microvascular endothelial cells. Interestingly, we found that during the development of the blood-brain barrier, although the tight junctions gradually mature, endothelial cell transcytosis is gradually enhanced, resulting in an increase in the permeability of the blood-brain barrier. This study suggested that under physiological conditions, low vesicle transport is playing an important role in maintaining the integrity of the blood-brain barrier. This study not only summarized the unique characteristics of microvascular endothelial cells, but also illustrated a clarified mechanism of the development and maintenance of BBB which can provide new therapeutic opportunities for central nervous system drug delivery. Raw data of RNA sequencing were deposited in NCBI Sequence Read Archive database (PRJNA790676).
Collapse
Affiliation(s)
- Wei Li
- Department of Neurology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Jinlong Zou
- Department of Neurology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Junkui Shang
- Department of Neurology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Chenhao Gao
- Department of Neurology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Ruihua Sun
- Department of Neurology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Ruijie Liu
- Department of Neurology, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Huixia Cao
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Yanliang Wang
- Department of Nephrology, Henan Provincial Key Laboratory of Kidney Disease and Immunology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Henan University People’s Hospital, Zhengzhou, China
| | - Jiewen Zhang
- Department of Neurology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
59
|
Striano P, Auvin S, Collins A, Horvath R, Scheffer IE, Tzadok M, Miller I, Koenig MK, Lacy A, Davis R, Garcia-Cazorla A, Saneto RP, Brandabur M, Blair S, Koutsoukos T, De Vivo D. A randomized, double-blind trial of triheptanoin for drug-resistant epilepsy in glucose transporter I deficiency syndrome (Glut1DS). Epilepsia 2022; 63:1748-1760. [PMID: 35441706 PMCID: PMC9546029 DOI: 10.1111/epi.17263] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Evaluate efficacy and long-term safety of triheptanoin in patients >1 year old, not on a ketogenic diet, with drug-resistant seizures associated with Glucose Transporter Type 1 Deficiency Syndrome (Glut1DS). METHODS UX007G-CL201 was a randomized, double-blind, placebo-controlled trial. Following a 6-week baseline period, eligible patients were randomized 3:1 to triheptanoin or placebo. Dosing was titrated to 35% total daily calories over 2 weeks. After an 8-week placebo-controlled period, all patients received open-label triheptanoin through Week 52. RESULTS The study included 36 patients (15 children; 13 adolescents; 8 adults). A median 12.6% reduction in overall seizure frequency was observed in the triheptanoin arm relative to baseline and a 13.5% difference was observed relative to placebo (p = .58). In patients with absence seizures only (n = 9), a median 62.2% reduction in seizure frequency was observed in the triheptanoin arm relative to baseline. Only one patient with absence seizures only was present in the control group, preventing comparison. No statistically significant differences in seizure frequency were observed. Common treatment-emergent adverse events (TEAEs) included diarrhea, vomiting, abdominal pain, and nausea, most mild or moderate in severity. No serious AEs were considered treatment related. One patient discontinued due to status epilepticus. SIGNIFICANCE Triheptanoin did not significantly reduce seizure frequency in patients with Glut1DS not on the ketogenic diet. Treatment was associated with mild to moderate GI treatment-related events; most resolved following dose reduction or interruption and/or medication for treatment. Triheptanoin was not associated with any long-term safety concerns when administered at dose levels up to 35% total daily caloric intake for up to one year.
Collapse
Affiliation(s)
- Pasquale Striano
- IRCCS Istituto 'G. Gaslini', Genoa, Italy.,Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Stéphane Auvin
- Robert-Debré University Hospital and Université de Paris, Paris, France.,Institut Universitaire de France (IUF), Paris, France
| | | | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Ingrid E Scheffer
- Austin and Royal Children's Hospital, Florey and Murdoch Institutes, University of Melbourne, Melbourne, Vic., Australia
| | - Michal Tzadok
- Pediatric Neurology Units, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Affiliated to the Sackler Faculty of Medicine, Tel-Aviv University, Ramat Gan, Israel
| | - Ian Miller
- Miami Children's Research Institute, Miami, Florida, USA
| | | | - Adrian Lacy
- Cook Children's Medical Center, Fort Worth, Texas, USA
| | - Ronald Davis
- Neurology & Epilepsy Research Center, DBO Pediatric Neurology, P.A., Orlando, Florida, USA
| | | | - Russell P Saneto
- Department of Neurology, Division of Pediatric Neurology, University of Washington/ Seattle Children's Hospital, Seattle, Washington, USA
| | | | - Susan Blair
- Ultragenyx Pharmaceutical Inc., Novato, California, USA
| | | | - Darryl De Vivo
- Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
60
|
Sánchez-Lijarcio O, Yubero D, Leal F, Couce ML, Luis GGS, López-Laso E, García-Cazorla À, Pías-Peleteiro L, de Azua Brea B, Ibáñez-Micó S, Martínez GM, Schifferli MT, Enriquez SW, Ugarte M, Artuch R, Pérez B. The clinical and biochemical hallmarks generally associated with GLUT1DS may be caused by defects in genes other than SLC2A1. Clin Genet 2022; 102:40-55. [PMID: 35388452 PMCID: PMC9325084 DOI: 10.1111/cge.14138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/27/2022]
Abstract
Glucose transporter 1 deficiency syndrome (GLUT1DS) is a neurometabolic disorder caused by haploinsufficiency of the GLUT1 glucose transporter (encoded by SLC2A1) leading to defective glucose transport across the blood–brain barrier. This work describes the genetic analysis of 56 patients with clinical or biochemical GLUT1DS hallmarks. 55.4% of these patients had a pathogenic variant of SLC2A1, and 23.2% had a variant in one of 13 different genes. No pathogenic variant was identified for the remaining patients. Expression analysis of SLC2A1 indicated a reduction in SLC2A1 mRNA in patients with pathogenic variants of this gene, as well as in one patient with a pathogenic variant in SLC9A6, and in three for whom no candidate variant was identified. Thus, the clinical and biochemical hallmarks generally associated with GLUT1DS may be caused by defects in genes other than SLC2A1.
Collapse
Affiliation(s)
- Obdulia Sánchez-Lijarcio
- Centro de Diagnóstico de Enfermedades Moleculares, Center of Molecular Biology Severo Ochoa (CBMSO), Autonomous University of Madrid, CIBERER, IdiPAZ, Madrid, Spain
| | - Delia Yubero
- Sant Joan de Déu Research Institute, CIBERER, Barcelona, Spain
| | - Fátima Leal
- Centro de Diagnóstico de Enfermedades Moleculares, Center of Molecular Biology Severo Ochoa (CBMSO), Autonomous University of Madrid, CIBERER, IdiPAZ, Madrid, Spain
| | - María L Couce
- Unit for the Diagnosis and Treatment of Congenital Metabolic Diseases, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, University of Santiago de Compostela, CIBERER, MetabERN, Santiago de Compostela, Spain
| | | | - Eduardo López-Laso
- Paediatric Neurology Unit, Department of Paediatrics, University Hospital Reina Sofía, Maimónides Institute of Biomedical Investigation of Cordoba (IMIBIC) and CIBERER, Córdoba, Spain
| | | | | | | | - Salvador Ibáñez-Micó
- Neuropaediatrics Unit, Department of Pediatrics, Virgen de la Arrixaca University Hospital, Murcia, Spain
| | | | | | - Scarlet Witting Enriquez
- Child Neurology Service, Clinical Hospital San Borja Arriarán, University of Chile, Santiago, Chile
| | - Magdalena Ugarte
- Centro de Diagnóstico de Enfermedades Moleculares, Center of Molecular Biology Severo Ochoa (CBMSO), Autonomous University of Madrid, CIBERER, IdiPAZ, Madrid, Spain
| | - Rafael Artuch
- Sant Joan de Déu Research Institute, CIBERER, Barcelona, Spain
| | - Belén Pérez
- Centro de Diagnóstico de Enfermedades Moleculares, Center of Molecular Biology Severo Ochoa (CBMSO), Autonomous University of Madrid, CIBERER, IdiPAZ, Madrid, Spain
| |
Collapse
|
61
|
Brain Metabolic Alterations in Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23073785. [PMID: 35409145 PMCID: PMC8998942 DOI: 10.3390/ijms23073785] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
The brain is one of the most energy-consuming organs in the body. Satisfying such energy demand requires compartmentalized, cell-specific metabolic processes, known to be complementary and intimately coupled. Thus, the brain relies on thoroughly orchestrated energy-obtaining agents, processes and molecular features, such as the neurovascular unit, the astrocyte-neuron metabolic coupling, and the cellular distribution of energy substrate transporters. Importantly, early features of the aging process are determined by the progressive perturbation of certain processes responsible for adequate brain energy supply, resulting in brain hypometabolism. These age-related brain energy alterations are further worsened during the prodromal stages of neurodegenerative diseases, namely Alzheimer's disease (AD), preceding the onset of clinical symptoms, and are anatomically and functionally associated with the loss of cognitive abilities. Here, we focus on concrete neuroenergetic features such as the brain's fueling by glucose and lactate, the transporters and vascular system guaranteeing its supply, and the metabolic interactions between astrocytes and neurons, and on its neurodegenerative-related disruption. We sought to review the principles underlying the metabolic dimension of healthy and AD brains, and suggest that the integration of these concepts in the preventive, diagnostic and treatment strategies for AD is key to improving the precision of these interventions.
Collapse
|
62
|
Study of paediatric patients with the clinical and biochemical phenotype of glucose transporter type 1 deficiency syndrome. NEUROLOGÍA (ENGLISH EDITION) 2022; 37:91-100. [PMID: 35279228 DOI: 10.1016/j.nrleng.2018.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/30/2018] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Glucose transporter type 1 (GLUT1) deficiency syndrome may present a range of phenotypes, including epilepsy, intellectual disability, and movement disorders. The majority of patients present low CSF glucose levels and/or defects in the SLC2A1 gene; however, some patients do not present low CSF glucose or SLC2A1 mutations, and may have other mutations in other genes with compatible phenotypes. AIMS We describe the clinical, biochemical, and genetic characteristics of the disease and perform a univariate analysis of a group of patients with clinical and biochemical phenotype of GLUT1 deficiency syndrome, with or without SLC2A1 mutations. MATERIAL AND METHODS The study included 13 patients meeting clinical and biochemical criteria for GLUT1 deficiency syndrome. SLC2A1 sequencing and multiplex ligation-dependent probe amplification were performed; exome sequencing was performed for patients with negative results. RESULTS Six patients presented the classic phenotype; 2 paroxysmal dyskinesia, 2 complex movement disorders, 2 early-onset absence seizures, and one presented drug-resistant childhood absence epilepsy. Six patients were positive for SLC2A1 mutations; in the other 5, another genetic defect was identified. No significant differences were observed between the 2 groups for age of onset, clinical presentation, microcephaly, intellectual disability, or response to ketogenic diet. Patients with SLC2A1 mutations presented more clinical changes in relation to diet (66.7%, vs 28.6% in the SLC2A1-negative group) and greater persistence of motor symptoms (66% vs 28.6%); these differences were not statistically significant. Significant differences were observed for CSF glucose level (34.5 vs 46mg/dL, P=.04) and CSF/serum glucose ratio (0.4 vs 0.48, P<.05). CONCLUSIONS GLUT1 deficiency syndrome may be caused by mutations to genes other than SLC2A1 in patients with compatible phenotype, low CSF glucose level, and good response to the ketogenic diet.
