1
|
Yamazaki S, Kaneko S, Shimbo A, Irabu H, Ogino R, Miyamoto T, Izawa K, Segawa Y, Kakizaki J, Mori M, Shimizu M. Overlapping Aicardi-Goutières and Singleton-Merten syndromes with a heterozygous gain-of-function mutation in IFIH1 mimicking juvenile idiopathic arthritis. Immunol Med 2025:1-5. [PMID: 40116369 DOI: 10.1080/25785826.2025.2479148] [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: 09/03/2024] [Accepted: 03/01/2025] [Indexed: 03/23/2025] Open
Abstract
Aicardi-Goutières syndrome (AGS) and Singleton-Merten syndrome (SMS) are associated with heterozygous gain-of-function mutations in the interferon induced with helicase C domain 1 (IFIH1) gene. Recent reports describe patients exhibiting overlapping clinical features of AGS and SMS, along with marked type I interferon (IFN) overproduction. However, the clinical characteristics and optimal treatment strategies remain unclear. Herein, we present a patient with overlapping clinical features of AGS and SMS who was initially misdiagnosed with juvenile idiopathic arthritis. Surgical soft tissue release of the hip and knee joints improved joint deformities and spastic paraparesis. Baricitinib effectively treated refractory chilblains and skin ulcers while reducing IFN-stimulated gene overexpression in peripheral blood. These findings indicate that baricitinib may be a safe and effective treatment for AGS-SMS overlap, and surgical intervention may be a viable option for refractory joint deformities with spastic paraparesis.
Collapse
Affiliation(s)
- Susumu Yamazaki
- Department of Pediatrics and Adolescent Medicine, Juntendo University Nerima Hospital, Tokyo, Japan
- Department of Lifetime Clinical Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shuya Kaneko
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Asami Shimbo
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hitoshi Irabu
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryo Ogino
- Department of Pediatrics, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Miyamoto
- Department of Pediatrics, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Kazushi Izawa
- Department of Pediatrics, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Yuko Segawa
- Department of Orthopedic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jun Kakizaki
- Department of Orthopedic Surgery, Chiba Children's Hospital, Chiba, Japan
| | - Masaaki Mori
- Department of Lifetime Clinical Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaki Shimizu
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
2
|
Gavazzi F, Charsar B, Hamilton E, Erler JA, Patel V, Woidill S, Sevagamoorthy A, Helman G, Schmidt J, Pizzino A, Muirhead K, Takanohashi A, Bonkowsky JL, Meyerhoffer K, Simons C, Doi H, Satoko M, Matsumoto N, Delgado MR, Sanchez-Castillo M, Wang J, de Carvalho DR, Tournev I, Chamova T, Jordanova A, Clegg NJ, Nicita F, Bertini E, Teng M, Williams D, Tonduti D, Houlden H, Stellingwerff M, Wassmer E, Garcia-Cazorla A, Bernard G, Mirchi A, Toutounchi H, Wolf NI, van der Knaap MS, Shults J, Adang LA, Vanderver AL. The natural history of variable subtypes in pediatric-onset TUBB4A-related leukodystrophy. Mol Genet Metab 2025; 144:109048. [PMID: 39951964 PMCID: PMC11875891 DOI: 10.1016/j.ymgme.2025.109048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/21/2024] [Accepted: 01/29/2025] [Indexed: 02/17/2025]
Abstract
We establish the natural history of pediatric-onset TUBB4A-related leukodystrophy to improve clinical trial readiness through a medical record-based longitudinal study. An international cohort of 216 individuals with pediatric-onset TUBB4A-related leukodystrophy was included. Demographic information and medical events were extracted from medical records or publications. Retrospective scores (Gross Motor Function - Metachromatic Leukodystrophy [GMFC-MLD] and Communication Function Classification System [CFCS]) were applied to assess function. Survival analysis distinguished differences in longitudinal neurocognitive function and time to event outcomes between subtypes. A decision tree predicted independent ambulation from early motor milestones. Genotype (p.Asp249Asn vs non-p.Asp249Asn) and independent sitting by age 9 months predicted ambulation by 3 years, and stratification into three subgroups: early-infantile (non- sitting by 9 months), late-infantile (normal early milestones without the common p.Asp249Asn mutation), and a cohort of p.Asp249Asn late-infantile onset individuals. Median age at symptom onset was 0.71 years (interquartile range: [0.33, 1.50]). Common symptoms at onset include delayed development and tone abnormalities (n = 125, 66.5 % and n = 77, 43.0 %). The most common medical complications included scoliosis (N = 51/142), hip dislocation (N = 30/101), and seizures (N = 51/163). The early-infantile more severely affected cohort had a greater prevalence of G-tube placement, scoliosis, and seizure compared to the late-infantile form (p < 0.01). Peak motor and communication abilities were comparable between the p.Asp249Asn and the late infantile cohorts. Despite the acquisition of early milestones, individuals with p.Asp249Asn showed a more rapid decline of functional abilities compared to other late infantile forms (log-rank p = 0.0002). Better understanding of TUBB4A-related leukodystrophy subtypes will improve clinical care, allow targeted preventive interventions, and permit disease stratification for future disease-modifying clinical trials.
Collapse
Affiliation(s)
- Francesco Gavazzi
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Brittany Charsar
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Eline Hamilton
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Netherlands
| | - Jacqueline A Erler
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Virali Patel
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sarah Woidill
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anjana Sevagamoorthy
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Guy Helman
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Johanna Schmidt
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amy Pizzino
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kayla Muirhead
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Asako Takanohashi
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joshua L Bonkowsky
- Division of Pediatric Neurology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Personalized Medicine, Primary Children's Hospital, Salt Lake City, UT, USA
| | - Kelsee Meyerhoffer
- Division of Pediatric Neurology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Cas Simons
- Murdoch Childrens Research Institute, Melbourne, Australia
| | - Hiroshi Doi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Miyatake Satoko
- Department of Human Genetics, Yokoama City University, Yokohama, Japan
| | | | - Mauricio R Delgado
- Department of Neurology, University of Texas Southwestern Medical Center, USA
| | | | - Jingming Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | | | - Ivailo Tournev
- Clinic of Nervous Diseases, University Hospital Aleksandrovska, Department of Neurology, Medical University-Sofia, Bulgaria; Department of Cognitive Science and Psychology, New Bulgarian University, Bulgaria
| | - Teodora Chamova
- Department of Neurology, University Hospital Alexandrovska, Medical University Sofia, Sofia, Bulgaria
| | - Albena Jordanova
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical University - Sofia, Sofia, Bulgaria; Molecular Neurogenomics Group, Center for Molecular Neurology, VIB, Antwerpen, Belgium; Molecular Neurogenomics Group, Center for Molecular Neurology, Department of Biomedical Sciences, University of Antwerp, Antwerpen, Belgium
| | - Nancy J Clegg
- Division of Clinical Research, Department of Research, Scottish Rite for Children, Dallas, TX, USA
| | - Francesco Nicita
- Unit of Neuromuscular and Neurodegenerative Diseases, Department of Neuroscience and Neurorehabilitation, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Enrico Bertini
- Research Unit of Neuromuscular and Neurodegenerative Diseases, Bambino Gesu' Children's Hospital, IRCCS, Rome, Italy
| | | | | | - Davide Tonduti
- Unit of Pediatric Neurology, C.O.A.L.A (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children's Hospital, Milan, Italy; Department of Biomedical and Clinical Sciences, L. Sacco University Hospital, Università degli Studi di Milano, Milan, Italy
| | - Henry Houlden
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Evangeline Wassmer
- Neurology Department, Birmingham Children's Hospital, Institute of Health and Neurodevelopment, Aston University, Birmingham, UK
| | - Angeles Garcia-Cazorla
- Institut de Recerca Sant Joan de Déu, Barcelona, Spain; Neurology Department, Neurometabolic Unit and Synaptic Metabolism Lab, Institut Pediàtric de Recerca, Hospital Sant Joan de Déu, metabERN and CIBERER-ISCIII, Barcelona, Spain
| | - Geneviève Bernard
- Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University, Montreal, Quebec, Canada; Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Centre, Montreal, Quebec, Canada
| | - Amytice Mirchi
- Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Helia Toutounchi
- Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Nicole I Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, Netherlands
| | - Marjo S van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, Netherlands
| | - Justine Shults
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura A Adang
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adeline L Vanderver
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
3
|
Al Zayer L, Al Zayer M, Alrujaib A, Buhasan A, Jadah RHSH. Delayed Diagnosis of Aicardi-Goutières Syndrome in a 10-Year-Old Female Child With TREX1 Mutation: A Case Report. Cureus 2025; 17:e79730. [PMID: 40161088 PMCID: PMC11953621 DOI: 10.7759/cureus.79730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Aicardi-Goutières syndrome (AGS) is a rare hereditary autoinflammatory disease of subacute encephalopathy, characterized by a wide range of neurological and extra-neurological manifestations, primarily affecting the brain and skin. Key features include increased expression of interferon-stimulated genes (ISGs), acquired microcephaly, dystonia, spasticity, chilblains, and panniculitis. Radiological findings include cerebral calcifications, leukodystrophy, cerebral atrophy, and cerebrospinal fluid abnormalities such as chronic lymphocytosis and elevated interferon-alpha (INF-α) levels. Seven pathogenic genes have been identified in association with AGS. The management of AGS is primarily supportive, as there is currently no definitive cure for the condition. The primary goals are to address symptoms, improve quality of life, and prevent complications. The aim of this study was to emphasize the importance of early diagnosis of this rare genetic condition, as timely identification enables prompt intervention and management. Early diagnosis improves clinical outcomes, enhances the quality of life for affected individuals, and provides valuable guidance for family planning and genetic counseling.
Collapse
Affiliation(s)
- Leen Al Zayer
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, BHR
| | | | | | - Asal Buhasan
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, BHR
| | | |
Collapse
|
4
|
Elitt CM, Volpe JJ. Degenerative Disorders of the Newborn. VOLPE'S NEUROLOGY OF THE NEWBORN 2025:967-1007.e17. [DOI: 10.1016/b978-0-443-10513-5.00033-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Politano D, Tonduti D, Battini R, Fazzi E, Orcesi S. Exploring emerging JAK inhibitors in the treatment of Aicardi-Goutières syndrome. Expert Opin Emerg Drugs 2024:1-19. [PMID: 39704072 DOI: 10.1080/14728214.2024.2445508] [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: 07/15/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION Aicardi-Goutières syndrome (AGS) is a genetically heterogeneous monogenic autoinflammatory disorder classified as an 'interferonopathy'. Nine genes have been implicated in AGS, encoding proteins involved in nucleic acid clearance, repair, sensing, or histone pre-mRNA processing. Dysregulation in these pathways leads to excessive type I interferon production, the primary driver of the disease. AGS typically presents with early-life neurological regression, followed by stabilization with varying degrees of neurological impairment and common extra-neurological features, such as chilblains. Advances in understanding AGS pathogenesis have enabled the development of new therapies, with JAK inhibitors emerging as the most studied option for reducing interferon-mediated effects. AREAS COVERED This review discusses the clinical features, genetic basis, and molecular pathways of AGS while tracing the evolution of its therapeutic strategies. Particular emphasis is placed on JAK inhibitors, which target proteins activated by type I interferons, providing a novel direction in treatment. EXPERT OPINION Inhibitors effectively reduce extra-neurological symptoms in AGS, though their impact on neurological outcomes remains unclear. The unknown natural history of AGS limits treatment evaluation. Despite growing insights, key aspects of pathogenesis and treatment optimization - including timing, administration, and long-term effects - remain unresolved, highlighting the need for further research.