Collapse
|
63
|
Boyé K, Geraldo LH, Furtado J, Pibouin-Fragner L, Poulet M, Kim D, Nelson B, Xu Y, Jacob L, Maissa N, Agalliu D, Claesson-Welsh L, Ackerman SL, Eichmann A. Endothelial Unc5B controls blood-brain barrier integrity. Nat Commun 2022; 13:1169. [PMID: 35246514 PMCID: PMC8897508 DOI: 10.1038/s41467-022-28785-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 01/18/2022] [Indexed: 12/20/2022] Open
Abstract
Blood-brain barrier (BBB) integrity is critical for proper function of the central nervous system (CNS). Here, we show that the endothelial Unc5B receptor controls BBB integrity by maintaining Wnt/β-catenin signaling. Inducible endothelial-specific deletion of Unc5B in adult mice leads to BBB leak from brain capillaries that convert to a barrier-incompetent state with reduced Claudin-5 and increased PLVAP expression. Loss of Unc5B decreases BBB Wnt/β-catenin signaling, and β-catenin overexpression rescues Unc5B mutant BBB defects. Mechanistically, the Unc5B ligand Netrin-1 enhances Unc5B interaction with the Wnt co-receptor LRP6, induces its phosphorylation and activates Wnt/β-catenin downstream signaling. Intravenous delivery of antibodies blocking Netrin-1 binding to Unc5B causes a transient BBB breakdown and disruption of Wnt signaling, followed by neurovascular barrier resealing. These data identify Netrin-1-Unc5B signaling as a ligand-receptor pathway that regulates BBB integrity, with implications for CNS diseases.
Collapse
Affiliation(s)
- Kevin Boyé
- Cardiovascular Research Center, Department of Internal Medicine, 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
- Université de Paris, INSERM, PARCC, F-75015, Paris, France
| | - Jessica Furtado
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | - Mathilde Poulet
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Université de Paris, INSERM, PARCC, F-75015, Paris, France
| | - Doyeun Kim
- OliX pharmaceuticals, Suwon, Republic of Korea
| | - Bryce Nelson
- Department of Pharmacology, Cancer Biology Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Yunling Xu
- Université de Paris, INSERM, PARCC, F-75015, Paris, France
| | - Laurent Jacob
- Université de Paris, INSERM, PARCC, F-75015, Paris, France
| | - Nawal Maissa
- Université de Paris, INSERM, PARCC, F-75015, Paris, France
| | - Dritan Agalliu
- Departments of Neurology and Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Susan L Ackerman
- Division of Biological Sciences Section of Neurobiology and Department of Cellular and Molecular Medicine, University of California San Diego and Howard Hughes Medical Institute, La Jolla, CA, USA
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
- Université de Paris, INSERM, PARCC, F-75015, Paris, France.
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
64
|
Jiménez Legido M, Cortés Ledesma C, Bernardino Cuesta B, López Marín L, Cantarín Extremera V, Pérez-Cerdá C, Pérez González B, López Martín E, González Gutiérrez-Solana L. Study of paediatric patients with the clinical and biochemical phenotype of glucose transporter type 1 deficiency syndrome. Neurologia 2022; 37:91-100. [PMID: 31047728 DOI: 10.1016/j.nrl.2018.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/16/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Glucose transporter type 1 (GLUT1) deficiency syndrome may present a range of phenotypes, including epilepsy, intellectual disability, and movement disorders. The majority of patients present low CSF glucose levels and/or defects in the SLC2A1 gene; however, some patients do not present low CSF glucose or SLC2A1 mutations, and may have other mutations in other genes with compatible phenotypes. AIMS We describe the clinical, biochemical, and genetic characteristics of the disease and perform a univariate analysis of a group of patients with clinical and biochemical phenotype of GLUT1 deficiency syndrome, with or without SLC2A1 mutations. MATERIAL AND METHODS The study included 13 patients meeting clinical and biochemical criteria for GLUT1 deficiency syndrome. SLC2A1 sequencing and multiplex ligation-dependent probe amplification were performed; exome sequencing was performed for patients with negative results. RESULTS Six patients presented the classic phenotype; 2 paroxysmal dyskinesia, 2 complex movement disorders, 2 early-onset absence seizures, and one presented drug-resistant childhood absence epilepsy. Six patients were positive for SLC2A1 mutations; in the other 5, another genetic defect was identified. No significant differences were observed between the 2 groups for age of onset, clinical presentation, microcephaly, intellectual disability, or response to ketogenic diet. Patients with SLC2A1 mutations presented more clinical changes in relation to diet (66.7% vs. 28.6% in the SLC2A1-negative group) and greater persistence of motor symptoms (66% vs. 28.6%); these differences were not statistically significant. Significant differences were observed for CSF glucose level (34.5 vs. 46mg/dL, P=.04) and CSF/serum glucose ratio (0.4 vs. 0.48, P<.05). CONCLUSIONS GLUT1 deficiency syndrome may be caused by mutations to genes other than SLC2A1 in patients with compatible phenotype, low CSF glucose level, and good response to the ketogenic diet.
Collapse
Affiliation(s)
- M Jiménez Legido
- Sección de Neuropediatría, Hospital Infantil Universitario Niño Jesús, Madrid, España.
| | - C Cortés Ledesma
- Sección de Neuropediatría, Hospital Infantil Universitario Niño Jesús, Madrid, España
| | - B Bernardino Cuesta
- Sección de Neuropediatría, Hospital Infantil Universitario Niño Jesús, Madrid, España
| | - L López Marín
- Sección de Neuropediatría, Hospital Infantil Universitario Niño Jesús, Madrid, España; Grupo Clínico Vinculado a CIBERER (GCV6)
| | - V Cantarín Extremera
- Sección de Neuropediatría, Hospital Infantil Universitario Niño Jesús, Madrid, España; Grupo Clínico Vinculado a CIBERER (GCV6)
| | - C Pérez-Cerdá
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, España
| | - B Pérez González
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), Madrid, España
| | - E López Martín
- Instituto de Investigación de Enfermedades Raras (IIER) & Centro de Investigación Biomédica en Red para Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, España
| | - L González Gutiérrez-Solana
- Sección de Neuropediatría, Hospital Infantil Universitario Niño Jesús, Madrid, España; Grupo Clínico Vinculado a CIBERER (GCV6)
| |
Collapse
|
65
|
Yang R, Chen M, Zheng J, Li X, Zhang X. The Role of Heparin and Glycocalyx in Blood-Brain Barrier Dysfunction. Front Immunol 2022; 12:754141. [PMID: 34992593 PMCID: PMC8724024 DOI: 10.3389/fimmu.2021.754141] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/29/2021] [Indexed: 11/13/2022] Open
Abstract
The blood-brain barrier (BBB) functions as a dynamic boundary that protects the central nervous system from blood and plays an important role in maintaining the homeostasis of the brain. Dysfunction of the BBB is a pathophysiological characteristic of multiple neurologic diseases. Glycocalyx covers the luminal side of vascular endothelial cells(ECs). Damage of glycocalyx leads to disruption of the BBB, while inhibiting glycocalyx degradation maintains BBB integrity. Heparin has been recognized as an anticoagulant and it protects endothelial glycocalyx from destruction. In this review, we summarize the role of glycocalyx in BBB formation and the therapeutic potency of heparin to provide a theoretical basis for the treatment of neurological diseases related to BBB breakdown.
Collapse
Affiliation(s)
- Rui Yang
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mingming Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jiayin Zheng
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xin Li
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaojuan Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
66
|
Xia X, Wang Y, Qin Y, Zhao S, Zheng JC. Exosome: A novel neurotransmission modulator or non-canonical neurotransmitter? Ageing Res Rev 2022; 74:101558. [PMID: 34990846 DOI: 10.1016/j.arr.2021.101558] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/13/2021] [Accepted: 12/30/2021] [Indexed: 02/08/2023]
Abstract
Neurotransmission is the electrical impulse-triggered propagation of signals between neurons or between neurons and other cell types such as skeletal muscle cells. Recent studies point out the involvement of exosomes, a type of small bilipid layer-enclosed extracellular vesicles, in regulating neurotransmission. Through horizontally transferring proteins, lipids, and nucleic acids, exosomes can modulate synaptic activities rapidly by controlling neurotransmitter release or progressively by regulating neural plasticity including synapse formation, neurite growth & removal, and axon guidance & elongation. In this review, we summarize the similarities and differences between exosomes and synaptic vesicles in their biogenesis, contents, and release. We also highlight the recent progress made in demonstrating the biological roles of exosome in regulating neurotransmission, and propose a modified model of neurotransmission, in which exosomes act as novel neurotransmitters. Lastly, we provide a comprehensive discussion of the enlightenment of the current knowledge on neurotransmission to the future directions of exosome research.
Collapse
|
67
|
Genetic paroxysmal neurological disorders featuring episodic ataxia and epilepsy. Eur J Med Genet 2022; 65:104450. [DOI: 10.1016/j.ejmg.2022.104450] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 01/25/2023]
|
68
|
Fame RM, Lehtinen MK. Mitochondria in Early Forebrain Development: From Neurulation to Mid-Corticogenesis. Front Cell Dev Biol 2021; 9:780207. [PMID: 34888312 PMCID: PMC8650308 DOI: 10.3389/fcell.2021.780207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/10/2021] [Indexed: 01/07/2023] Open
Abstract
Function of the mature central nervous system (CNS) requires a substantial proportion of the body’s energy consumption. During development, the CNS anlage must maintain its structure and perform stage-specific functions as it proceeds through discrete developmental stages. While key extrinsic signals and internal transcriptional controls over these processes are well appreciated, metabolic and mitochondrial states are also critical to appropriate forebrain development. Specifically, metabolic state, mitochondrial function, and mitochondrial dynamics/localization play critical roles in neurulation and CNS progenitor specification, progenitor proliferation and survival, neurogenesis, neural migration, and neurite outgrowth and synaptogenesis. With the goal of integrating neurodevelopmental biologists and mitochondrial specialists, this review synthesizes data from disparate models and processes to compile and highlight key roles of mitochondria in the early development of the CNS with specific focus on forebrain development and corticogenesis.