Collapse
Affiliation(s)
- Davide Politano
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Davide Tonduti
- Unit of Pediatric Neurology, COALA Center for Diagnosis and Treatment of Leukodystrophies, V. Buzzi Children's Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elisa Fazzi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Unit of Child Neurology and Psychiatry, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Simona Orcesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
6
|
Qiang W, Deng WJ, Song SL, Pan LH. Identification and analysis correlation between hub genes and immune cell infiltration related to LPS-induced cognitive impairment. Heliyon 2024; 10:e37101. [PMID: 39286150 PMCID: PMC11403500 DOI: 10.1016/j.heliyon.2024.e37101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 08/19/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Background The occurrence of immunity and inflammation outside the central nervous system frequently results in acute cognitive impairment among elderly patients. However, there is currently a lack of standardized methods for diagnosing acute cognitive impairment. The objective of our study was to identify potential mRNA biomarkers and investigate the pathogenesis of acute cognitive impairment in mice brains. Methods To analyze changes in hub genes associated with acute cognitive impairment, bioinformatics analysis was performed on the mouse brain injury data of sterile saline control group and lipopolysaccharide (LPS) induced experimental group in Gene Expression Omnibus (GEO). Functional analysis was conducted using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), which facilitated to identify some potential mRNA biomarkers for hub gene expression in mice brains. Additionally, the "CIBERSORT X″ R kit was employed to examine immune cell infiltrations of mice brains in LPS group and saline group. Results In the LPS and saline group, 102 significantly upregulated differentially expressed genes (DEGs) and 32 downregulated DEGs were identified. The pathway enrichment analysis using GO and KEGG revealed that these DEGs were mainly related to the regulation of cytokine, cytokine-cytokine receptor interaction, as well as protein interaction with cytokine and cytokine receptor. Immune cell infiltration analysis indicated potential involvement of M1 macrophages, NK cells resting, T cells CD4 memory, and T cells CD8 naive in the process of cognitive impairment. By constructing a protein-protein interaction (PPI) network, five hub genes (Cxcl10, Cxcl12, Cxcr3, Gbp2, and Ifih1) showed significant associations with immune cell types by using a threshold Spearman's rank correlation coefficient of R > 0.50 and p < 0.05. Conclusion The mRNA expression profile of the mice brain tissues in the LPS group differed from that in the normal saline group. These significantly expressed mRNAs may act an importance in the pathogenesis of acute cognitive impairment through mechanisms involving immunity and neuroinflammation.
Collapse
Affiliation(s)
- Wang Qiang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Guangxi, China
| | - Wen Juan Deng
- Department of Radiology, Guangxi Medical University Cancer Hospital, Guangxi, China
| | - Shu Ling Song
- Department of Radiology, Guangxi Medical University Cancer Hospital, Guangxi, China
- The Fourth People's Hospital of Nanning, Guangxi, China
| | - Ling Hui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Guangxi, China
- Guangxi Clinical Research Center for Anesthesiology, Guangxi, China
- Guangxi Engineering Research Center for Tissue & Organ Injury and Repair Medicine, Guangxi, China
- Guangxi Health Commission Key Laboratory of Basic Science and Prevention of Perioperative Organ Disfunction, Guangxi, China
| |
Collapse
|
7
|
Zhang S, Zhang W, Ding C, Ren X, Fang F, Wu Y. Neurophenotype and genetic analysis of children with Aicardi-Goutières syndrome in China. Pediatr Investig 2024; 8:193-200. [PMID: 39347527 PMCID: PMC11427897 DOI: 10.1002/ped4.12428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/27/2024] [Indexed: 10/01/2024] Open
Abstract
Importance Aicardi-Goutières syndrome (AGS) is a rare genetic disorder mainly affecting the central nervous system and autoimmunity. However, research on AGS among Chinese patients is limited. Objective To summarize the neurologic phenotypes and genetic causes in pediatric AGS patients, providing insights for early recognition and diagnosis in the Chinese population. Methods Clinical features and neuroimaging results of the patients diagnosed with AGS from Beijing Children's Hospital between January 2018 and January 2022 were collected. Whole exome sequencing was used for genetic analysis. Results A total of 15 patients was included, all presenting with various neurological symptoms, including developmental delay (100%), motor skill impairment (100%), language disability (78.6%), dystonia (93.3%), microcephaly (73.3%), sleep disorders (26.7%), regression (20.0%), vessel disease (6.7%), and epilepsy (6.7%). Neuroimaging revealed intracranial calcification (86.7%), cerebral atrophy (73.3%), and leukodystrophy (73.3%). Seven genes were identified, with TREX1 being the most common (40.0%, 6/15), followed by IFIH1 (20.0%, 3/15). Variant c.294dupA (p.C99Mfs*3) was detected in four unrelated patients, accounting for 66.7% (4/6) patients with the TREX1 variant. A literature review showed that TREX1 gene mutations in 35.6% (21/59) of AGS patients among the Chinese population. Interpretation Neurological symptoms are the most prevalent and severe presentation of AGS. Diagnosis may be considered when symptoms such as developmental delay, dystonia, microcephaly, brain calcification, and leukodystrophy emerge. TREX1 mutations are predominant in the Chinese population.
Collapse
Affiliation(s)
- Shen Zhang
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Weihua Zhang
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Changhong Ding
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Xiaotun Ren
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Fang Fang
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| | - Yun Wu
- Department of Neurology Beijing Children's Hospital Capital Medical University National Center for Children's Health Beijing China
| |
Collapse
|
8
|
Gavazzi F, Gonzalez CD, Arnold K, Swantkowski M, Charlton L, Modesti N, Dar AA, Vanderver A, Bennett M, Adang LA. Nucleotide metabolism, leukodystrophies, and CNS pathology. J Inherit Metab Dis 2024; 47:860-875. [PMID: 38421058 PMCID: PMC11358362 DOI: 10.1002/jimd.12721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
The balance between a protective and a destructive immune response can be precarious, as exemplified by inborn errors in nucleotide metabolism. This class of inherited disorders, which mimics infection, can result in systemic injury and severe neurologic outcomes. The most common of these disorders is Aicardi Goutières syndrome (AGS). AGS results in a phenotype similar to "TORCH" infections (Toxoplasma gondii, Other [Zika virus (ZIKV), human immunodeficiency virus (HIV)], Rubella virus, human Cytomegalovirus [HCMV], and Herpesviruses), but with sustained inflammation and ongoing potential for complications. AGS was first described in the early 1980s as familial clusters of "TORCH" infections, with severe neurology impairment, microcephaly, and basal ganglia calcifications (Aicardi & Goutières, Ann Neurol, 1984;15:49-54) and was associated with chronic cerebrospinal fluid (CSF) lymphocytosis and elevated type I interferon levels (Goutières et al., Ann Neurol, 1998;44:900-907). Since its first description, the clinical spectrum of AGS has dramatically expanded from the initial cohorts of children with severe impairment to including individuals with average intelligence and mild spastic paraparesis. This broad spectrum of potential clinical manifestations can result in a delayed diagnosis, which families cite as a major stressor. Additionally, a timely diagnosis is increasingly critical with emerging therapies targeting the interferon signaling pathway. Despite the many gains in understanding about AGS, there are still many gaps in our understanding of the cell-type drivers of pathology and characterization of modifying variables that influence clinical outcomes and achievement of timely diagnosis.
Collapse
Affiliation(s)
- Francesco Gavazzi
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Kaley Arnold
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Meghan Swantkowski
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lauren Charlton
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nicholson Modesti
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Asif A. Dar
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Adeline Vanderver
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mariko Bennett
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Laura A. Adang
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Peixoto de Barcelos I, Jan AK, Modesti N, Woidill S, Gavazzi F, Isaacs D, D'Aiello R, Sevagamoorthy A, Charlton L, Pizzino A, Schmidt J, van Haren K, Keller S, Eichler F, Emrick LT, Fraser JL, Shults J, Vanderver A, Adang LA. Systemic complications of Aicardi Goutières syndrome using real-world data. Mol Genet Metab 2024; 143:108578. [PMID: 39332260 DOI: 10.1016/j.ymgme.2024.108578] [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: 05/29/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVE Aicardi Goutières Syndrome (AGS) is a rare genetic interferonopathy associated with diverse multisystemic complications. A critical gap exists in our understanding of its longitudinal, systemic disease burden, complicated by delayed diagnosis. To address this need, real-world data extracted from existing medical records were used to characterize the longitudinal disease burden. METHODS All subjects (n = 167) with genetically confirmed AGS enrolled in the Myelin Disorders Biorepository Project (MDBP) were included. As available in medical records, information was collected on subject demographics, age of onset, and disease complications. Information from published cases of AGS (2007-2022; n = 129) with individual-level data was also collected. Neurologic severity at the last available encounter was determined by retrospectively assigning the AGS Severity Scale [severe (0-3), moderate (4-8), and mild (9-11)]. RESULTS The genotype frequency in the natural history cohort was TREX1 (n = 26, 15.6 %), RNASEH2B (n = 50, 29.9 %), RNASEH2C (n = 3, 1.8 %), RNASEH2A (n = 7, 4.2 %), SAMHD1 (n = 25, 15.0 %), ADAR (n = 34, 20.4 %), IFIH1 (n = 19, 11.4 %), and RNU7-1 (n = 3, 1.8 %). The median age of systemic onset was 0.15 years [IQR = 0.67 years; median range by genotype: 0 (TREX1) - 0.62 (ADAR) years], while the median neurological onset was 0.33 years [IQR = 0.82 years; median range by genotype: 0.08 (TREX1) - 0.90 (ADAR) year]. The most common early systemic complications were gastrointestinal, including dysphagia or feeding intolerance (n = 124) and liver abnormalities (n = 67). Among postnatal complications, thrombocytopenia appeared earliest (n = 29, median 0.06 years). Tone abnormalities (axial hypotonia: n = 145, 86.8 %; dystonia: n = 123, 73.7 %), irritability (n = 115, 68.9 %), and gross motor delay (n = 112, 7.1 %) emerged as the most prevalent neurological symptoms. Previously published case reports demonstrated similar patterns. The median AGS score for the entire cohort was 4 (IQR = 7). The most severe neurologic phenotype occurred in TREX1-related AGS (n = 19, median AGS severity score 2, IQR = 2). Time to feeding tube placement, chilblains, early gross motor delay, early cognitive delay, and motor regression were significantly associated with genotype (Fleming-Harrington log-rank: p = 0.0002, p < 0.0001, p = 0.0038, p < 0.0001, p = 0.0001, respectively). Microcephaly, feeding tube placement, and seizures were associated with lower AGS scores (All: Wilcoxon rank sum test, p < 0.0001). Among the qualifying case reports (n = 129), tone abnormalities were the most prevalent disease feature, with spastic quadriplegia reported in 37 of 96 cases (38.5 %) and dystonia in 30 of 96 cases (31.2 %). CONCLUSIONS AGS is a heterogeneous disease with multi-organ system dysfunction that compounds throughout the clinical course, resulting in profound neurological and extra-neurological disease impact. Systemic symptoms precede neurologic disease features in most cases. Disease onset before the age of one year, microcephaly, feeding tube placement, and seizures were associated with worse neurological outcomes. This work will inform evidence-based clinical monitoring guidelines and clinical trial design.
Collapse
Affiliation(s)
| | - Amanda K Jan
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nicholson Modesti
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sarah Woidill
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Francesco Gavazzi
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - David Isaacs
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Russell D'Aiello
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anjana Sevagamoorthy
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lauren Charlton
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amy Pizzino
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Johanna Schmidt
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Keith van Haren
- Department of Neurology, Stanford University, Stanford, CA, USA
| | - Stephanie Keller
- Division of Pediatric Neurology, Emory University, Atlanta, GA, USA
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Lisa T Emrick
- Division of Neurology and Developmental Neuroscience and Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Jamie L Fraser
- Rare Disease Institute, Division of Genetics and Metabolism, Children's National Hospital, Washington, DC, USA
| | - Justine Shults
- Department of Statistics, University of Pennsylvania, Philadelphia, PA, USA
| | - Adeline Vanderver
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura A Adang
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Mendonça LO, Frémond ML. Interferonopathies: From concept to clinical practice. Best Pract Res Clin Rheumatol 2024; 38:101975. [PMID: 39122631 DOI: 10.1016/j.berh.2024.101975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/12/2024] [Accepted: 07/08/2024] [Indexed: 08/12/2024]
Abstract
The horror autoinflammaticus derived from aberrant type I interferon secretion determines a special group of autoinflammatory diseases named interferonopathies. Diverse mechanisms involved in nucleic acids sensing, metabolizing or the lack of interferon signaling retro-control are responsible for the phenotypes associated to Aicardi-Goutières Syndrome (AGS), Proteasome-Associated Autoinflammatory Diseases (PRAAS), STING-Associated Vasculopathy with Infancy Onset (SAVI) and certain forms of monogenic Systemic lupus erythematosus (SLE). This review approaches interferonopathies from the basic immunogenetic concept to diagnosis and treatment.
Collapse
Affiliation(s)
- Leonardo Oliveira Mendonça
- Division of Clinical Immunology and Allergy, School of Medicine, University of São Paulo, São Paulo, Brazil; Discipline of Clinical Immunology and Allergy, Department of Internal Medicine, Universidade de Santo Amaro (UNISA), São Paulo, Brazil.
| | - Marie-Louise Frémond
- Department of Paediatric Hematology-Immunology and Rheumatology, Necker-Enfants Malades Hospital, AP-HP, Paris, France; Laboratory of Neurogenetics and Neuroinflammation Imagine Institute, INSERM UMR1163, Paris, France
| |
Collapse
|
11
|
Adang LA, Groeschel S, Grzyb C, D'Aiello R, Gavazzi F, Sherbini O, Bronner N, Patel A, Vincent A, Sevagamoorthy A, Mutua S, Muirhead K, Schmidt J, Pizzino A, Yu E, Jin D, Eichler F, Fraser JL, Emrick L, Van Haren K, Boulanger JM, Ruzhnikov M, Sylvain M, Nguyen CTÉ, Potic A, Keller S, Fatemi A, Uebergang E, Poe M, Yazdani PA, Bernat J, Lindstrom K, Bonkowsky JL, Bernard G, Stutterd CA, Orchard P, Gupta AO, Ljungberg M, Groenborg S, Zambon A, Locatelli S, Fumagalli F, Elguen S, Kehrer C, Krägeloh-Mann I, Shults J, Vanderver A, Escolar ML. Developmental delay can precede neurologic regression in early onset metachromatic leukodystrophy. Mol Genet Metab 2024; 142:108521. [PMID: 38964050 PMCID: PMC11348664 DOI: 10.1016/j.ymgme.2024.108521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/19/2024] [Accepted: 06/24/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE Metachromatic leukodystrophy (MLD) is a rare neurodegenerative disorder. Emerging therapies are most effective in the presymptomatic phase, and thus defining this window is critical. We hypothesize that early development delay may precede developmental plateau. With the advent of presymptomatic screening platforms and transformative therapies, it is essential to define the onset of neurologic disease. METHODS The specific ages of gain and loss of developmental milestones were captured from the medical records of individuals affected by MLD. Milestone acquisition was characterized as: on target (obtained before the age limit of 90th percentile plus 2 standard deviations compared to a normative dataset), delayed (obtained after 90th percentile plus 2 standard deviations), or plateau (skills never gained). Regression was defined as the age at which skills were lost. LI-MLD was defined by age at onset before 2.5 years. RESULTS Across an international cohort, 351 subjects were included (n = 194 LI-MLD subcohort). The median age at presentation of the LI-MLD cohort was 1.4 years (25th-75th %ile: 1.0-1.5). Within the LI-MLD cohort, 75/194 (39%) had developmental delay (or plateau) prior to MLD clinical presentation. Among the LI-MLD cohort with a minimum of 1.5 years of follow-up (n = 187), 73 (39.0%) subjects never attained independent ambulation. Within LI-MLD + delay subcohort, the median time between first missed milestone target to MLD decline was 0.60 years (maximum distance from delay to onset: 1.9 years). INTERPRETATION Early developmental delay precedes regression in a subset of children affected by LI-MLD, defining the onset of neurologic dysfunction earlier than previously appreciated. The use of realworld data prior to diagnosis revealed an early deviation from typical development. Close monitoring for early developmental delay in presymptomatic individuals may help in earlier diagnosis with important consequences for treatment decisions.
Collapse
Affiliation(s)
- Laura Ann Adang
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | - Samuel Groeschel
- University Children's Hospital, Hoppe-Seyler-Str.1, Tuebingen, DE 72070, USA.
| | - Chloe Grzyb
- University of Pittsburgh, Department of Pediatrics, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| | - Russell D'Aiello
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | - Francesco Gavazzi
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | - Omar Sherbini
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | - Nowa Bronner
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA; Uniformed Services University of Health Sciences, Bethesda, MD, USA
| | - Akshilkumar Patel
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA; Penn State Health Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Ariel Vincent
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | | | - Sylvia Mutua
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | - Kayla Muirhead
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA
| | - Johanna Schmidt
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | - Amy Pizzino
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | - Emily Yu
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | | | | | - Jamie L Fraser
- Children's National Medical Center, Washington, DC 20010-2978, USA.
| | - Lisa Emrick
- Baylor College of Medicine, Houston, TX 77030-3411, USA.
| | | | | | | | - Michel Sylvain
- Université Laval, Division of Pediatric Neurology, Centre Mère-Enfant Soleil, Quebec, QC, Canada
| | - Cam-Tu Émilie Nguyen
- Centre Hospitalier Universitaire Sainte-Justine, Department of Neurosciences and Pediatrics, Division of Pediatric Neurology, Montreal, QC, Canada.
| | - Ana Potic
- University of Belgrade Faculty of Medicine, Department of Neurology, Clinic for Child Neurology and Psychiatry, Beograd, Republic of Serbia
| | - Stephanie Keller
- Children's Healthcare of Atlanta Scottish Rite Hospital, Atlanta, GA, USA.
| | - Ali Fatemi
- Kennedy Krieger Institute and Department of Neurology, Johns Hopkins University, Baltimore, MD, USA.
| | - Eloise Uebergang
- Royal Children's Hospital, Murdoch Children's Research Institute and University of Melbourne, Department of Paediatrics, Melbourne, VIC, Australia.
| | - Michele Poe
- University of Pittsburgh, Department of Pediatrics, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| | - Pouneh Amir Yazdani
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| | - John Bernat
- University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
| | - Kristen Lindstrom
- BioMarin Pharmaceutical Inc, Novato, CA, USA; Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Joshua L Bonkowsky
- University of Utah School of Medicine, Division of Pediatric Neurology, Department of Pediatrics, 295 Chipeta Way, Williams Building, Salt Lake City, UT 84108, USA.
| | - Genevieve Bernard
- Montreal Children's Hospital, McGill University Health Center, Departments of Pediatrics, Neurology and Neurosurgery, 2300 Tupper, Room A-506, Montreal, QC H3H 1P3, Canada.
| | - Chloe A Stutterd
- Royal Children's Hospital, Murdoch Children's Research Institute and University of Melbourne, Department of Paediatrics, Melbourne, VIC, Australia.
| | | | | | - Merete Ljungberg
- Copenhagen University Hospital, Centre Inherited Metabolic Disease, Department of Pediatrics and Adolescent Medicine, Kobenhavn, Denmark.
| | - Sabine Groenborg
- Copenhagen University Hospital, Centre Inherited Metabolic Disease, Department of Pediatrics and Adolescent Medicine, Kobenhavn, Denmark.
| | - Alberto Zambon
- San Raffaele Scientific Institute, Division of Neuroscience, Milan, Italy.
| | - Sara Locatelli
- San Raffaele Hospital, Paediatric Immunohematology and Unit of Neurology, Milano, Lombardia, Italy.
| | | | - Saskia Elguen
- University Children's Hospital, Hoppe-Seyler-Str.1, Tuebingen, DE 72070, USA.
| | - Christiane Kehrer
- University Children's Hospital, Hoppe-Seyler-Str.1, Tuebingen, DE 72070, USA.
| | | | - Justine Shults
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Adeline Vanderver
- The Children's Hospital of Philadelphia, Neurology, Philadelphia, PA, USA.