Collapse
Affiliation(s)
- Ryann M Fame
- Department of Pathology, Boston Children's Hospital, Boston, MA, United States
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
69
|
Hu Q, Shen Y, Su T, Liu Y, Xu S. Clinical and Genetic Characteristics of Chinese Children With GLUT1 Deficiency Syndrome: Case Report and Literature Review. Front Genet 2021; 12:734481. [PMID: 34880899 PMCID: PMC8645975 DOI: 10.3389/fgene.2021.734481] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/18/2021] [Indexed: 11/24/2022] Open
Abstract
Objective: GLUT1 deficiency syndrome (GLUT1-DS) is a rare, treatable neurometabolic disorder. However, its diagnosis may be challenging due to the various and evolving phenotypes. Here we report the first Chinese familial cases with genetically confirmed GLUT1-DS and analyze the characteristics of Chinese children with GLUT1-DS from clinical, laboratory, and genetic aspects. Methods: We reported a Chinese family with three members affected with GLUT1-DS and searched for relevant articles up to September 2020 from PubMed, WOS, CNKI, and WanFang databases. A total of 30 Chinese patients diagnosed with GLUT1-DS (three newly identified patients in one family and 27 previously reported ones) were included and analyzed in this study. Results: The median age of onset of the 30 patients (male: 18, female: 12) was 8.5 months (range, 33 days to 10 years). Epileptic seizures were found in 25 patients, most with generalized tonic–clonic and focal ones. Movement disorders were found in 20 patients—frequently with ataxia and dystonia, developmental delay in 25 patients, and microcephaly only in six patients. The cerebrospinal fluid (CSF) analysis showed decreased CSF glucose (median: 1.63 mmol/L, range: 1.1–2.6 mmol/L) and glucose ratio of CSF to blood (median: 0.340; range: 0.215–0.484). The genetic testing performed in 28 patients revealed 27 cases with pathogenic variations of the SLC2A1 gene, including 10 missense, nine frameshift, three nonsense, three large fragment deletions, and two splice-site mutations. Most patients had a good response to the treatment of ketogenic diet or regular diet with increased frequency. Although three patients in this Chinese family carried the same pathogenic mutation c.73C > T (p.Q25X) in the SLC2A1 gene, their symptoms and responses to treatment were not exactly the same. Conclusion: The clinical manifestations of GLUT1-DS are heterogeneous, even among family members sharing the same mutation. For children with unexplained epileptic seizures, developmental delay, and complex movement disorders, detection of low CSF glucose or SLC2A1 gene mutations is helpful for the diagnosis of GLUT1-DS. Early initiation of ketogenic diet treatment significantly improves the symptoms and prognosis of GLUT1-DS.
Collapse
Affiliation(s)
- Qingqing Hu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuechi Shen
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tangfeng Su
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sanqing Xu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
70
|
Mir A, Almudhry M, Alghamdi F, Albaradie R, Ibrahim M, Aldurayhim F, Alhedaithy A, Alamr M, Bawazir M, Mohammad S, Abdelhay S, Bashir S, Housawi Y. SLC gene mutations and pediatric neurological disorders: diverse clinical phenotypes in a Saudi Arabian population. Hum Genet 2021; 141:81-99. [PMID: 34797406 DOI: 10.1007/s00439-021-02404-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
The uptake and efflux of solutes across a plasma membrane is controlled by transporters. There are two main superfamilies of transporters, adenosine 5'-triphosphate (ATP) binding cassettes (ABCs) and solute carriers (SLCs). In the brain, SLC transporters are involved in transporting various solutes across the blood-brain barrier, blood-cerebrospinal fluid barrier, astrocytes, neurons, and other brain cell types including oligodendrocytes and microglial cells. SLCs play an important role in maintaining normal brain function. Hence, mutations in the genes that encode SLC transporters can cause a variety of neurological disorders. We identified the following SLC gene variants in 25 patients in our cohort: SLC1A2, SLC2A1, SLC5A1, SLC6A3, SLC6A5, SLC6A8, SLC9A6, SLC9A9, SLC12A6, SLC13A5, SLC16A1, SLC17A5, SLC19A3, SLC25A12, SLC25A15, SLC27A4, SLC45A1, SLC46A1, and SLC52A3. Eight patients harbored pathogenic or likely pathogenic mutations (SLC5A1, SLC9A6, SLC12A6, SLC16A1, SLC19A3, and SLC52A3), and 12 patients were found to have variants of unknown clinical significance (VOUS); these variants occurred in 11 genes (SLC1A2, SLC2A1, SLC6A3, SLC6A5, SLC6A8, SLC9A6, SLC9A9, SLC13A5, SLC25A12, SLC27A4, and SLC45A1). Five patients were excluded as they were carriers. In the remaining 20 patients with SLC gene variants, we identified 16 possible distinct neurological disorders. Based on the clinical presentation, we categorized them into genes causing intellectual delay (ID) or autism spectrum disorder (ASD), those causing epilepsy, those causing vitamin-related disorders, and those causing other neurological diseases. Several variants were detected that indicated possible personalized therapies: SLC2A1 led to dystonia or epilepsy, which can be treated with a ketogenic diet; SLC6A3 led to infantile parkinsonism-dystonia 1, which can be treated with levodopa; SLC6A5 led to hyperekplexia 3, for which unnecessary treatment with antiepileptic drugs should be avoided; SLC6A8 led to creatine deficiency syndrome type 1, which can be treated with creatine monohydrate; SLC16A1 led to monocarboxylate transporter 1 deficiency, which causes seizures that should not be treated with a ketogenic diet; SLC19A3 led to biotin-thiamine-responsive basal ganglia disease, which can be treated with biotin and thiamine; and SLC52A3 led to Brown-Vialetto-Van-Laere syndrome 1, which can be treated with riboflavin. The present study examines the prevalence of SLC gene mutations in our cohort of children with epilepsy and other neurological disorders. It highlights the diverse phenotypes associated with mutations in this large family of SLC transporter proteins, and an opportunity for personalized genomics and personalized therapeutics.
Collapse
Affiliation(s)
- Ali Mir
- Department of Pediatric Neurology, Neuroscience Center, King Fahad Specialist Hospital, Ammar Bin Thabit Street, Dammam, 31444, Kingdom of Saudi Arabia.
| | - Montaha Almudhry
- Department of Pediatric Neurology, Neuroscience Center, King Fahad Specialist Hospital, Ammar Bin Thabit Street, Dammam, 31444, Kingdom of Saudi Arabia
| | - Fouad Alghamdi
- Department of Pediatric Neurology, Neuroscience Center, King Fahad Specialist Hospital, Ammar Bin Thabit Street, Dammam, 31444, Kingdom of Saudi Arabia
| | - Raidah Albaradie
- Department of Pediatric Neurology, Neuroscience Center, King Fahad Specialist Hospital, Ammar Bin Thabit Street, Dammam, 31444, Kingdom of Saudi Arabia
| | - Mona Ibrahim
- Department of Pediatric Neurology, Neuroscience Center, King Fahad Specialist Hospital, Ammar Bin Thabit Street, Dammam, 31444, Kingdom of Saudi Arabia
| | - Fatimah Aldurayhim
- Department of Pediatric Neurology, Neuroscience Center, King Fahad Specialist Hospital, Ammar Bin Thabit Street, Dammam, 31444, Kingdom of Saudi Arabia
| | - Abdullah Alhedaithy
- Department of Pediatric Neurology, Neuroscience Center, King Fahad Specialist Hospital, Ammar Bin Thabit Street, Dammam, 31444, Kingdom of Saudi Arabia
| | - Mushari Alamr
- Genetic and Metabolic Department, King Fahad Specialist Hospital, Dammam, Kingdom of Saudi Arabia
| | - Maryam Bawazir
- Genetic and Metabolic Department, King Fahad Specialist Hospital, Dammam, Kingdom of Saudi Arabia
| | - Sahar Mohammad
- Department of Pediatric, King Fahad Specialist Hospital, Dammam, Kingdom of Saudi Arabia
| | - Salma Abdelhay
- Department of Pediatric, King Fahad Specialist Hospital, Dammam, Kingdom of Saudi Arabia
| | - Shahid Bashir
- Department of Pediatric Neurology, Neuroscience Center, King Fahad Specialist Hospital, Ammar Bin Thabit Street, Dammam, 31444, Kingdom of Saudi Arabia
| | - Yousef Housawi
- Genetic and Metabolic Department, King Fahad Specialist Hospital, Dammam, Kingdom of Saudi Arabia
| |
Collapse
|
71
|
Marini C, Giardino M. Novel treatments in epilepsy guided by genetic diagnosis. Br J Clin Pharmacol 2021; 88:2539-2551. [PMID: 34778987 DOI: 10.1111/bcp.15139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/22/2021] [Accepted: 11/04/2021] [Indexed: 12/21/2022] Open
Abstract
In recent years, precision medicine has emerged as a new paradigm for improved and more individualized patient care. Its key objective is to provide the right treatment, to the right patient at the right time, by basing medical decisions on individual characteristics, including specific genetic biomarkers. In order to realize this objective researchers and physicians must first identify the underlying genetic cause; over the last 10 years, advances in genetics have made this possible for several monogenic epilepsies. Through next generation techniques, a precise genetic aetiology is attainable in 30-50% of genetic epilepsies beginning in the paediatric age. While committed in such search for novel genes carrying disease-causing variants, progress in the study of experimental models of epilepsy has also provided a better understanding of the mechanisms underlying the condition. Such advances are already being translated into improving care, management and treatment of some patients. Identification of a precise genetic aetiology can already direct physicians to prescribe treatments correcting specific metabolic defects, avoid antiseizure medicines that might aggravate functional consequences of the disease-causing variant or select the drugs that counteract the underlying, genetically determined, functional disturbance. Personalized, tailored treatments should not just focus on how to stop seizures but possibly prevent their onset and cure the disorder, often consisting of seizures and its comorbidities including cognitive, motor and behaviour deficiencies. This review discusses the therapeutic implications following a specific genetic diagnosis and the correlation between genetic findings, pathophysiological mechanisms and tailored seizure treatment, emphasizing the impact on current clinical practice.
Collapse
Affiliation(s)
- Carla Marini
- Child Neurology and Psychiatric Unit, Pediatric Hospital G. Salesi, United Hospitals of Ancona, Ancona, Italy
| | - Maria Giardino
- Child Neurology and Psychiatric Unit, Pediatric Hospital G. Salesi, United Hospitals of Ancona, Ancona, Italy
| |
Collapse
|
72
|
Inherited Developmental and Epileptic Encephalopathies. Neurol Int 2021; 13:555-568. [PMID: 34842787 PMCID: PMC8628919 DOI: 10.3390/neurolint13040055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 01/13/2023] Open
Abstract
Epileptic encephalopathies often have a genetic etiology. The epileptic activity itself exerts a direct detrimental effect on neurodevelopment, which may add to the cognitive impairment induced by the underlying mutation (“developmental and epileptic encephalopathy”). The focus of this review is on inherited syndromes. The phenotypes of genetic disorders affecting ion channels, metabolic signalling, membrane trafficking and exocytosis, cell adhesion, cell growth and proliferation are discussed. Red flags suggesting family of genes or even specific genes are highlighted. The knowledge of the phenotypical spectrum can indeed prompt the clinician to suspect specific etiologies, expediting the diagnosis.