| | - Maria L Escolar
- University of Pittsburgh, Department of Pediatrics, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| |
Collapse
|
12
|
Gavazzi F, Vaia Y, Woidill S, Formanowski B, Peixoto de Barcelos I, Sevagamoorthy A, Modesti NB, Charlton L, Cusack SV, Vincent A, D'Aiello R, Jawad A, Galli J, Varesio C, Fazzi E, Orcesi S, Glanzman AM, Lorch S, DeMauro SB, Guez-Barber D, Waldman AT, Vanderver A, Adang LA. Nonverbal Cognitive Skills in Children With Aicardi Goutières Syndrome. Neurology 2024; 103:e209541. [PMID: 38857477 PMCID: PMC11226315 DOI: 10.1212/wnl.0000000000209541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/10/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Aicardi Goutières syndrome (AGS) is type I interferonopathy characterized by severe neurologic impairment. Although many children with AGS demonstrate motor and expressive language deficits, the magnitude of receptive language impairment is uncharacterized. We sought to characterize cognitive function in AGS-affected children using assessment tools with reduced dependence on motor abilities and compare cognitive testing outcomes with overall severity and parental assessment of adaptive behavior. METHODS We performed a cross-sectional study. Children were recruited as part of the Myelin Disorders Biorepository Project at the Children's Hospital of Philadelphia. We included individuals with a confirmed diagnosis of AGS. We administered the Leiter International Performance Scale, third edition (Leiter-3), and the Vineland Adaptive Behavior Scale, third edition (VABS-3), in the context of research encounters. Motor skills were categorized by AGS Severity Scale mobility levels. Descriptive statistics and Spearman's rank correlation were used to compare assessments. Mann-Whitney and Kruskal-Wallis tests with correction with Dunn's multiple comparison test were used to compare test performance between mobility groups. RESULTS Cognitive and adaptive behavior performance was captured in 57 children. The mean age at encounters was 8.51 (SD 5.15) years. The median (IQR) Leiter-3 score was 51 (interquartile range [IQR] 60), with administration failure in 20 of 57 (35%) individuals. On the VABS-3, the Motor Domain (median 29, IQR 36.25) was more impacted than the Communication (median 50, IQR 52), Daily Living Skills (median 52, IQR 31), and Socialization (median 54, IQR 40) Domains (p < 0.0001). The AGS Scale correlated with VABS-3 (r = 0.86, p < 0.0001) and Leiter-3 (r = 0.87, p < 0.0001). There was correlation between VABS-3 Domains and Leiter-3 (r-range 0.83-0.97). Gross motor and fine motor categories, respectively, correlated with VABS-3 (H = 39.37, p < 0.0001; U = 63, p < 0.0001) and Leiter-3 (H = 40.43, p < 0.0001; U = 66, p < 0.0001). Within each gross motor and fine motor category of the AGS Scale, a subset of children scored within normal IQ range. DISCUSSION Parental assessment of function by the VABS-3 correlated with directly assessed performance measures. Our data underscore the potential value of VABS-3 and Leiter-3 as tools to assess psychometric function in AGS. With a deeper understanding of our patients' abilities, we can better guide clinicians and families to provide appropriate support and personalized interventions to empower children with leukodystrophies to maximize their communication and educational potential.
Collapse
Affiliation(s)
- Francesco Gavazzi
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ylenia Vaia
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Sarah Woidill
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Brielle Formanowski
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Isabella Peixoto de Barcelos
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Anjana Sevagamoorthy
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Nicholson B Modesti
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Lauren Charlton
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Stacy V Cusack
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ariel Vincent
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Russell D'Aiello
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Abbas Jawad
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jessica Galli
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Costanza Varesio
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Elisa Fazzi
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Simona Orcesi
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Allan M Glanzman
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Scott Lorch
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Sara B DeMauro
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Danielle Guez-Barber
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Amy T Waldman
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Adeline Vanderver
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Laura A Adang
- From the Divisions of Neurology (F.G., Y.V., S.W., I.P.d.B., A.S., N.B.M., L.C., D.G.-B., A.T.W., A. Vanderver, L.A.A.), and Neonatology (B.F., S.L., S.B.D.), and Departments of Occupational Therapy (S.V.C., A. Vincent), Biomedical & Health Informatics (R.D.A.), Pediatrics (A.J.), and Physical Therapy (A.M.G.), Children's Hospital of Philadelphia, PA; University of Milan (Y.V.); Department of Clinical and Experimental Sciences (J.G., E.F.), University of Brescia; Unit of Child Neurology and Psychiatry (J.G., E.F.), ASST Spedali Civili of Brescia; Unit of Child Neurology and Psychiatry (C.V., S.O.), Istituto di Ricovero e Cura a Carattere Scientifico Mondino Foundation, Pavia; Department of Brain and Behavioral Sciences (S.O.), University of Pavia, Italy; and Department of Neurology (D.G.-B., A.T.W., A. Vanderver, L.A.A.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
13
|
Liu X, Lei M, Xue Y, Li H, Yin J, Li D, Shu J, Cai C. Multi-dimensional Insight into the Coexistence of Pathogenic Genes for ADAR1 and TSC2: Careful Consideration is Essential for Interpretation of ADAR1 Variants. Biochem Genet 2024; 62:1811-1826. [PMID: 37740860 DOI: 10.1007/s10528-023-10488-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/06/2023] [Indexed: 09/25/2023]
Abstract
Aicardi-Goutières syndrome 6 (AGS6) is a serious auto-immunization-associated acute neurologic decompensation. AGS6 manifests as acute onset of severe generalized dystonia of limbs and developmental regression secondary to febrile illness mostly. Dyschromatosis symmetrica hereditaria (DSH), as pigmentary genodermatosis, is a characterized mixture of hyperpigmented and hypopigmented macules. Both AGS6 and DSH are associated with ADAR1 pathogenic variants. To explore the etiology of a proband with developmental regression with mixture of hyperpigmentation and hypopigmentation macules, we used the trio-WES. Later, to clarify the association between variants and diseases, we used guidelines of ACMG for variants interpretation and quantitative Real-time PCR for verifying elevated expression levels of interferon-stimulated genes, separately. By WES, we detected 2 variants in ADAR1 and a variant in TSC2, respectively, were NM_001111.5:c.1096_1097del, NM_001111.5:c.518A>G, and NM_000548.5:c.1864C>T. Variants interpretation suggested that these 3 variants were both pathogenic. Expression levels of interferon-stimulated genes also elevated as expected. We verified the co-occurrence of pathogenic variants of ADAR1 and TSC2 in AGS6 patients with DSH. Our works contributed to the elucidation of ADAR1 pathogenic mechanism, given the specific pathogenic mechanism of ADAR1, and it is necessary to consider with caution when variants were found in ADAR1.
Collapse
Affiliation(s)
- Xiangyu Liu
- Graduate College of Tianjin Medical University, Tianjin, 300070, China
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, 300134, China
| | - Meifang Lei
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, 300134, China
- Department of Neurology, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China
| | - Yan Xue
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, 300134, China
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Tianjin University Children's Hospital), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China
| | - Hong Li
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, 300134, China
- Department of Neurology, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China
| | - Jing Yin
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, 300134, China
- Department of Immunology, Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, 300134, China
| | - Dong Li
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, 300134, China.
- Department of Neurology, Tianjin Children's Hospital (Tianjin University Children's Hospital), No. 238 Longyan Road, Beichen District, Tianjin, 300134, China.
| | - Jianbo Shu
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, 300134, China.
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Tianjin University Children's Hospital), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China.
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, 300134, China.
| | - Chunquan Cai
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, 300134, China.
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Tianjin University Children's Hospital), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China.
- Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, 300134, China.
| |
Collapse
|
14
|
de Barcelos IP, Woidill S, Gavazzi F, Modesti NB, Sevagamoorthy A, Vanderver A, Adang L. Systematic analysis of genotype-phenotype variability in siblings with Aicardi Goutières Syndrome (AGS). Mol Genet Metab 2024; 142:108346. [PMID: 38368708 PMCID: PMC11431181 DOI: 10.1016/j.ymgme.2024.108346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/11/2023] [Accepted: 02/11/2024] [Indexed: 02/20/2024]
Abstract
OBJECTIVE Aicardi Goutières Syndrome (AGS) is a genetic interferonopathy associated with multisystemic heterogeneous disease and neurologic dysfunction. AGS includes a broad phenotypic spectrum which is only partially explained by genotype. To better characterize this variability, we will perform a systematic analysis of phenotypic variability in familial cases of AGS. METHODS Among thirteen families, twenty-six siblings diagnosed with AGS were identified from the Myelin Disorders and Biorepository Project (MDBP) at the Children's Hospital of Philadelphia. Data were collected on the age of onset, genotype, neurologic impairment, and systemic complications. Neurologic impairment was assessed by a disease-specific scale (AGS Severity Scale) at the last available clinical encounter (range: 0-11 representing severe - attenuated phenotypes). The concordance of clinical severity within sibling pairs was categorized based on the difference in AGS Scale (discordant defined as >2-unit difference). The severity classifications were compared between sibling sets and by genotype. RESULTS Five genotypes were represented: TREX1 (n = 4 subjects), RNASEH2B (n = 8), SAMHD1 (n = 8) ADAR1 (n = 4), and IFIH1 (n = 2). The older sibling was diagnosed later relative to the younger affected sibling (median age 7.32 years [IQR = 14.1] compared to 1.54 years [IQR = 10.3]). Common presenting neurologic symptoms were tone abnormalities (n = 10/26) and gross motor dysfunction (n = 9/26). Common early systemic complications included dysphagia and chilblains. The overall cohort median AGS severity score at the last encounter was 8, while subjects presenting with symptoms before one year had a median score of 5. The TREX1 cohort presented at the youngest age and with the most severe phenotype on average. AGS scores were discordant for 5 of 13 sibling pairs, most commonly in the SAMHD1 pairs. Microcephaly, feeding tube placement, seizures and earlier onset sibling were associated with lower AGS scores (respectively, Wilcoxon rank sum: p = 0.0001, p < 0.0001, p = 0.0426, and Wilcoxon signed rank: p = 0.0239). CONCLUSIONS In this systematic analysis of phenotypic variability in familial cases, we found discordance between siblings affected by AGS. Our results underscore the heterogeneity of AGS and suggest factors beyond AGS genotype may affect phenotype. Understanding the critical variables associated with disease onset and severity can guide future therapeutic interventions and clinical monitoring. This report reinforces the need for further studies to uncover potential factors to better understand this phenotypic variability, and consequently identify potential targets for interventions in attempt to change the natural history of the disease.
Collapse
Affiliation(s)
| | - Sarah Woidill
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Francesco Gavazzi
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nicholson B Modesti
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anjana Sevagamoorthy
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adeline Vanderver
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, usa
| | - Laura Adang
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, usa.
| |
Collapse
|
15
|
Adang LA, Sevagamoorthy A, Sherbini O, Fraser JL, Bonkowsky JL, Gavazzi F, D'Aiello R, Modesti NB, Yu E, Mutua S, Kotes E, Shults J, Vincent A, Emrick LT, Keller S, Van Haren KP, Woidill S, Barcelos I, Pizzino A, Schmidt JL, Eichler F, Fatemi A, Vanderver A. Longitudinal natural history studies based on real-world data in rare diseases: Opportunity and a novel approach. Mol Genet Metab 2024; 142:108453. [PMID: 38522179 PMCID: PMC11131438 DOI: 10.1016/j.ymgme.2024.108453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/13/2024] [Accepted: 03/16/2024] [Indexed: 03/26/2024]
Abstract
Growing interest in therapeutic development for rare diseases necessitate a systematic approach to the collection and curation of natural history data that can be applied consistently across this group of heterogenous rare diseases. In this study, we discuss the challenges facing natural history studies for leukodystrophies and detail a novel standardized approach to creating a longitudinal natural history study using existing medical records. Prospective studies are uniquely challenging for rare diseases. Delays in diagnosis and overall rarity limit the timely collection of natural history data. When feasible, prospective studies are often cross-sectional rather than longitudinal and are unlikely to capture pre- or early- symptomatic disease trajectories, limiting their utility in characterizing the full natural history of the disease. Therapeutic development in leukodystrophies is subject to these same obstacles. The Global Leukodystrophy Initiative Clinical Trials Network (GLIA-CTN) comprises of a network of research institutions across the United States, supported by a multi-center biorepository protocol, to map the longitudinal clinical course of disease across leukodystrophies. As part of GLIA-CTN, we developed Standard Operating Procedures (SOPs) that delineated all study processes related to staff training, source documentation, and data sharing. Additionally, the SOP detailed the standardized approach to data extraction including diagnosis, clinical presentation, and medical events, such as age at gastrostomy tube placement. The key variables for extraction were selected through face validity, and common electronic case report forms (eCRF) across leukodystrophies were created to collect analyzable data. To enhance the depth of the data, clinical notes are extracted into "original" and "imputed" encounters, with imputed encounter referring to a historic event (e.g., loss of ambulation 3 months prior). Retrospective Functional Assessments were assigned by child neurologists, using a blinded dual-rater approach and score discrepancies were adjudicated by a third rater. Upon completion of extraction, data source verification is performed. Data missingness was evaluated using statistics. The proposed methodology will enable us to leverage existing medical records to address the persistent gap in natural history data within this unique disease group, allow for assessment of clinical trajectory both pre- and post-formal diagnosis, and promote recruitment of larger cohorts.
Collapse
Affiliation(s)
- Laura Ann Adang
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Anjana Sevagamoorthy
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Omar Sherbini
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jamie L Fraser
- Rare Disease Institute, Children's National Medical Center, Washington, DC, USA; Leukodystrophy and Myelin Disorders Program, Children's National Medical Center, Washington, DC, USA
| | - Joshua L Bonkowsky
- Division of Pediatric Neurology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Personalized Medicine, Primary Children's Hospital, Salt Lake City, UT, USA
| | - Francesco Gavazzi
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Russel D'Aiello
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nicholson B Modesti
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emily Yu
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sylvia Mutua
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emma Kotes
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Justine Shults
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ariel Vincent
- CHOP Research Institute, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lisa T Emrick
- Division of Neurology and Developmental Neuroscience in Department Pediatrics, Baylor College Medicine and Texas Children's Hospital, Houston, TX, USA; Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Stephanie Keller
- Children's Healthcare of Atlanta Scottish Rite Hospital, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Sarah Woidill
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Isabella Barcelos
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amy Pizzino
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Johanna L Schmidt
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Ali Fatemi
- Moser Center for Leukodystrophies, Kennedy Krieger Institute, Baltimore, MD, USA; Departments of Neurology & Pediatrics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Adeline Vanderver
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
16
|
Cusack SV, Gavazzi F, Peixoto de Barcelos I, Modesti NB, Woidill S, Formanowski B, DeMauro SB, Lorch S, Vincent A, Jawad AF, Estilow T, Glanzman AM, Vanderver A, Adang LA. Characterization of Fine Motor and Visual Motor Skills in Aicardi-Goutières Syndrome. J Child Neurol 2024; 39:147-154. [PMID: 38532733 PMCID: PMC11098691 DOI: 10.1177/08830738241241786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Aicardi-Goutières syndrome is a genetic inflammatory disorder resulting in dispersed neurologic dysfunction. Despite a recognition of overall motor impairment, fine and visual motor skills are undercharacterized. We hypothesize that there is a spectrum of fine and visual motor skills in the Aicardi-Goutières syndrome population as captured by a standard outcome measure, the Peabody Developmental Motor Scales (PDMS-2), which will be proportional to overall disease severity.In a cohort of 74 subjects, the Peabody Developmental Motor Scales-2 grasping and visual-motor integration subtests were administered concurrently with the Aicardi-Goutières syndrome Severity Scale (severe [range 0-3], moderate [range 4-8], and attenuated [range 9-11]). The cohort was also compared by genotype and performance as defined by raw scores. The distribution of Peabody Developmental Motor Scales-2 scores within a genotype was assessed by interquartile ranges (IQRs).Peabody Developmental Motor Scales-2 grasping and visual-motor integration performance was the least variable in the TREX1-cohort (IQR: 10.00-12.00) versus the SAMHD1 and IFIH1 cohorts (IQR: 51.00-132.00 and 48.50-134.00, respectively). Neurologic severity highly correlated with both fine and visual motor skills (Spearman correlation: r = 0.87, 0.91, respectively). A floor effect (lowest 10% of possible scores) was observed within the severe cohort (n = 32/35), whereas a ceiling effect (top 10%) was observed in the attenuated cohort (n = 13/17).This study characterized the spectrum of fine and visual motor function in the Aicardi-Goutières syndrome population, which correlated with overall neurologic dysfunction. The Peabody Developmental Motor Scales-2 grasping and visual-motor integration showed promise as potential assessment tools in moderate and attenuated Aicardi-Goutières syndrome cohorts. A better understanding of fine and visual motor function in this population will benefit clinical care and clinical trial design.
Collapse
Affiliation(s)
- Stacy V. Cusack
- Department of Occupational Therapy, Children’s Hospital of Philadelphia
| | - Francesco Gavazzi
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Nicholson B. Modesti
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sarah Woidill
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brielle Formanowski
- Department of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sara B. DeMauro
- Department of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Scott Lorch
- Department of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ariel Vincent
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Abbas F. Jawad
- Division of General Peds, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Timothy Estilow
- Department of Occupational Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Allan M. Glanzman
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adeline Vanderver
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics , Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Laura A. Adang
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
17
|
Joung J, Gallison K, Sollee JJ, Vigilante N, Cooper H, Liu GW, Ballester L, Faig W, Waldman AT. Acquisition and Loss of Developmental Milestones and Time to Disease-Related Outcomes in Cerebral Alexander Disease. J Child Neurol 2023; 38:672-678. [PMID: 37920915 DOI: 10.1177/08830738231210040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Objective: To determine the ages at acquisition of developmental milestones, loss of motor function, and clinical symptoms in Alexander disease. Methods: Patients with confirmed cerebral Alexander disease were included. Data abstraction of developmental and disease-specific milestones was performed from medical records, physical exams, and questionnaires. Mixed effects logistic regression was used to determine if key clinical features were associated with milestone achievement, controlling for patient age. Results: 51 patients with cerebral/infantile Alexander disease were evaluated at a mean age of 10.96 years (range 2.29-31.08 years). Developmental milestones in Alexander disease were often achieved but delayed. Ambulation was achieved in 44 subjects (86%); 34 (67%) subjects walked independently (mean age 1.9 years, range 0.91-3.25 years) and an additional 10 (20%) subjects walked with assistance (mean age 3.9 years, range 1.8-8 years) but did not progress to independent ambulation. Developmental delay was the earliest and most prevalent symptom (N = 48 [94%], mean age 0.58 years), compared to an initial seizure (N = 41 [80%], mean age 2.80 years), and macrocephaly (N = 28 [55%], mean age 4.04 years), P < .0001 between these ages of onset. Loss of independent ambulation occurred in 11 of the 34 (32%) children who had acquired ambulation (range 3.41-15.10 years). Presence of seizures or macrocephaly did not predict the achievement or loss of ambulation. Conclusions: The clinical triad of developmental delay, seizures, and macrocephaly are not universally present in cerebral Alexander disease. Clinicians should have a high index of suspicion for Alexander disease in patients with mild delays and a first seizure.
Collapse
Affiliation(s)
- Joshua Joung
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kathryn Gallison
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - John Jack Sollee
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nicholas Vigilante
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hannah Cooper
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Geraldine W Liu
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lance Ballester
- Biostatistics and Data Management Core, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Walter Faig
- Biostatistics and Data Management Core, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amy T Waldman
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Departments of Neurology and Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
18
|
Świerczyńska M, Tronina A, Filipek E. Aicardi-Goutières Syndrome with Congenital Glaucoma Caused by Novel TREX1 Mutation. J Pers Med 2023; 13:1609. [PMID: 38003924 PMCID: PMC10672266 DOI: 10.3390/jpm13111609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Aicardi-Goutières syndrome (AGS) is a rare genetic disorder characterized by microcephaly, white matter lesions, numerous intracranial calcifications, chilblain skin lesions and high levels of interferon-α (IFN-α) in the cerebrospinal fluid (CSF). However, ocular involvement is reported significantly less frequently. CASE PRESENTATION We present a case of a neonate with hypotrophy, microcephaly, frostbite-like skin lesions, thrombocytopenia, elevated liver enzymes and hepatosplenomegaly. Magnetic resonance imaging (MRI) of the brain showed multiple foci of calcification, white matter changes, cerebral atrophy, and atrophic dilatation of the ventricular system. The inflammatory parameters were not elevated, and the infectious etiology was excluded. Instead, elevated levels of IFN-α in the serum were detected. Based on the related clinical symptoms, imaging and test findings, the diagnosis of AGS was suspected. Genetic testing revealed two pathogenic mutations, c.490C>T and c.222del (novel mutation), in the three prime repair exonuclease 1 (TREX1) gene, confirming AGS type 1 (AGS1). An ophthalmologic examination of the child at 10 months of age revealed an impaired pupillary response to light, a corneal haze with Haab lines in the right eye (RE), pale optic nerve discs and neuropathy in both eyes (OU). The intraocular pressure (IOP) was 51 mmHg in the RE and 49 in the left eye (LE). The flash visual evoked potential (FVEP) showed prolonged P2 latencies of up to 125% in the LE and reduced amplitudes of up to approximately 10% OU. This girl was diagnosed with congenital glaucoma, and it was managed with a trabeculectomy with a basal iridectomy of OU, resulting in a reduction and stabilization in the IOP to 12 mmHg in the RE and 10 mmHg in the LE without any hypotensive eyedrops. CONCLUSIONS We present the clinical characteristics, electrophysiological and imaging findings, as well as the genetic test results of a patient with AGS1. Our case contributes to the extended ophthalmic involvement of the pathogenic c.490C>T and c.222del mutations in TREX1.
Collapse
Affiliation(s)
- Marta Świerczyńska
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-514 Katowice, Poland
- Department of Ophthalmology, Kornel Gibiński University Clinical Center, Medical University of Silesia, 40-514 Katowice, Poland
| | - Agnieszka Tronina
- Department of Pediatric Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-514 Katowice, Poland; (A.T.); (E.F.)
- Department of Pediatric Ophthalmology, Kornel Gibiński University Clinical Center, Medical University of Silesia, 40-514 Katowice, Poland
| | - Erita Filipek
- Department of Pediatric Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-514 Katowice, Poland; (A.T.); (E.F.)
- Department of Pediatric Ophthalmology, Kornel Gibiński University Clinical Center, Medical University of Silesia, 40-514 Katowice, Poland
| |
Collapse
|
19
|
Liu A, Ying S. Aicardi-Goutières syndrome: A monogenic type I interferonopathy. Scand J Immunol 2023; 98:e13314. [PMID: 37515439 DOI: 10.1111/sji.13314] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/26/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023]
Abstract
Aicardi-Goutières syndrome (AGS) is a rare monogenic autoimmune disease that primarily affects the brains of children patients. Its main clinical features include encephalatrophy, basal ganglia calcification, leukoencephalopathy, lymphocytosis and increased interferon-α (IFN-α) levels in the patient's cerebrospinal fluid (CSF) and serum. AGS may be caused by mutations in any one of nine genes (TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR1, IFIH1, LSM11 and RNU7-1) that result in accumulation of self-nucleic acids in the cytoplasm or aberrant sensing of self-nucleic acids. This triggers overproduction of type I interferons (IFNs) and subsequently causes AGS, the prototype of type I interferonopathies. This review describes the discovery history of AGS with various genotypes and provides the latest knowledge of clinical manifestations and causative genes of AGS. The relationship between AGS and type I interferonopathy and potential therapeutic methods for AGS are also discussed in this review.