Collapse
|
73
|
Archie SR, Al Shoyaib A, Cucullo L. Blood-Brain Barrier Dysfunction in CNS Disorders and Putative Therapeutic Targets: An Overview. Pharmaceutics 2021; 13:pharmaceutics13111779. [PMID: 34834200 PMCID: PMC8622070 DOI: 10.3390/pharmaceutics13111779] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 01/22/2023] Open
Abstract
The blood-brain barrier (BBB) is a fundamental component of the central nervous system (CNS). Its functional and structural integrity is vital to maintain the homeostasis of the brain microenvironment by controlling the passage of substances and regulating the trafficking of immune cells between the blood and the brain. The BBB is primarily composed of highly specialized microvascular endothelial cells. These cells’ special features and physiological properties are acquired and maintained through the concerted effort of hemodynamic and cellular cues from the surrounding environment. This complex multicellular system, comprising endothelial cells, astrocytes, pericytes, and neurons, is known as the neurovascular unit (NVU). The BBB strictly controls the transport of nutrients and metabolites into brain parenchyma through a tightly regulated transport system while limiting the access of potentially harmful substances via efflux transcytosis and metabolic mechanisms. Not surprisingly, a disruption of the BBB has been associated with the onset and/or progression of major neurological disorders. Although the association between disease and BBB disruption is clear, its nature is not always evident, specifically with regard to whether an impaired BBB function results from the pathological condition or whether the BBB damage is the primary pathogenic factor prodromal to the onset of the disease. In either case, repairing the barrier could be a viable option for treating and/or reducing the effects of CNS disorders. In this review, we describe the fundamental structure and function of the BBB in both healthy and altered/diseased conditions. Additionally, we provide an overview of the potential therapeutic targets that could be leveraged to restore the integrity of the BBB concomitant to the treatment of these brain disorders.
Collapse
Affiliation(s)
- Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (S.R.A.); (A.A.S.)
| | - Abdullah Al Shoyaib
- Department of Pharmaceutical Sciences, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; (S.R.A.); (A.A.S.)
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
- Correspondence: ; Tel.: +1-248-370-3884; Fax: +1-248-370-4060
| |
Collapse
|
74
|
Maiorana A, Caviglia S, Greco B, Alfieri P, Cumbo F, Campana C, Bernabei SM, Cusmai R, Mosca A, Dionisi-Vici C. Ketogenic diet as elective treatment in patients with drug-unresponsive hyperinsulinemic hypoglycemia caused by glucokinase mutations. Orphanet J Rare Dis 2021; 16:424. [PMID: 34635134 PMCID: PMC8507241 DOI: 10.1186/s13023-021-02045-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/19/2021] [Indexed: 11/10/2022] Open
Abstract
Background Hyperinsulinemic hypoglycemia (HI) is the most frequent cause of recurrent hypoglycemia in children. Despite diagnostic and therapeutic advances, it remains an important cause of morbidity, leading to neurological complications, such as psychomotor retardation and epilepsy. Patients with diffuse drug-unresponsive HI manifest neurological impairment and neurobehavioral problems, even though surgically treated with a near-total pancreatectomy. Based on the analogies between HI and GLUT1 deficiency, both presenting with neuroglycopenia and lack of alternative cerebral energy sources, we administered a ketogenic diet (KD) in three drug-unresponsive GCK-HI patients with the aim of preserving neurodevelopment and avoiding the need of a near-total pancreatectomy. They presented recurrent symptomatic hypoglycemia, intellectual disability and refractory epilepsy. Patients were treated with classical KD for 79, 27 and 18 months, respectively. Results All patients became asymptomatic in a few days and showed an important improvement of the alert state. Epilepsy disappeared and no appearance of novel hypoglycemic lesions was detected with a brain MRI. Cognitive and adaptive abilities rapidly improved and normalized. IQ rose significantly from 81 to 111 (p = 0.04) in patient 1, from 82 vs 95 (p = 0.04) in patient 2, from 60 to 90 (p = 0.04) in patient 3. Conclusions We demonstrated the safety and efficacy of KD in the treatment of drug-unresponsive GCK-HI at a short and long-term. The neuroprotective effects of KD determined the recovery from epilepsy and intellectual disabilities and averted the need of a near-total pancreatectomy. All patients and their families reported an improvement of physical and psychosocial well-being, with a substantial improvement of their quality of life. These results might change the course and the quality of life of these patients and their families, having a relevant impact on human lives. Therefore, KD might be considered the elective treatment in unresponsive forms of GCK-HI. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-02045-3.
Collapse
Affiliation(s)
- Arianna Maiorana
- Division of Metabolism, Department of Pediatric Subspecialties, Ospedale Pediatrico Bambino Gesù, IRCSS, Piazza S. Onofrio 4, 00165, Rome, Italy.
| | - Stefania Caviglia
- Psychology Clinic Unit, Department of Neuroscience, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Benedetta Greco
- Psychology Clinic Unit, Department of Neuroscience, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Paolo Alfieri
- Child and Adolescent Psychiatric Unit, Department of Neuroscience, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Francesca Cumbo
- Child and Adolescent Psychiatric Unit, Department of Neuroscience, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Carmen Campana
- Division of Metabolism, Department of Pediatric Subspecialties, Ospedale Pediatrico Bambino Gesù, IRCSS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Silvia Maria Bernabei
- Division of Artificial Nutrition, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Raffaella Cusmai
- Neurology Unit, Department of Neuroscience, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Antonella Mosca
- Department of Hepatology, Gastroenterology and Nutrition, Ospedale Pediatrico Bambino Gesù, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Carlo Dionisi-Vici
- Division of Metabolism, Department of Pediatric Subspecialties, Ospedale Pediatrico Bambino Gesù, IRCSS, Piazza S. Onofrio 4, 00165, Rome, Italy
| |
Collapse
|
75
|
Montana V, Flint D, Waagepetersen HS, Schousboe A, Parpura V. Two Metabolic Fuels, Glucose and Lactate, Differentially Modulate Exocytotic Glutamate Release from Cultured Astrocytes. Neurochem Res 2021; 46:2551-2579. [PMID: 34057673 PMCID: PMC9015689 DOI: 10.1007/s11064-021-03340-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022]
Abstract
Astrocytes have a prominent role in metabolic homeostasis of the brain and can signal to adjacent neurons by releasing glutamate via a process of regulated exocytosis. Astrocytes synthesize glutamate de novo owing to the pyruvate entry to the citric/tricarboxylic acid cycle via pyruvate carboxylase, an astrocyte specific enzyme. Pyruvate can be sourced from two metabolic fuels, glucose and lactate. Thus, we investigated the role of these energy/carbon sources in exocytotic glutamate release from astrocytes. Purified astrocyte cultures were acutely incubated (1 h) in glucose and/or lactate-containing media. Astrocytes were mechanically stimulated, a procedure known to increase intracellular Ca2+ levels and cause exocytotic glutamate release, the dynamics of which were monitored using single cell fluorescence microscopy. Our data indicate that glucose, either taken-up from the extracellular space or mobilized from the intracellular glycogen storage, sustained glutamate release, while the availability of lactate significantly reduced the release of glutamate from astrocytes. Based on further pharmacological manipulation during imaging along with tandem mass spectrometry (proteomics) analysis, lactate alone, but not in the hybrid fuel, caused metabolic changes consistent with an increased synthesis of fatty acids. Proteomics analysis further unveiled complex changes in protein profiles, which were condition-dependent and generally included changes in levels of cytoskeletal proteins, proteins of secretory organelle/vesicle traffic and recycling at the plasma membrane in aglycemic, lactate or hybrid-fueled astrocytes. These findings support the notion that the availability of energy sources and metabolic milieu play a significant role in gliotransmission.
Collapse
Affiliation(s)
- Vedrana Montana
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Daniel Flint
- Luxumbra Strategic Research, LLC, Arlington, VA, USA
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
76
|
Tang M, Monani UR. Glut1 deficiency syndrome: New and emerging insights into a prototypical brain energy failure disorder. Neurosci Insights 2021; 16:26331055211011507. [PMID: 34589708 PMCID: PMC8474335 DOI: 10.1177/26331055211011507] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/01/2021] [Indexed: 11/16/2022] Open
Abstract
Considering its small size relative to the rest of the body, the
mammalian brain has a disproportionately high energy requirement. This
energy is supplied to the brain mainly in the form of glucose through
the principal cerebral glucose transporter, Glut1. Inactivation of
even a single copy of the Glut1 gene, SLC2A1, has
dire consequences for the brain, starving cerebral neurons of energy
and triggering the debilitating neurodevelopmental disorder, Glut1
deficiency syndrome (Glut1 DS). Considering the monogenic nature of
Glut1 DS, the disease serves as an excellent paradigm to study the
larger family of brain energy failure syndromes. Here we review how
studies of Glut1 DS are proving instructive to the brain’s energy
needs, focusing first on the requirements, both spatial and temporal
of the transporter, second, on proposed mechanisms linking low Glut1
to brain dysfunction and, finally on efforts to treat the disease and
thus restore nutritional support to the brain. These studies promise
not only to inform mechanisms and treatments for the relatively rare
Glut1 DS but also the myriad other conditions involving the Glut1
protein.
Collapse
Affiliation(s)
- Maoxue Tang
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, NY, USA
| | - Umrao R Monani
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, NY, USA.,Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
77
|
Jacobson RI. Discovery of Glut 1 Deficiency Syndrome: Cerebrospinal Fluid Inspiration and Serendipity. Pediatr Neurol 2021; 122:65-67. [PMID: 34303586 DOI: 10.1016/j.pediatrneurol.2021.05.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/23/2021] [Indexed: 10/21/2022]
Affiliation(s)
- Ronald I Jacobson
- Chief of Pediatric Neurology, Boston Children's Health Physicians, Maria Fareri Children's Hospital at Westchester Medical Center, Clinical Associate Professor Neurology and Pediatrics, New York Medical College, Valhalla, New York.
| |
Collapse
|
78
|
GLUT1 Deficiency Syndrome-Early Treatment Maintains Cognitive Development? (Literature Review and Case Report). Genes (Basel) 2021; 12:genes12091379. [PMID: 34573360 PMCID: PMC8472230 DOI: 10.3390/genes12091379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/22/2021] [Accepted: 08/30/2021] [Indexed: 01/13/2023] Open
Abstract
Glucose transporter type 1 (GLUT1) is the most important energy carrier of the brain across the blood-brain barrier, and a genetic defect of GLUT1 is known as GLUT1 deficiency syndrome (GLUT1DS). It is characterized by early infantile seizures, developmental delay, microcephaly, ataxia, and various paroxysmal neurological phenomena. In most cases, GLUT1DS is caused by heterozygous single-nucleotide variants (SNVs) in the SLC2A1 gene that provoke complete or severe impairment of the functionality and/or expression of GLUT1 in the brain. Despite the rarity of these diseases, GLUT1DS is of high clinical interest since a very effective therapy, the ketogenic diet, can improve or reverse symptoms, especially if it is started as early as possible. We present a clinical phenotype, biochemical analysis, electroencephalographic and neuropsychological features of an 11-month-old boy with myoclonic seizures, hypogammaglobulinemia, and mildly impaired gross motor development. Using sequence analysis and deletion/duplication testing, deletion of an entire coding sequence in the SLC2A1 gene was detected. Early introduction of a modified Atkins diet maintained a seizure-free period without antiseizure medications and normal cognitive development in the follow-up period. Our report summarizes the clinical features of GLUT1 syndromes and discusses the importance of early identification and molecular confirmation of GLUT1DS as a treatable metabolic disorder.
Collapse
|
79
|
Sommonte F, Arduino I, Racaniello GF, Lopalco A, Lopedota AA, Denora N. The Complexity of the Blood-Brain Barrier and the Concept of Age-Related Brain Targeting: Challenges and Potential of Novel Solid Lipid-Based Formulations. J Pharm Sci 2021; 111:577-592. [PMID: 34469749 DOI: 10.1016/j.xphs.2021.08.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022]
Abstract
Diseases that affect the Central Nervous System (CNS) are one of the most exciting challenges of recent years, as they are ubiquitous and affect all ages. Although these disorders show different etiologies, all treatments share the same difficulty represented by the Blood-Brain Barrier (BBB). This barrier acts as a protective system of the delicate cerebral microenvironment, isolating it and making extremely arduous delivering drugs to the brain. To overtake the obstacles provided by the BBB it is essential to explore the changes that affect it, to understand how to exploit these findings in the study and design of innovative brain targeted formulations. Interestingly, the concept of age-related targeting could prove to be a winning choice, as it allows to consider the type of treatment according to the different needs and peculiarities depending on the disease and the age of onset. In this review was considered the prospective contribution of lipid-based formulations, namely Solid Lipid Nanoparticles (SLNs) and Nanostructured Lipid Carriers (NLCs), which have been highlighted as able to overcome some limitations of other innovative approaches, thus representing a promising strategy for the non-invasive specific treatment of CNS-related diseases.
Collapse
Affiliation(s)
- Federica Sommonte
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", 4 Orabona St., 70125, Bari, Italy
| | - Ilaria Arduino
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", 4 Orabona St., 70125, Bari, Italy
| | | | - Antonio Lopalco
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", 4 Orabona St., 70125, Bari, Italy
| | - Angela Assunta Lopedota
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", 4 Orabona St., 70125, Bari, Italy
| | - Nunzio Denora
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro", 4 Orabona St., 70125, Bari, Italy.
| |
Collapse
|
80
|
Zaganas I, Vorgia P, Spilioti M, Mathioudakis L, Raissaki M, Ilia S, Giorgi M, Skoula I, Chinitrakis G, Michaelidou K, Paraskevoulakos E, Grafakou O, Kariniotaki C, Psyllou T, Zafeiris S, Tzardi M, Briassoulis G, Dinopoulos A, Mitsias P, Evangeliou A. Genetic cause of epilepsy in a Greek cohort of children and young adults with heterogeneous epilepsy syndromes. Epilepsy Behav Rep 2021; 16:100477. [PMID: 34568804 PMCID: PMC8449081 DOI: 10.1016/j.ebr.2021.100477] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/01/2022] Open
Abstract
We describe a cohort of 10 unrelated Greek patients (4 females, 6 males; median age 6.5 years, range 2-18 years) with heterogeneous epilepsy syndromes with a genetic basis. In these patients, causative genetic variants, including two novel ones, were identified in 9 known epilepsy-related genes through whole exome sequencing. A patient with glycine encephalopathy was a compound heterozygote for the p.Arg222Cys and the p.Ser77Leu AMT variant. A patient affected with Lafora disease carried the homozygous p.Arg171His EPM2A variant. A de novo heterozygous variant in the GABRG2 gene (p.Pro282Thr) was found in one patient and a pathogenic variant in the GRIN2B gene (p.Gly820Val) in another patient. Infantile-onset lactic acidosis with seizures was associated with the p.Arg446Ter PDHX gene variant in one patient. In two additional epilepsy patients, the p.Ala1662Val and the novel non-sense p.Phe1330Ter SCN1A gene variants were found. Finally, in 3 patients we observed a novel heterozygous missense variant in SCN2A (p.Ala1874Thr), a heterozygous splice site variant in SLC2A1 (c.517-2A>G), as a cause of Glut1 deficiency syndrome, and a pathogenic variant in STXBP1 (p.Arg292Leu), respectively. In half of our cases (patients with variants in the GRIN2B, SCN1A, SCN2A and SLC2A1 genes), a genetic cause with potential management implications was identified.
Collapse
Affiliation(s)
- Ioannis Zaganas
- Neurogenetics Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
- Neurology Department, University Hospital of Heraklion, Crete, Greece
| | - Pelagia Vorgia
- Neurogenetics Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Martha Spilioti
- AHEPA General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lambros Mathioudakis
- Neurogenetics Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Maria Raissaki
- Department of Radiology, University Hospital of Heraklion, Crete, Greece
| | - Stavroula Ilia
- Pediatric Intensive Care Unit, University Hospital of Heraklion, Crete, Greece
| | | | - Irene Skoula
- Neurogenetics Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | | | - Kleita Michaelidou
- Neurogenetics Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | | | - Olga Grafakou
- Pediatric Department, Venizelion General Hospital, Heraklio, Crete, Greece
| | - Chariklia Kariniotaki
- Neurogenetics Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Thekla Psyllou
- Neurogenetics Laboratory, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Spiros Zafeiris
- Neurology Department, University Hospital of Heraklion, Crete, Greece
| | - Maria Tzardi
- Pathology Department, Medical School, University of Crete, Greece
| | - George Briassoulis
- Pediatric Intensive Care Unit, University Hospital of Heraklion, Crete, Greece
| | | | - Panayiotis Mitsias
- Neurology Department, University Hospital of Heraklion, Crete, Greece
- Department of Neurology, Henry Ford Hospital/Wayne State University, Detroit, MI, USA
| | - Athanasios Evangeliou
- Papageorgiou General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
81
|
Rutland CS, Cockcroft JM, Lothion-Roy J, Harris AE, Jeyapalan JN, Simpson S, Alibhai A, Bailey C, Ballard-Reisch AC, Rizvanov AA, Dunning MD, de Brot S, Mongan NP. Immunohistochemical Characterisation of GLUT1, MMP3 and NRF2 in Osteosarcoma. Front Vet Sci 2021; 8:704598. [PMID: 34414229 PMCID: PMC8369506 DOI: 10.3389/fvets.2021.704598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma (OSA) is an aggressive bone malignancy. Unlike many other malignancies, OSA outcomes have not improved in recent decades. One challenge to the development of better diagnostic and therapeutic methods for OSA has been the lack of well characterized experimental model systems. Spontaneous OSA in dogs provides a good model for the disease seen in people and also remains an important veterinary clinical challenge. We recently used RNA sequencing and qRT-PCR to provide a detailed molecular characterization of OSA relative to non-malignant bone in dogs. We identified differential mRNA expression of the solute carrier family 2 member 1 (SLC2A1/GLUT1), matrix metallopeptidase 3 (MMP3) and nuclear factor erythroid 2–related factor 2 (NFE2L2/NRF2) genes in canine OSA tissue in comparison to paired non-tumor tissue. Our present work characterizes protein expression of GLUT1, MMP3 and NRF2 using immunohistochemistry. As these proteins affect key processes such as Wnt activation, heme biosynthesis, glucose transport, understanding their expression and the enriched pathways and gene ontologies enables us to further understand the potential molecular pathways and mechanisms involved in OSA. This study further supports spontaneous OSA in dogs as a model system to inform the development of new methods to diagnose and treat OSA in both dogs and people.
Collapse
Affiliation(s)
- Catrin S Rutland
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - James M Cockcroft
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Jennifer Lothion-Roy
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom.,Faculty of Medicine and Health Science, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Anna E Harris
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom.,Faculty of Medicine and Health Science, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Jennie N Jeyapalan
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom.,Faculty of Medicine and Health Science, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Siobhan Simpson
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Aziza Alibhai
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Clara Bailey
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Albert A Rizvanov
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom.,Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Mark D Dunning
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom.,Willows Veterinary Centre and Referral Service, Solihull, United Kingdom
| | - Simone de Brot
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom.,COMPATH, Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Nigel P Mongan
- School of Veterinary Medicine and Science, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, United Kingdom.,Faculty of Medicine and Health Science, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom.,Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
82
|
Wang H, Qi W, Zou C, Xie Z, Zhang M, Naito MG, Mifflin L, Liu Z, Najafov A, Pan H, Shan B, Li Y, Zhu ZJ, Yuan J. NEK1-mediated retromer trafficking promotes blood-brain barrier integrity by regulating glucose metabolism and RIPK1 activation. Nat Commun 2021; 12:4826. [PMID: 34376696 PMCID: PMC8355301 DOI: 10.1038/s41467-021-25157-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/24/2021] [Indexed: 12/14/2022] Open
Abstract
Loss-of-function mutations in NEK1 gene, which encodes a serine/threonine kinase, are involved in human developmental disorders and ALS. Here we show that NEK1 regulates retromer-mediated endosomal trafficking by phosphorylating VPS26B. NEK1 deficiency disrupts endosomal trafficking of plasma membrane proteins and cerebral proteome homeostasis to promote mitochondrial and lysosomal dysfunction and aggregation of α-synuclein. The metabolic and proteomic defects of NEK1 deficiency disrupts the integrity of blood-brain barrier (BBB) by promoting lysosomal degradation of A20, a key modulator of RIPK1, thus sensitizing cerebrovascular endothelial cells to RIPK1-dependent apoptosis and necroptosis. Genetic inactivation of RIPK1 or metabolic rescue with ketogenic diet can prevent postnatal lethality and BBB damage in NEK1 deficient mice. Inhibition of RIPK1 reduces neuroinflammation and aggregation of α-synuclein in the brains of NEK1 deficient mice. Our study identifies a molecular mechanism by which retromer trafficking and metabolism regulates cerebrovascular integrity, cerebral proteome homeostasis and RIPK1-mediated neuroinflammation.
Collapse
Affiliation(s)
- Huibing Wang
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Weiwei Qi
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Chengyu Zou
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Zhangdan Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Mengmeng Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | | | - Lauren Mifflin
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Zhen Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ayaz Najafov
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Heling Pan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Bing Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ying Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
83
|
New avenues in molecular genetics for the diagnosis and application of therapeutics to the epilepsies. Epilepsy Behav 2021; 121:106428. [PMID: 31400936 DOI: 10.1016/j.yebeh.2019.07.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/14/2019] [Accepted: 07/06/2019] [Indexed: 11/22/2022]
Abstract
Genetic epidemiology studies have shown that most epilepsies involve some genetic cause. In addition, twin studies have helped strengthen the hypothesis that in most patients with epilepsy, a complex inheritance is involved. More recently, with the development of high-density single-nucleotide polymorphism (SNP) microarrays and next-generation sequencing (NGS) technologies, the discovery of genes related to the epilepsies has accelerated tremendously. Especially, the use of whole exome sequencing (WES) has had a considerable impact on the identification of rare genetic variants with large effect sizes, including inherited or de novo mutations in severe forms of childhood epilepsies. The identification of pathogenic variants in patients with these childhood epilepsies provides many benefits for patients and families, such as the confirmation of the genetic nature of the diseases. This process will allow for better genetic counseling, more accurate therapy decisions, and a significant positive emotional impact. However, to study the genetic component of the more common forms of epilepsy, the use of high-density SNP arrays in genome-wide association studies (GWAS) seems to be the strategy of choice. As such, researchers can identify loci containing genetic variants associated with the common forms of epilepsy. The knowledge generated over the past two decades about the effects of the mutations that cause the monogenic epilepsy is tremendous; however, the scientific community is just starting to apply this information in order to generate better target treatments.