Collapse
Affiliation(s)
- Anran Liu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
- First School of Clinical Medicine, Anhui Medical University, Hefei, China
| | - Songcheng Ying
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
20
|
Natoli V, Charras A, Hahn G, Hedrich CM. Neuropsychiatric involvement in juvenile-onset systemic lupus erythematosus (jSLE). Mol Cell Pediatr 2023; 10:5. [PMID: 37556020 PMCID: PMC10412509 DOI: 10.1186/s40348-023-00161-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/26/2023] [Indexed: 08/10/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a rare autoimmune/inflammatory disease with significant morbidity and mortality. Approximately 15-20% of SLE patients develop the disease during childhood or adolescence (juvenile-onset SLE/jSLE). Patients with jSLE exhibit more variable and severe disease when compared to patients with disease-onset during adulthood. Neuropsychiatric (NP) involvement is a clinically heterogenous and potentially severe complication. Published reports on the incidence and prevalence of NP-jSLE are scarce, and the exact pathophysiology is poorly understood.This manuscript provides a review of the existing literature, suggesting NP involvement in 13.5-51% of jSLE patients. Among patients with NP-jSLE affecting the CNS, we propose two main subgroups: (i) a chronic progressive, predominantly type 1 interferon-driven form that poorly responds to currently used treatments, and (ii) an acutely aggressive form that usually presents early during the disease that may be primarily mediated by auto-reactive effector lymphocytes. While this hypothesis requires to be tested in large collaborative international cohort studies, it may offer future patient stratification and individualised care.
Collapse
Affiliation(s)
- Valentina Natoli
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Rheumatology, Institute in the Park, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
- Dipartimento di Neuroscienze, Riabilitazione, Oftalmologia, Genetica e Scienze Materno-Infantili, Università degli Studi di Genova, Genoa, Italy
| | - Amandine Charras
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Gabriele Hahn
- Department of Radiology, Universitätsklinikum Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Christian M Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.
- Department of Rheumatology, Institute in the Park, Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
21
|
Frémond ML, Hully M, Fournier B, Barrois R, Lévy R, Aubart M, Castelle M, Chabalier D, Gins C, Sarda E, Al Adba B, Couderc S, D' Almeida C, Berat CM, Durrleman C, Espil C, Lambert L, Méni C, Périvier M, Pillet P, Polivka L, Schiff M, Todosi C, Uettwiller F, Lepelley A, Rice GI, Seabra L, Sanquer S, Hulin A, Pressiat C, Goldwirt L, Bondet V, Duffy D, Moshous D, Bader-Meunier B, Bodemer C, Robin-Renaldo F, Boddaert N, Blanche S, Desguerre I, Crow YJ, Neven B. JAK Inhibition in Aicardi-Goutières Syndrome: a Monocentric Multidisciplinary Real-World Approach Study. J Clin Immunol 2023; 43:1436-1447. [PMID: 37171742 PMCID: PMC10175907 DOI: 10.1007/s10875-023-01500-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/19/2023] [Indexed: 05/13/2023]
Abstract
The paradigm type I interferonopathy Aicardi-Goutières syndrome (AGS) is most typically characterized by severe neurological involvement. AGS is considered an immune-mediated disease, poorly responsive to conventional immunosuppression. Premised on a chronic enhancement of type I interferon signaling, JAK1/2 inhibition has been trialed in AGS, with clear improvements in cutaneous and systemic disease manifestations. Contrastingly, treatment efficacy at the level of the neurological system has been less conclusive. Here, we report our real-word approach study of JAK1/2 inhibition in 11 patients with AGS, providing extensive assessments of clinical and radiological status; interferon signaling, including in cerebrospinal fluid (CSF); and drug concentrations in blood and CSF. Over a median follow-up of 17 months, we observed a clear benefit of JAK1/2 inhibition on certain systemic features of AGS, and reproduced results reported using the AGS neurologic severity scale. In contrast, there was no change in other scales assessing neurological status; using the caregiver scale, only patient comfort, but no other domain of everyday-life care, was improved. Serious bacterial infections occurred in 4 out of the 11 patients. Overall, our data lead us to conclude that other approaches to treatment are urgently required for the neurologic features of AGS. We suggest that earlier diagnosis and adequate central nervous system penetration likely remain the major factors determining the efficacy of therapy in preventing irreversible brain damage, implying the importance of early and rapid genetic testing and the consideration of intrathecal drug delivery.
Collapse
Affiliation(s)
- Marie-Louise Frémond
- Paediatric Haematology-Immunology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris Cité, 149 rue de Sèvres, 75015, Paris, France
- Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, Inserm UMR 1163, Université Paris Cité, 24 boulevard du Montparnasse, 75015, Paris, France
| | - Marie Hully
- Paediatric Neurology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | - Benjamin Fournier
- Paediatric Haematology-Immunology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris Cité, 149 rue de Sèvres, 75015, Paris, France
| | - Rémi Barrois
- Paediatric Neurology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | - Romain Lévy
- Paediatric Haematology-Immunology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris Cité, 149 rue de Sèvres, 75015, Paris, France
| | - Mélodie Aubart
- Paediatric Neurology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | - Martin Castelle
- Paediatric Haematology-Immunology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris Cité, 149 rue de Sèvres, 75015, Paris, France
| | - Delphine Chabalier
- Paediatric Neurology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | - Clarisse Gins
- Paediatric Neurology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | - Eugénie Sarda
- Paediatric Neurology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | - Buthaina Al Adba
- Department of Paediatric Rheumatology, Sidra Medicine, Doha, Qatar
| | - Sophie Couderc
- Neonatal Department, Poissy Saint-Germain Hospital, Poissy, France
| | - Céline D' Almeida
- Paediatrics Department, Castres-Mazamet Intercommunal Hospital, Castres, France
| | - Claire-Marine Berat
- Reference Center of Inherited Metabolic Disorders, Necker Hospital, APHP, Université Paris Cité, 75015, Paris, France
| | - Chloé Durrleman
- Paediatric Neurology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | - Caroline Espil
- Paediatric Neurology Department, Bordeaux University Hospital, Bordeaux, France
| | - Laetitia Lambert
- Genetics Department, Nancy University Hospital, 54000, Nancy, France
| | - Cécile Méni
- Paediatric Dermatology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | | | - Pascal Pillet
- Paediatric Rheumatology Department, Bordeaux University Hospital, Bordeaux, France
| | - Laura Polivka
- Paediatric Dermatology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | - Manuel Schiff
- Reference Center of Inherited Metabolic Disorders, Necker Hospital, APHP, Université Paris Cité, 75015, Paris, France
- Imagine Institute, Inserm UMR 1163, 75015, Paris, France
| | - Calina Todosi
- Paediatric Neurology Unit, Children's Medicine Department, Children's Hospital, Nancy University Hospital, 54000, Nancy, France
| | - Florence Uettwiller
- Paediatric Rheumatology Department, Tours University Hospital, Tours, France
| | - Alice Lepelley
- Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, Inserm UMR 1163, Université Paris Cité, 24 boulevard du Montparnasse, 75015, Paris, France
| | - Gillian I Rice
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Luis Seabra
- Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, Inserm UMR 1163, Université Paris Cité, 24 boulevard du Montparnasse, 75015, Paris, France
| | - Sylvia Sanquer
- Biochemistry, Metabolomics and Proteomics Department, Necker Hospital, AP-HP Centre, Université Paris Cité, 75015, Paris, France
| | - Anne Hulin
- Pharmacology and Toxicology Laboratory, Henri Mondor University Hospital, APHP, 94000, Créteil, France
| | - Claire Pressiat
- Pharmacology and Toxicology Laboratory, Henri Mondor University Hospital, APHP, 94000, Créteil, France
| | - Lauriane Goldwirt
- Pharmacology Department, Saint-Louis University Hospital, APHP, 75010, Paris, France
| | - Vincent Bondet
- Translational Immunology Unit, Institut Pasteur, Université de Paris Cité, F75015, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université de Paris Cité, F75015, Paris, France
| | - Despina Moshous
- Paediatric Haematology-Immunology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris Cité, 149 rue de Sèvres, 75015, Paris, France
- Imagine Institute, Inserm UMR 1163, 75015, Paris, France
| | - Brigitte Bader-Meunier
- Paediatric Haematology-Immunology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris Cité, 149 rue de Sèvres, 75015, Paris, France
| | - Christine Bodemer
- Genetics Department, Nancy University Hospital, 54000, Nancy, France
| | - Florence Robin-Renaldo
- Paediatric Neurology Department, Trousseau Hospital, APHP, Sorbonne Université, 75012, Paris, France
| | - Nathalie Boddaert
- Paediatric Radiology Department, AP-HP, Hôpital Necker Enfants Malades, Université Paris cité, Institut Imagine INSERM U1163 and U1299, 75015, Paris, France
| | - Stéphane Blanche
- Paediatric Haematology-Immunology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris Cité, 149 rue de Sèvres, 75015, Paris, France
| | - Isabelle Desguerre
- Paediatric Neurology Department, Necker Hospital, APHP Centre, Université Paris Cité, 75015, Paris, France
| | - Yanick J Crow
- Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, Inserm UMR 1163, Université Paris Cité, 24 boulevard du Montparnasse, 75015, Paris, France.
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Edinburgh, UK.
| | - Bénédicte Neven
- Paediatric Haematology-Immunology and Rheumatology Unit, Necker Hospital, APHP Centre, Université Paris Cité, 149 rue de Sèvres, 75015, Paris, France.
- Imagine Institute, Laboratory of Immunogenetics of Paediatric Autoimmunity, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France.
| |
Collapse
|
22
|
Galli J, Cattalini M, Loi E, Ferraro RM, Giliani S, Orcesi S, Pinelli L, Badolato R, Fazzi E. Treatment response to Janus kinase inhibitor in a child affected by Aicardi-Goutières syndrome. Clin Case Rep 2023; 11:e7724. [PMID: 37534202 PMCID: PMC10390657 DOI: 10.1002/ccr3.7724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/14/2023] [Accepted: 07/09/2023] [Indexed: 08/04/2023] Open
Abstract
Key Clinical Message Baricitinib, a Janus kinase inhibitor (JAK-inhibitor), seems to contribute to an improvement of a child affected by Aicardi-Goutières syndrome (AGS), reducing the interferon score and determining a recovery of cognitive, communicative, and relational dysfunctions, while the gross motor deficit persisted. Abstract We report the treatment response to baricitinib, a JAK-inhibitor, in a 4-year-old girl affected by Aicardi-Goutières syndrome (AGS2, RNASEH2B mutation). Using quantitative measures, we detected a significant amelioration characterized by a complete recovery of cognitive, communicative, and relational skills after 8 and 16 months from the beginning of therapy.
Collapse
Affiliation(s)
- Jessica Galli
- Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
- Unit of Child Neurology and PsychiatryASST Spedali Civili of BresciaBresciaItaly
| | - Marco Cattalini
- Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
- Pediatrics ClinicASST Spedali Civili of BresciaBresciaItaly
| | - Erika Loi
- Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
| | - Rosalba Monica Ferraro
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- “Angelo Nocivelli” Institute for Molecular Medicine, ASST Spedali Civili of BresciaBresciaItaly
| | - Silvia Giliani
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- “Angelo Nocivelli” Institute for Molecular Medicine, ASST Spedali Civili of BresciaBresciaItaly
| | - Simona Orcesi
- Child Neurology and Psychiatry UnitIRCCS Mondino FoundationPaviaItaly
- Department of Brain and Behavioral SciencesUniversity of PaviaPaviaItaly
| | - Lorenzo Pinelli
- Neuroradiology Unit, Section of Pediatric NeuroradiologyASST Spedali Civili of BresciaBresciaItaly
| | - Raffaele Badolato
- Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
- Pediatrics ClinicASST Spedali Civili of BresciaBresciaItaly
| | - Elisa Fazzi
- Department of Clinical and Experimental SciencesUniversity of BresciaBresciaItaly
- Unit of Child Neurology and PsychiatryASST Spedali Civili of BresciaBresciaItaly
| |
Collapse
|
23
|
Gavazzi F, Glanzman AM, Woidill S, Formanowski B, Dixit A, Isaacs D, Kornafel T, Balance E, Pierce SR, Modesti N, Barcelos I, Cusack SV, Jan AK, Flores Z, Sherbini O, Vincent A, D’Aiello R, Lorch SA, DeMauro SB, Jawad A, Vanderver A, Adang L. Exploration of Gross Motor Function in Aicardi-Goutières Syndrome. J Child Neurol 2023; 38:518-527. [PMID: 37499181 PMCID: PMC10530058 DOI: 10.1177/08830738231188753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Background: Aicardi-Goutières syndrome (AGS) is a rare genetic disorder characterized by a spectrum of motor abilities. While the Aicardi-Goutières syndrome severity score favors severely impacted individuals, there is an unmet need to define tools measuring function across the Aicardi-Goutières syndrome spectrum as potential outcome assessments for future clinical trials. Methods: Gross Motor Function Measure-88 (GMFM-88) and AGS Severity Scale were administered in individuals affected by Aicardi-Goutières syndrome (n = 71). We characterized the performance variability by genotype. Derived versions of the GMFM-88, including the GMFM-66, GMFM-66 item set (GMFM-66IS), and GMFM-66 Basal&Ceiling (GMFM-66BC) were calculated. The Aicardi-Goutières syndrome cohort was divided into severe (AGS Severity Scale score <4) or attenuated (≥4). Performance on the AGS Severity Scale highly correlated with total GMFM-88 scores (Spearman Correlation: R = 0.91). To assess variability of the GMFM-88 within genotypic subcohorts, interquartile ranges (IQRs) were compared. Results: GMFM-88 performance in the TREX1 cohort had least variability while the SAMHD1 cohort had the largest IQR (4.23 vs 81.8). Floor effect was prominent, with most evaluations scoring below 20% (n = 46, 64.79%), particularly in TREX1- and RNASEH2-cohorts. Performance by the GMFM-66, GMFM-66IS, and GMFM-66BC highly correlated with the full GMFM-88. The Aicardi-Goutières syndrome population represents a broad range of gross motor skills. Conclusions: This work identified the GMFM-88 as a potential clinical outcome assessment in subsets of the Aicardi-Goutières syndrome population but underscores the need for additional validation of outcome measures reflective of the diverse gross motor function observed in this population, including low motor function. When time is limited by resources or patient endurance, shorter versions of the GMFM-88 may be a reasonable alternative.
Collapse
Affiliation(s)
- Francesco Gavazzi
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Molecular and Translational Medicine, University of Brescia, Italy
| | - Allan M. Glanzman
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Sarah Woidill
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brielle Formanowski
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Agrani Dixit
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - David Isaacs
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tracy Kornafel
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Elizabeth Balance
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Samuel R. Pierce
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Nicholson Modesti
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Isabella Barcelos
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stacy V Cusack
- Department of Occupational Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Amanda K. Jan
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zaida Flores
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Omar Sherbini
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ariel Vincent
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Russell D’Aiello
- Department of Biomedical & Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Scott A. Lorch
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Sara B. DeMauro
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Abbas Jawad
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Adeline Vanderver
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Laura Adang
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
24
|
Gavazzi F, Patel V, Charsar B, Glanzman A, Erler J, Sevagamoorthy A, McKenzie E, Kornafel T, Ballance E, Pierce SR, Teng M, Formanowski B, Woidill S, Shults J, Wassmer E, Tonduti D, Magrinelli F, Bernard G, Van Der Knaap M, Wolf N, Adang L, Vanderver A. Gross Motor Function in Pediatric Onset TUBB4A-Related Leukodystrophy: GMFM-88 Performance and Validation of GMFC-MLD in TUBB4A. J Child Neurol 2023; 38:498-504. [PMID: 37461315 PMCID: PMC10527384 DOI: 10.1177/08830738231188159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
TUBB4A pathogenic variants are associated with a spectrum of neurologic impairments including movement disorders and leukodystrophy. With the development of targeted therapies, there is an urgent unmet need for validated tools to measure mobility impairment. Our aim is to explore gross motor function in a pediatric-onset TUBB4A-related leukodystrophy cohort with existing gross motor outcome tools. Gross Motor Function Measure-88 (GMFM-88), Gross Motor Function Classification System (GMFCS-ER), and Gross Motor Function Classification-Metachromatic Leukodystrophy (GMFC-MLD) were selected through face validity. Subjects with a confirmed clinical and molecular diagnosis of TUBB4A-related leukodystrophy were enrolled. Participants' sex, age, genotype, and age at disease onset were collected, together with GMFM-88 and concurrent GMFCS-ER and GMFC-MLD. Performances on each measure were compared. GMFM-88 floor effect was defined as total score below 20%. A total of 35 subjects participated. Median performance by GMFM-88 was 16.24% (range 0-97.31), with 42.9% (n = 15) of individuals performing above the floor. GMFM-88 Dimension A (Lying and Rolling) was the best-performing dimension in the GMFM-88 (n = 29 above the floor). All levels of the Classification Scales were represented, with the exception of the GMFC-MLD level 0. Evaluation by GMFM-88 was strongly correlated with the Classification Scales (Spearman correlations: GMFCS-ER:GMFM-88 r = 0.90; GMFC-MLD:GMFM-88 r = 0.88; GMFCS-ER:GMFC-MLD: r = 0.92). Despite overall observation of a floor effect, the GMFM-88 is able to accurately capture the performance of individuals with attenuated phenotypes. GMFM-88 Dimension A shows no floor effect. GMFC-MLD shows a strong correlation with GMFCS-ER and GMFM-88, supporting its use as an age-independent functional score in TUBB4A-related leukodystrophy.
Collapse
Affiliation(s)
- Francesco Gavazzi
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Virali Patel
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brittany Charsar
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Allan Glanzman
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jacqueline Erler
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Anjana Sevagamoorthy
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emma McKenzie
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Tracy Kornafel
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth Ballance
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Samuel R. Pierce
- Department of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Michelle Teng
- Synaptixbio Ltd, Fermi Avenue, Harwell, Oxfordshire OX11 0QX
| | - Brielle Formanowski
- Division of Neonatology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Sarah Woidill
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Justine Shults
- Synaptixbio Ltd, Fermi Avenue, Harwell, Oxfordshire OX11 0QX
| | - Evangeline Wassmer
- Neurology Department, Birmingham Children’s Hospital, Institute of Health and Neurodevelopment, Aston University, Birmingham, United Kingdom
| | - Davide Tonduti
- Unit of Pediatric Neurology, C.O.A.L.A (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, L. Sacco University Hospital, Università degli Studi di Milano, Milan, Italy
| | - Francesca Magrinelli
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Geneviève Bernard
- Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University, Montreal, Quebec, Canada
- Department Specialized Medicine, Division of Medical Genetics, McGill University Health Centre, Montreal, Quebec, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Marjo Van Der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, The Netherlands
| | - Nicole Wolf
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands
| | - Laura Adang
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adeline Vanderver
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Waugh KA, Minter R, Baxter J, Chi C, Galbraith MD, Tuttle KD, Eduthan NP, Kinning KT, Andrysik Z, Araya P, Dougherty H, Dunn LN, Ludwig M, Schade KA, Tracy D, Smith KP, Granrath RE, Busquet N, Khanal S, Anderson RD, Cox LL, Estrada BE, Rachubinski AL, Lyford HR, Britton EC, Fantauzzo KA, Orlicky DJ, Matsuda JL, Song K, Cox TC, Sullivan KD, Espinosa JM. Triplication of the interferon receptor locus contributes to hallmarks of Down syndrome in a mouse model. Nat Genet 2023; 55:1034-1047. [PMID: 37277650 PMCID: PMC10260402 DOI: 10.1038/s41588-023-01399-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 04/14/2023] [Indexed: 06/07/2023]
Abstract
Down syndrome (DS), the genetic condition caused by trisomy 21, is characterized by variable cognitive impairment, immune dysregulation, dysmorphogenesis and increased prevalence of diverse co-occurring conditions. The mechanisms by which trisomy 21 causes these effects remain largely unknown. We demonstrate that triplication of the interferon receptor (IFNR) gene cluster on chromosome 21 is necessary for multiple phenotypes in a mouse model of DS. Whole-blood transcriptome analysis demonstrated that IFNR overexpression associates with chronic interferon hyperactivity and inflammation in people with DS. To define the contribution of this locus to DS phenotypes, we used genome editing to correct its copy number in a mouse model of DS, which normalized antiviral responses, prevented heart malformations, ameliorated developmental delays, improved cognition and attenuated craniofacial anomalies. Triplication of the Ifnr locus modulates hallmarks of DS in mice, suggesting that trisomy 21 elicits an interferonopathy potentially amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Katherine A Waugh
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross Minter
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jessica Baxter
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Congwu Chi
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew D Galbraith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kathryn D Tuttle
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Neetha P Eduthan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kohl T Kinning
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zdenek Andrysik
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Paula Araya
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hannah Dougherty
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lauren N Dunn
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Michael Ludwig
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kyndal A Schade
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dayna Tracy
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Keith P Smith
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ross E Granrath
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nicolas Busquet
- Animal Behavior Core, NeuroTechnology Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Santosh Khanal
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ryan D Anderson
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Liza L Cox
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Belinda Enriquez Estrada
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Angela L Rachubinski
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Pediatrics, Section of Developmental Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Hannah R Lyford
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Eleanor C Britton
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katherine A Fantauzzo
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David J Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer L Matsuda
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, USA
| | - Kunhua Song
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- The Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Timothy C Cox
- Department of Oral and Craniofacial Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
- Department of Pediatrics, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Kelly D Sullivan
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
26
|
Fang L, Ying S, Xu X, Wu D. TREX1 cytosolic DNA degradation correlates with autoimmune disease and cancer immunity. Clin Exp Immunol 2023; 211:193-207. [PMID: 36745566 PMCID: PMC10038326 DOI: 10.1093/cei/uxad017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023] Open
Abstract
The N-terminal domain of Three Prime Repair Exonuclease 1 (TREX1) is catalytically active and can degrade dsDNA or ssDNA in the cytosol, whereas the C-terminal domain is primarily involved in protein localization. TREX1 deficiency induces cytosolic DNA accumulation as well as activation of the cGAS-STING-IFN signaling pathway, which results in tissue inflammation and autoimmune diseases. Furthermore, TREX1 expression in cancer immunity can be adaptively regulated to promote tumor proliferation, making it a promising therapeutic target.