Collapse
|
84
|
Bozkurt T, Alanay Y, Isik U, Sezerman U. Re-analysis of whole-exome sequencing data reveals a novel splicing variant in the SLC2A1 in a patient with GLUT1 Deficiency Syndrome 1 accompanied by hemangioma: a case report. BMC Med Genomics 2021; 14:197. [PMID: 34332575 PMCID: PMC8325841 DOI: 10.1186/s12920-021-01045-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 07/22/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND GLUT1 Deficiency Syndrome 1 (GLUT1DS1) is a neurological disorder caused by either heterozygous or homozygous mutations in the Solute Carrier Family 2, Member 1 (SLC2A1) gene. SLC2A1 encodes Glucose transporter type 1 (GLUT1) protein, which is the primary glucose transporter at the blood-brain barrier. A ketogenic diet (KD) provides an alternative fuel for brain metabolism to treat impaired glucose transport. By reanalyzing exome data, we identified a de novo heterozygous SLC2A1 variant in a girl with epilepsy. After reversed phenotyping with neurometabolic tests, she was diagnosed with GLUT1DS1 and started on a KD. The patient's symptoms responded to the diet. Here, we report a patient with GLUT1DS1 with a novel SLC2A1 mutation. She also has a hemangioma which has not been reported in association with this syndrome before. CASE PRESENTATION A 5-year 8-month girl with global developmental delay, spasticity, intellectual disability, dysarthric speech, abnormal eye movements, and hemangioma. The electroencephalography (EEG) result revealed that she had epilepsy. Magnetic resonance imaging (MRI) showed that non-specific white matter abnormalities. Whole Exome Sequencing (WES) was previously performed, but the case remained unsolved. The re-analysis of WES data revealed a heterozygous splicing variant in the SLC2A1 gene. Segregation analysis with parental DNA samples indicated that the variant occurred de novo. Lumbar puncture (LP) confirmed the diagnosis, and the patient started on a KD. Her seizures responded to the KD. She has been seizure-free since shortly after the initiation of the diet. She also had decreased involuntary movements, her speech became more understandable, and her vocabulary increased after the diet. CONCLUSIONS We identified a novel de novo variant in the SLC2A1 gene in a patient who previously had a negative WES result. The patient has been diagnosed with GLUT1DS1. The syndrome is a treatable condition, but the differential diagnosis is not an easy process due to showing a wide range of phenotypic spectrum and the overlapping symptoms with other neurological diseases. The diagnosis necessitates a genomic testing approach. Our findings also highlight the importance of re-analysis to undiagnosed cases after initial WES to reveal disease-causing variants.
Collapse
Affiliation(s)
- Tugce Bozkurt
- Biostatistics and Bioinformatics Program, Graduate School of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Yasemin Alanay
- Division of Pediatric Genetics, Department of Pediatrics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ugur Isik
- Division of Pediatric Neurology, Department of Pediatrics, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ugur Sezerman
- Biostatistics and Bioinformatics Program, Graduate School of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
| |
Collapse
|
85
|
Lhomme T, Clasadonte J, Imbernon M, Fernandois D, Sauve F, Caron E, Lima N, Heras V, Martinez-Corral I, Müller-Fielitz H, Rasika S, Schwaninger M, Nogueiras R, Prevot V. Tanycytic networks mediate energy balance by feeding lactate to glucose-insensitive POMC neurons. J Clin Invest 2021; 131:e140521. [PMID: 34324439 DOI: 10.1172/jci140521] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Hypothalamic glucose sensing enables an organism to match energy expenditure and food intake to circulating levels of glucose, the main energy source of the brain. Here, we established that tanycytes of the hypothalamic arcuate nucleus, specialized glia that line the wall of the third ventricle, convert brain glucose supplies into lactate that they transmit through monocarboxylate transporters to arcuate proopiomelanocortin neurons, which integrate this signal to drive their activity and to adapt the metabolic response to meet physiological demands. Furthermore, this transmission required the formation of extensive Connexin-43 gap-junction-mediated metabolic networks by arcuate tanycytes. Selectively suppressing either tanycytic monocarboxylate transporters or gap junctions resulted in altered feeding behavior and energy metabolism. Tanycytic intercellular communication and lactate production are thus integral to the mechanism by which hypothalamic neurons that regulate energy and glucose homeostasis efficiently perceive alterations in systemic glucose levels as a function of the physiological state of the organism.
Collapse
Affiliation(s)
- Tori Lhomme
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Jerome Clasadonte
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Monica Imbernon
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Daniela Fernandois
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Florent Sauve
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Emilie Caron
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Natalia Lima
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Violeta Heras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ines Martinez-Corral
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Helge Müller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - S Rasika
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Ruben Nogueiras
- CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, INSERM U1172, Lille, France
| |
Collapse
|
86
|
Pan PY, Zhu J, Rizvi A, Zhu X, Tanaka H, Dreyfus CF. Synaptojanin1 deficiency upregulates basal autophagosome formation in astrocytes. J Biol Chem 2021; 297:100873. [PMID: 34126070 PMCID: PMC8258991 DOI: 10.1016/j.jbc.2021.100873] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 01/07/2023] Open
Abstract
Macroautophagy dysregulation is implicated in multiple neurological disorders, such as Parkinson's disease. While autophagy pathways are heavily researched in heterologous cells and neurons, regulation of autophagy in the astrocyte, the most abundant cell type in the mammalian brain, is less well understood. Missense mutations in the Synj1 gene encoding Synaptojanin1 (Synj1), a neuron-enriched lipid phosphatase, have been linked to Parkinsonism with seizures. Our previous study showed that the Synj1 haploinsufficient (Synj1+/−) mouse exhibits age-dependent autophagy impairment in multiple brain regions. Here, we used cultured astrocytes from Synj1-deficient mice to investigate its role in astrocyte autophagy. We report that Synj1 is expressed in low levels in astrocytes and represses basal autophagosome formation. We demonstrate using cellular imaging that Synj1-deficient astrocytes exhibit hyperactive autophagosome formation, represented by an increase in the size and number of GFP-microtubule-associated protein 1A/1B-light chain 3 structures. Interestingly, Synj1 deficiency is also associated with an impairment in stress-induced autophagy clearance. We show, for the first time, that the Parkinsonism-associated R839C mutation impacts autophagy in astrocytes. The impact of this mutation on the phosphatase function of Synj1 resulted in elevated basal autophagosome formation that mimics Synj1 deletion. We found that the membrane expression of the astrocyte-specific glucose transporter GluT-1 was reduced in Synj1-deficient astrocytes. Consistently, AMP-activated protein kinase activity was elevated, suggesting altered glucose sensing in Synj1-deficient astrocytes. Expressing exogenous GluT-1 in Synj1-deficient astrocytes reversed the autophagy impairment, supporting a role for Synj1 in regulating astrocyte autophagy via disrupting glucose-sensing pathways. Thus, our work suggests a novel mechanism for Synj1-related Parkinsonism involving astrocyte dysfunction.
Collapse
Affiliation(s)
- Ping-Yue Pan
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Justin Zhu
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Asma Rizvi
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Xinyu Zhu
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Hikari Tanaka
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Cheryl F Dreyfus
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
87
|
Patanè F, Pasquetti E, Sullo F, Tosto M, Romano C, Salafia S, Falsaperla R. SLC2A1 and Its Related Epileptic Phenotypes. JOURNAL OF PEDIATRIC NEUROLOGY 2021. [DOI: 10.1055/s-0041-1728668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
AbstractGlucose transporter type 1 deficiency syndrome (GLUT1DS) is caused by heterozygous, mostly de novo, mutations in SLC2A1 gene encoding the glucose transporter GLUT1, the most relevant energy transporter in the blood–brain barrier. GLUT1DS includes a broad spectrum of neurologic disturbances, from severe encephalopathy with developmental delay, to epilepsy, movement disorders, acquired microcephaly and atypical mild forms. For diagnosis, lumbar puncture and genetic analysis are necessary and complementary; an immediate response to ketogenic diet supports the diagnosis in case of high suspicion of disease and negative exams. The ketogenic diet is the first-line treatment and should be established at the initial stages of disease.
Collapse
Affiliation(s)
- Francesca Patanè
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Elisa Pasquetti
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Federica Sullo
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Monica Tosto
- Pediatric Postgraduate Residency Program, Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | | | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
- Unit of Neonatal Intensive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| |
Collapse
|
88
|
Good JM, Atallah I, Castro Jimenez M, Benninger D, Kuntzer T, Superti-Furga A, Tran C. NGS-Based Diagnosis of Treatable Neurogenetic Disorders in Adults: Opportunities and Challenges. Genes (Basel) 2021; 12:genes12050695. [PMID: 34066437 PMCID: PMC8148126 DOI: 10.3390/genes12050695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/28/2022] Open
Abstract
The identification of neurological disorders by next-generation sequencing (NGS)-based gene panels has helped clinicians understand the underlying physiopathology, resulting in personalized treatment for some rare diseases. While the phenotype of distinct neurogenetic disorders is generally well-known in childhood, in adulthood, the phenotype can be unspecific and make the standard diagnostic approach more complex. Here we present three unrelated adults with various neurological manifestations who were successfully diagnosed using NGS, allowing for the initiation of potentially life-changing treatments. A 63-year-old woman with progressive cognitive decline, pyramidal signs, and bilateral cataract was treated by chenodeoxycholic acid following the diagnosis of cerebrotendinous xanthomatosis due to a homozygous variant in CYP27A1. A 32-year-old man with adult-onset spastic paraplegia, in whom a variant in ABCD1 confirmed an X-linked adrenoleukodystrophy, was treated with corticoids for adrenal insufficiency. The third patient, a 28-year-old woman with early-onset developmental delay, epilepsy, and movement disorders was treated with a ketogenic diet following the identification of a variant in SLC2A1, confirming a glucose transporter type 1 deficiency syndrome. This case study illustrates the challenges in the timely diagnosis of medically actionable neurogenetic conditions, but also the considerable potential for improving patient health through modern sequencing technologies.
Collapse
Affiliation(s)
- Jean-Marc Good
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (J.-M.G.); (I.A.); (A.S.-F.)
| | - Isis Atallah
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (J.-M.G.); (I.A.); (A.S.-F.)
| | - Mayte Castro Jimenez
- Division of Neurology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (M.C.J.); (D.B.); (T.K.)
| | - David Benninger
- Division of Neurology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (M.C.J.); (D.B.); (T.K.)
| | - Thierry Kuntzer
- Division of Neurology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (M.C.J.); (D.B.); (T.K.)
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (J.-M.G.); (I.A.); (A.S.-F.)
| | - Christel Tran
- Division of Genetic Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (J.-M.G.); (I.A.); (A.S.-F.)
- Correspondence: ; Tel.: +41-79-556-5325
| |
Collapse
|
89
|
Liao JY, Salles PA, Shuaib UA, Fernandez HH. Genetic updates on paroxysmal dyskinesias. J Neural Transm (Vienna) 2021; 128:447-471. [PMID: 33929620 DOI: 10.1007/s00702-021-02335-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022]
Abstract
The paroxysmal dyskinesias are a diverse group of genetic disorders that manifest as episodic movements, with specific triggers, attack frequency, and duration. With recent advances in genetic sequencing, the number of genetic variants associated with paroxysmal dyskinesia has dramatically increased, and it is now evident that there is significant genotype-phenotype overlap, reduced (or incomplete) penetrance, and phenotypic variability. In addition, a variety of genetic conditions can present with paroxysmal dyskinesia as the initial symptom. This review will cover the 34 genes implicated to date and propose a diagnostic workflow featuring judicious use of whole-exome or -genome sequencing. The goal of this review is to provide a common understanding of paroxysmal dyskinesias so basic scientists, geneticists, and clinicians can collaborate effectively to provide diagnoses and treatments for patients.
Collapse
Affiliation(s)
- James Y Liao
- Center for Neurological Restoration, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Philippe A Salles
- Center for Neurological Restoration, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Centro de Trastornos del Movimiento, CETRAM, Santiago, Chile
| | - Umar A Shuaib
- Center for Neurological Restoration, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Hubert H Fernandez
- Center for Neurological Restoration, Neurological Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| |
Collapse
|
90
|
de Gusmão CM, Garcia L, Mikati MA, Su S, Silveira-Moriyama L. Paroxysmal Genetic Movement Disorders and Epilepsy. Front Neurol 2021; 12:648031. [PMID: 33833732 PMCID: PMC8021799 DOI: 10.3389/fneur.2021.648031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/22/2021] [Indexed: 01/08/2023] Open
Abstract
Paroxysmal movement disorders include paroxysmal kinesigenic dyskinesia, paroxysmal non-kinesigenic dyskinesia, paroxysmal exercise-induced dyskinesia, and episodic ataxias. In recent years, there has been renewed interest and recognition of these disorders and their intersection with epilepsy, at the molecular and pathophysiological levels. In this review, we discuss how these distinct phenotypes were constructed from a historical perspective and discuss how they are currently coalescing into established genetic etiologies with extensive pleiotropy, emphasizing clinical phenotyping important for diagnosis and for interpreting results from genetic testing. We discuss insights on the pathophysiology of select disorders and describe shared mechanisms that overlap treatment principles in some of these disorders. In the near future, it is likely that a growing number of genes will be described associating movement disorders and epilepsy, in parallel with improved understanding of disease mechanisms leading to more effective treatments.
Collapse
Affiliation(s)
- Claudio M. de Gusmão
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
- Department of Neurology, Universidade Estadual de Campinas (UNICAMP), São Paulo, Brazil
| | - Lucas Garcia
- Department of Medicine, Universidade 9 de Julho, São Paulo, Brazil
| | - Mohamad A. Mikati
- Division of Pediatric Neurology and Developmental Medicine, Duke University Medical Center, Durham, NC, United States
| | - Samantha Su
- Division of Pediatric Neurology and Developmental Medicine, Duke University Medical Center, Durham, NC, United States
| | - Laura Silveira-Moriyama
- Department of Neurology, Universidade Estadual de Campinas (UNICAMP), São Paulo, Brazil
- Department of Medicine, Universidade 9 de Julho, São Paulo, Brazil
- Education Unit, University College London Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
91
|
Classic Ketogenic Diet and Modified Atkins Diet in SLC2A1 Positive and Negative Patients with Suspected GLUT1 Deficiency Syndrome: A Single Center Analysis of 18 Cases. Nutrients 2021; 13:nu13030840. [PMID: 33806661 PMCID: PMC8000344 DOI: 10.3390/nu13030840] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/10/2021] [Accepted: 02/26/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Glucose transporter type 1 deficiency syndrome (GLUT1DS) is caused by mutations in the SLC2A1 gene and produces seizures, neurodevelopmental impairment, and movement disorders. Ketogenic dietary therapies (KDT) are the gold standard treatment. Similar symptoms may appear in SLC2A1 negative patients. The purpose is to evaluate the effectiveness of KDT in children with GLUT1DS suspected SLC2A1 (+) and (-), side effects (SE), and the impact on patients nutritional status. Methods: An observational descriptive study was conducted to describe 18 children (January 2009–August 2020). SLC2A1 analysis, seizures, movement disorder, anti-epileptic drugs (AEDS), anthropometry, SE, and laboratory assessment were monitored baseline and at 3, 6, 12, and 24 months after the onset of KDT. Results: 6/18 were SLC2A1(+) and 13/18 had seizures. In these groups, the age for debut of symptoms was higher. The mean time from debut to KDT onset was higher in SLC2A1(+). The modified Atkins diet (MAD) was used in 12 (5 SLC2A1(+)). Movement disorder improved (4/5), and a reduction in seizures >50% compared to baseline was achieved in more than half of the epileptic children throughout the follow-up. No differences in effectiveness were found according to the type of KDT. Early SE occurred in 33%. Long-term SE occurred in 10, 5, 7, and 5 children throughout the follow-up. The most frequent SE were constipation, hypercalciuria, and hyperlipidaemia. No differences in growth were found according to the SLC2A1 mutation or type of KDT. Conclusions: CKD and MAD were effective for SLC2A1 positive and negative patients in our cohort. SE were frequent, but mild. Permanent monitoring should be made to identify SE and nutritional deficits.
Collapse
|
92
|
Tajima D, Nakamura T, Ichinose F, Okamoto N, Tomonoh Y, Uda K, Furukawa R, Tashiro K, Matsuo M. Transient hypoglycorrhachia with paroxysmal abnormal eye movement in early infancy. Brain Dev 2021; 43:482-485. [PMID: 33248857 DOI: 10.1016/j.braindev.2020.11.006] [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: 06/24/2020] [Revised: 10/30/2020] [Accepted: 11/08/2020] [Indexed: 11/17/2022]
Abstract
Paroxysmal abnormal eye movement in early infancy is one of the initial symptoms of glucose transporter 1 deficiency syndrome (GLUT1DS). We describe four early infants with transient hypoglycorrhachia presenting with abnormal eye movements. Their symptoms disappeared after the introduction of a ketogenic diet (KD), and their development was normal. Since no variants in SLC2A1 were detected, the CSF-to-blood glucose ratios (C/B) were re-examined, and within normal range. None of the four patients displayed recurrent symptoms after withdrawal from the KD. Because long-term KD has potential adverse effects and could affect the quality of life of patients and their families, re-examination of CSF glucose during late infancy should be considered in the case of absence of the SLC2A1 pathogenic variant.
Collapse
Affiliation(s)
- Daisuke Tajima
- Department of Pediatrics, Faculty of Medicine, Saga University, Japan; Department of Pediatrics, Karatsu Red Cross Hospital, Japan.
| | - Takuji Nakamura
- Department of Pediatrics, Faculty of Medicine, Saga University, Japan
| | - Fumio Ichinose
- Department of Pediatrics, Faculty of Medicine, Saga University, Japan
| | - Nobuhiko Okamoto
- Department of Medical Genetics, Osaka Women's and Children's Hospital, Japan
| | - Yuko Tomonoh
- Department of Pediatrics, Faculty of Medicine, Fukuoka University, Japan
| | - Keiko Uda
- Department of Pediatrics, Faculty of Medicine, Saga University, Japan
| | - Rie Furukawa
- Department of Pediatrics, Faculty of Medicine, Saga University, Japan; Department of Pediatrics, Karatsu Red Cross Hospital, Japan
| | - Katsuya Tashiro
- Department of Pediatrics, Faculty of Medicine, Saga University, Japan; Department of Pediatrics, Karatsu Red Cross Hospital, Japan
| | - Muneaki Matsuo
- Department of Pediatrics, Faculty of Medicine, Saga University, Japan
| |
Collapse
|
93
|
Tang M, Park SH, Petri S, Yu H, Rueda CB, Abel ED, Kim CY, Hillman EM, Li F, Lee Y, Ding L, Jagadish S, Frankel WN, De Vivo DC, Monani UR. An early endothelial cell-specific requirement for Glut1 is revealed in Glut1 deficiency syndrome model mice. JCI Insight 2021; 6:145789. [PMID: 33351789 PMCID: PMC7934852 DOI: 10.1172/jci.insight.145789] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/10/2020] [Indexed: 12/30/2022] Open
Abstract
Paucity of the glucose transporter-1 (Glut1) protein resulting from haploinsufficiency of the SLC2A1 gene arrests cerebral angiogenesis and disrupts brain function to cause Glut1 deficiency syndrome (Glut1 DS). Restoring Glut1 to Glut1 DS model mice prevents disease, but the precise cellular sites of action of the transporter, its temporal requirements, and the mechanisms linking scarcity of the protein to brain cell dysfunction remain poorly understood. Here, we show that Glut1 functions in a cell-autonomous manner in the cerebral microvasculature to affect endothelial tip cells and, thus, brain angiogenesis. Moreover, brain endothelial cell–specific Glut1 depletion not only triggers a severe neuroinflammatory response in the Glut1 DS brain, but also reduces levels of brain-derived neurotrophic factor (BDNF) and causes overt disease. Reduced BDNF correlated with fewer neurons in the Glut1 DS brain. Controlled depletion of the protein demonstrated that brain pathology and disease severity was greatest when Glut1 scarcity was induced neonatally, during brain angiogenesis. Reducing Glut1 at later stages had mild or little effect. Our results suggest that targeting brain endothelial cells during early development is important to ensure proper brain angiogenesis, prevent neuroinflammation, maintain BDNF levels, and preserve neuron numbers. This requirement will be essential for any disease-modifying therapeutic strategy for Glut1 DS.
Collapse
Affiliation(s)
- Maoxue Tang
- Department of Neurology and.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, New York, USA
| | - Sarah H Park
- Department of Neurology and.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, New York, USA
| | - Sabrina Petri
- Department of Genetics & Development and the Institute for Genomic Medicine, Columbia University, New York, New York, USA
| | - Hang Yu
- Departments of Biomedical Engineering and Radiology, Mortimer B. Zuckerman Mind Brain Behavior Institute and Kavli Institute for Brain Science, Columbia University, New York, New York, USA
| | - Carlos B Rueda
- Department of Neurology and.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, New York, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Carla Y Kim
- Departments of Biomedical Engineering and Radiology, Mortimer B. Zuckerman Mind Brain Behavior Institute and Kavli Institute for Brain Science, Columbia University, New York, New York, USA
| | - Elizabeth Mc Hillman
- Departments of Biomedical Engineering and Radiology, Mortimer B. Zuckerman Mind Brain Behavior Institute and Kavli Institute for Brain Science, Columbia University, New York, New York, USA
| | - Fanghua Li
- Department of Neurology and.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, New York, USA
| | - Yeojin Lee
- Columbia Stem Cell Initiative and Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - Lei Ding
- Columbia Stem Cell Initiative and Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - Smitha Jagadish
- Rare & Neurological Diseases Research, Sanofi Genzyme, Framingham, Massachusetts, USA
| | - Wayne N Frankel
- Department of Genetics & Development and the Institute for Genomic Medicine, Columbia University, New York, New York, USA
| | - Darryl C De Vivo
- Department of Neurology and.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, New York, USA
| | - Umrao R Monani
- Department of Neurology and.,Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, New York, USA.,Department of Pathology & Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
94
|
Chen X, Zhao Y, Gao Y, Qi Y, Du J. Outcomes in hepatocellular carcinoma patients undergoing sorafenib treatment: toxicities, cellular oxidative stress, treatment adherence, and quality of life: Erratum. Anticancer Drugs 2021; 32:345-364. [PMID: 33417326 DOI: 10.1097/cad.0000000000001029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Xiaotong Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Yunshuo Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yuanming Qi
- School of Life Sciences, Zhengzhou University, Zhengzhou
| | - Jiangfeng Du
- School of Life Sciences, Zhengzhou University, Zhengzhou
| |
Collapse
|
95
|
Natsume J, Ishihara N, Azuma Y, Nakata T, Takeuchi T, Tanaka M, Sakaguchi Y, Okai Y, Ito Y, Yamamoto H, Ohno A, Kidokoro H, Hattori A, Nabatame S, Kato K. Lenticular nuclei to thalamic ratio on PET is useful for diagnosis of GLUT1 deficiency syndrome. Brain Dev 2021; 43:69-77. [PMID: 32739099 DOI: 10.1016/j.braindev.2020.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To establish an objective method of [18F]-fluorodeoxyglucose positron emission tomography (FDG-PET) that can assist in the diagnosis of glucose transporter 1 deficiency syndrome (GLUT1-DS). METHODS FDG-PET was performed in 8 patients with a mean age of 12.5 years (range, 2-22 years) with GLUT1-DS. Their PET findings were compared with those of 45 controls with a mean age of 11.2 years (range, 2-21 years) by statistical parametric mapping (SPM12, Welcome Neurological Institute). The controls had epilepsy of unknown etiology and normal MRI findings. The age-adjusted ratios of mean radioactivities in regions of interest (ROIs) of bilateral lenticular nuclei, thalami, and the whole cerebral cortex were also measured. The sensitivities and specificities of the ratios for the differential diagnosis of GLUT1-DS were also determined. RESULTS SPM showed significantly decreased uptake in bilateral thalami and increased uptake in bilateral lenticular nuclei in patients with GLUT1-DS. There were no areas in the cerebral cortex with significant differences between patients and controls. On ROI analysis, by setting the cut-off value of the age-adjusted lenticular nuclei/thalami radioactivity ratio to 1.54, patients with GLUT1-DS were differentiated from controls with sensitivity of 1.00 and specificity of 0.98. CONCLUSION The age-adjusted lenticular nuclei/thalami radioactivity ratio on PET can distinguish patients with GLUT1-DS from patients with epilepsy of unknown etiology with high sensitivity and specificity. It is important to pay attention to the metabolism of the lenticular nuclei and thalami on PET for the diagnosis of GLUT1-DS.
Collapse
Affiliation(s)
- Jun Natsume
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Department of Developmental Disability Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan; Brain and Mind Research Center, Nagoya University, Nagoya, Japan.
| | - Naoko Ishihara
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshiteru Azuma
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomohiko Nakata
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoya Takeuchi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaharu Tanaka
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoko Sakaguchi
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yu Okai
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuji Ito
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Yamamoto
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsuko Ohno
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyuki Kidokoro
- Department of Pediatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan; Brain and Mind Research Center, Nagoya University, Nagoya, Japan
| | - Ayako Hattori
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shin Nabatame
- Department of Pediatrics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Katsuhiko Kato
- Department of Radiological and Medical Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
96
|
Gavrilovici C, Rho JM. Metabolic epilepsies amenable to ketogenic therapies: Indications, contraindications, and underlying mechanisms. J Inherit Metab Dis 2021; 44:42-53. [PMID: 32654164 DOI: 10.1002/jimd.12283] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 12/20/2022]
Abstract
Metabolic epilepsies arise in the context of rare inborn errors of metabolism (IEM), notably glucose transporter type 1 deficiency syndrome, succinic semialdehyde dehydrogenase deficiency, pyruvate dehydrogenase complex deficiency, nonketotic hyperglycinemia, and mitochondrial cytopathies. A common feature of these disorders is impaired bioenergetics, which through incompletely defined mechanisms result in a wide spectrum of neurological symptoms, such as epileptic seizures, developmental delay, and movement disorders. The ketogenic diet (KD) has been successfully utilized to treat such conditions to varying degrees. While the mechanisms underlying the clinical efficacy of the KD in IEM remain unclear, it is likely that the proposed heterogeneous targets influenced by the KD work in concert to rectify or ameliorate the downstream negative consequences of genetic mutations affecting key metabolic enzymes and substrates-such as oxidative stress and cell death. These beneficial effects can be broadly grouped into restoration of impaired bioenergetics and synaptic dysfunction, improved redox homeostasis, anti-inflammatory, and epigenetic activity. Hence, it is conceivable that the KD might prove useful in other metabolic disorders that present with epileptic seizures. At the same time, however, there are notable contraindications to KD use, such as fatty acid oxidation disorders. Clearly, more research is needed to better characterize those metabolic epilepsies that would be amenable to ketogenic therapies, both experimentally and clinically. In the end, the expanded knowledge base will be critical to designing metabolism-based treatments that can afford greater clinical efficacy and tolerability compared to current KD approaches, and improved long-term outcomes for patients.
Collapse
Affiliation(s)
- Cezar Gavrilovici
- Departments of Neurosciences and Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, California, USA
| | - Jong M Rho
- Departments of Neurosciences and Pediatrics, University of California San Diego, Rady Children's Hospital, San Diego, California, USA
| |
Collapse
|
97
|
Kramer J, Smith L. Ketogenic Diet in Glut 1 Deficiency Through the Life Cycle: Pregnancy to Neonate to Preschooler. Child Neurol Open 2021; 8:2329048X211034655. [PMID: 34541019 PMCID: PMC8444274 DOI: 10.1177/2329048x211034655] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/02/2021] [Indexed: 11/15/2022] Open
Abstract
A 19-year-old woman with glucose transporter type 1 deficiency syndrome (Glut1DS) treated with ketogenic diet therapy (KDT) became pregnant. Her pregnancy included close monitoring of her diet as well as the fetus. Shortly after delivery, a lumbar puncture was performed followed by confirmatory genetic test diagnosing the neonate with Glut1DS. The neonate was placed on KDT and has been maintained on diet since infancy. The child is now 5 years of age, asymptomatic, and excelling developmentally. This case presents 2 management challenges, that of a patient with Glut1DS during pregnancy followed by managing a neonate on KDT with minimal guidance available in the literature due to the relative rarity of the condition and this unique situation.
Collapse
Affiliation(s)
- Jennifer Kramer
- Riley Hospital for Children at Indiana University
Health, Indianapolis, IN, USA
| | - Lisa Smith
- Riley Hospital for Children at Indiana University
Health, Indianapolis, IN, USA
| |
Collapse
|
98
|
Fei Y, Shi R, Song Z, Wu J. Metabolic Control of Epilepsy: A Promising Therapeutic Target for Epilepsy. Front Neurol 2020; 11:592514. [PMID: 33363507 PMCID: PMC7753014 DOI: 10.3389/fneur.2020.592514] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022] Open
Abstract
Epilepsy is a common neurological disease that is not always controlled, and the ketogenic diet shows good antiepileptic effects drug-resistant epilepsy or seizures caused by specific metabolic defects via regulating the metabolism. The brain is a vital organ with high metabolic demands, and epileptic foci tend to exhibit high metabolic characteristics. Accordingly, there has been growing interest in the relationship between brain metabolism and epilepsy in recent years. To date, several new antiepileptic therapies targeting metabolic pathways have been proposed (i.e., inhibiting glycolysis, targeting lactate dehydrogenase, and dietary therapy). Promising strategies to treat epilepsy via modulating the brain's metabolism could be expected, while a lack of thorough understanding of the role of brain metabolism in the control of epilepsy remains. Herein, this review aims to provide insight into the state of the art concerning the brain's metabolic patterns and their association with epilepsy. Regulation of neuronal excitation via metabolic pathways and antiepileptic therapies targeting metabolic pathways are emphasized, which could provide a better understanding of the role of metabolism in epilepsy and could reveal potential therapeutic targets.
Collapse
Affiliation(s)
- Yanqing Fei
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ruting Shi
- Department of Rehabilitation, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhi Song
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jinze Wu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
99
|
McDonald TJW, Cervenka MC. Ketogenic Diet Therapies for Seizures and Status Epilepticus. Semin Neurol 2020; 40:719-729. [PMID: 33155184 DOI: 10.1055/s-0040-1719077] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ketogenic diet therapies are high-fat, low-carbohydrate diets designed to mimic a fasting state. Although initially developed nearly one century ago for seizure management, most clinical trials for the management of drug-resistant epilepsy in children as well as adults have been conducted over the last 3 decades. Moreover, ketogenic diets offer promising new adjunctive strategies in the critical care setting for the resolution of acute status epilepticus when traditional antiseizure drugs and anesthetic agents fail. Here, we review the history of ketogenic diet development, the clinical evidence supporting its use for the treatment of drug-resistant epilepsy in children and adults, and the early evidence supporting ketogenic diet feasibility, safety, and potential efficacy in the management of status epilepticus.
Collapse
|
100
|
Profaci CP, Munji RN, Pulido RS, Daneman R. The blood-brain barrier in health and disease: Important unanswered questions. J Exp Med 2020; 217:151582. [PMID: 32211826 PMCID: PMC7144528 DOI: 10.1084/jem.20190062] [Citation(s) in RCA: 419] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/21/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
The blood vessels vascularizing the central nervous system exhibit a series of distinct properties that tightly control the movement of ions, molecules, and cells between the blood and the parenchyma. This "blood-brain barrier" is initiated during angiogenesis via signals from the surrounding neural environment, and its integrity remains vital for homeostasis and neural protection throughout life. Blood-brain barrier dysfunction contributes to pathology in a range of neurological conditions including multiple sclerosis, stroke, and epilepsy, and has also been implicated in neurodegenerative diseases such as Alzheimer's disease. This review will discuss current knowledge and key unanswered questions regarding the blood-brain barrier in health and disease.
Collapse
Affiliation(s)
- Caterina P Profaci
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Roeben N Munji
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Robert S Pulido
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| | - Richard Daneman
- Department of Neurosciences, University of California, San Diego, San Diego, CA.,Department of Pharmacology, University of California, San Diego, San Diego, CA
| |
Collapse
|