Collapse
Affiliation(s)
- Liwei Fang
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Songcheng Ying
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Xi Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - De Wu
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Garau J, Charras A, Varesio C, Orcesi S, Dragoni F, Galli J, Fazzi E, Gagliardi S, Pansarasa O, Cereda C, Hedrich CM. Altered DNA methylation and gene expression predict disease severity in patients with Aicardi-Goutières syndrome. Clin Immunol 2023; 249:109299. [PMID: 36963449 DOI: 10.1016/j.clim.2023.109299] [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: 12/19/2022] [Revised: 02/06/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023]
Abstract
Aicardi-Goutières Syndrome (AGS) is a rare neuro-inflammatory disease characterized by increased expression of interferon-stimulated genes (ISGs). Disease-causing mutations are present in genes associated with innate antiviral responses. Disease presentation and severity vary, even between patients with identical mutations from the same family. This study investigated DNA methylation signatures in PBMCs to understand phenotypic heterogeneity in AGS patients with mutations in RNASEH2B. AGS patients presented hypomethylation of ISGs and differential methylation patterns (DMPs) in genes involved in "neutrophil and platelet activation". Patients with "mild" phenotypes exhibited DMPs in genes involved in "DNA damage and repair", whereas patients with "severe" phenotypes had DMPs in "cell fate commitment" and "organ development" associated genes. DMPs in two ISGs (IFI44L, RSAD2) associated with increased gene expression in patients with "severe" when compared to "mild" phenotypes. In conclusion, altered DNA methylation and ISG expression as biomarkers and potential future treatment targets in AGS.
Collapse
Affiliation(s)
- Jessica Garau
- Neurogenetics Research Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Amandine Charras
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Costanza Varesio
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy
| | - Simona Orcesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy
| | - Francesca Dragoni
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy; Molecular Biology and Transcriptomics, IRCCS Mondino Foundation, Pavia, Italy
| | - Jessica Galli
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Unit of Child Neurology and Psychiatry, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Elisa Fazzi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy; Unit of Child Neurology and Psychiatry, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Stella Gagliardi
- Molecular Biology and Transcriptomics, IRCCS Mondino Foundation, Pavia, Italy
| | - Orietta Pansarasa
- Cellular Model and Neuroepigenetics, IRCCS Mondino Foundation, Pavia, Italy
| | - Cristina Cereda
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Christian M Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, United Kingdom.
| |
Collapse
|
28
|
Withers SE, Rowlands CF, Tapia VS, Hedley F, Mosneag IE, Crilly S, Rice GI, Badrock AP, Hayes A, Allan SM, Briggs TA, Kasher PR. Characterization of a mutant samhd1 zebrafish model implicates dysregulation of cholesterol biosynthesis in Aicardi-Goutières syndrome. Front Immunol 2023; 14:1100967. [PMID: 36949945 PMCID: PMC10025490 DOI: 10.3389/fimmu.2023.1100967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Aicardi-Goutières syndrome (AGS1-9) is a genetically determined encephalopathy that falls under the type I interferonopathy disease class, characterized by excessive type I interferon (IFN-I) activity, coupled with upregulation of IFN-stimulated genes (ISGs), which can be explained by the vital role these proteins play in self-non-self-discrimination. To date, few mouse models fully replicate the vast clinical phenotypes observed in AGS patients. Therefore, we investigated the use of zebrafish as an alternative species for generating a clinically relevant model of AGS. Using CRISPR-cas9 technology, we generated a stable mutant zebrafish line recapitulating AGS5, which arises from recessive mutations in SAMHD1. The resulting homozygous mutant zebrafish larvae possess a number of neurological phenotypes, exemplified by variable, but increased expression of several ISGs in the head region, a significant increase in brain cell death, microcephaly and locomotion deficits. A link between IFN-I signaling and cholesterol biosynthesis has been highlighted by others, but not previously implicated in the type I interferonopathies. Through assessment of neurovascular integrity and qPCR analysis we identified a significant dysregulation of cholesterol biosynthesis in the zebrafish model. Furthermore, dysregulation of cholesterol biosynthesis gene expression was also observed through RNA sequencing analysis of AGS patient whole blood. From this novel finding, we hypothesize that cholesterol dysregulation may play a role in AGS disease pathophysiology. Further experimentation will lend critical insight into the molecular pathophysiology of AGS and the potential links involving aberrant type I IFN signaling and cholesterol dysregulation.
Collapse
Affiliation(s)
- Sarah E. Withers
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service (NHS) Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Charlie F. Rowlands
- Division of Evolution, Infection and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Victor S. Tapia
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service (NHS) Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Frances Hedley
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service (NHS) Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Ioana-Emilia Mosneag
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service (NHS) Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Siobhan Crilly
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service (NHS) Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Gillian I. Rice
- Division of Evolution, Infection and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Andrew P. Badrock
- Medical Research Council (MRC) Human Genetics Unit, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew Hayes
- Genomic Technologies Core Facility, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Stuart M. Allan
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service (NHS) Foundation Trust, The University of Manchester, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Tracy A. Briggs
- Division of Evolution, Infection and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Manchester University National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Paul R. Kasher
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance National Health Service (NHS) Foundation Trust, The University of Manchester, Manchester, United Kingdom
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
29
|
Gao X, Michel K, Griese M. Interstitial Lung Disease in Immunocompromised Children. Diagnostics (Basel) 2022; 13:diagnostics13010064. [PMID: 36611354 PMCID: PMC9818431 DOI: 10.3390/diagnostics13010064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The range of pulmonary complications beyond infections in pediatric immunocompromised patients is broad but not well characterized. Our goal was to assess the spectrum of disorders with a focus on interstitial lung diseases (ILD) in immunodeficient patients. METHODS We reviewed 217 immunocompromised children attending a specialized pneumology service during a period of 23 years. We assigned molecular diagnoses where possible and categorized the underlying immunological conditions into inborn errors of immunity or secondary immunodeficiencies according to the IUIS and the pulmonary conditions according to the chILD-EU classification system. RESULTS Among a wide array of conditions, opportunistic and chronic infections were the most frequent. ILD had a 40% prevalence. Of these children, 89% had a CT available, and 66% had a lung biopsy, which supported the diagnosis of ILD in 95% of cases. Histology was often lymphocyte predominant with the histo-pattern of granulomatous and lymphocytic interstitial lung disease (GLILD), follicular bronchiolitis or lymphocytic interstitial pneumonitis. Of interest, DIP, PAP and NSIP were also diagnosed. ILD was detected in several immunological disorders not yet associated with ILD. CONCLUSIONS Specialized pneumological expertise is necessary to manage the full spectrum of respiratory complications in pediatric immunocompromised patients.
Collapse
Affiliation(s)
| | | | - Matthias Griese
- Correspondence: ; Tel.: +49-89-4400-57870; Fax: +49-89-4400-57872
| |
Collapse
|
30
|
Modesti NB, Evans SH, Jaffe N, Vanderver A, Gavazzi F. Early recognition of patients with leukodystrophies. Curr Probl Pediatr Adolesc Health Care 2022; 52:101311. [PMID: 36470810 PMCID: PMC11326772 DOI: 10.1016/j.cppeds.2022.101311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Leukodystrophies are defined as differences in normal myelin development and maintenance in the central nervous system. They typically present as white matter imaging abnormalities in young children with delayed developmental milestones. As the scientific community begins to better understand and research the mechanisms underlying leukodystrophies, clinical trials and approved therapies for specific disorders are becoming available. These interventions, ranging from repurposing of existing small molecules to recently approved gene therapies, are highly dependent on early diagnosis. It is essential for pediatricians to identify affected individuals promptly, but they face challenges including lack of awareness of the disorders and nonspecific symptom presentation (e.g., cognitive or motor developmental delay). This review provides five hypothetical clinical presentations and describes the disease mechanisms, typical symptoms, and treatments currently available for common leukodystrophies: Krabbe Disease, Aicardi Goutières Syndrome (AGS), Metachromatic leukodystrophy (MLD), Alexander Disease (AxD), Pelizaeus-Merzbacher Disease (PMD), and X-Linked Adrenoleukodystrophy (X-ALD.) This review educates pediatricians to recognize the presentation of leukodystrophies in affected children. These clinical vignettes can serve as a framework for pediatricians to identify potentially treatable rare disorders among their patients.
Collapse
Affiliation(s)
- Nicholson B Modesti
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sarah Helen Evans
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nicole Jaffe
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adeline Vanderver
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Francesco Gavazzi
- Neurology Department, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
31
|
Incidence of Aicardi-Goutières syndrome and KCNT1-related epilepsy in Denmark. Mol Genet Metab Rep 2022; 33:100924. [PMID: 36262748 PMCID: PMC9574483 DOI: 10.1016/j.ymgmr.2022.100924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/07/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
Objective To estimate the incidence of Aicardi-Goutières syndrome (AGS) and potassium sodium-activated channel subfamily T member 1 (KCNT1)-related epilepsy in Denmark and to characterize the patients diagnosed with AGS and KCNT1-related epilepsy. Background AGS and KCNT1-related epilepsy are 2 distinct rare genetic disorders. Due to the rarity of AGS and KCNT1-related epilepsy, the epidemiology remains unclear. The incidences for these diseases or the carriers with disease-related genetic variants remain unknown. Materials and methods This is a retrospective, non-interventional, population-based study using aggregate data from the Danish population register and hospital-based patient-level data in Denmark to identify persons with genetically confirmed AGS between January 2010 to December 2020 and KCNT1-related epilepsies between January 2012 to December 2020. Cases of these disorders were identified from in-hospital databases, and pathogenic variants were identified and confirmed by Sanger and/or whole exome (panel-based) sequencing. The incidence of AGS and KCNT1-related epilepsy were estimated in separate statistical analyses. Results A total of 7 AGS patients were identified. The mean age at AGS diagnosis was 19.4 months (median age 14 months). TREX1 (n < 5) and RNASEH2B (n ≥ 5) genes were reported with confirmed pathogenic variants. The birth incidence of AGS was <0.7600 per 100,000 live births. The average annual incidence rate was calculated as 0.0539 (95% CI: 0.0217–0.1111) per 100,000 persons per year in the total population < 18 years (n = 7); the average annual incidence rate was <0.7538 per 100,000 persons per year (n < 5) in the population < 12 months, and the average annual incidence rate in the population ≥ 12 months and < 18 years was <0.0406 per 100,000 persons per year (n < 5). A total of 14 KCNT1-related epilepsy cases were identified during the study period (n = 5 in 2016, remaining 9 cases in 2013 and 2015). The mean age at diagnosis was 20.6 years (median 19 years) for KCNT1 cases. A total of 8 cases (57.1%) were ≥ 18 years, and 6 (42.9%) were < 18 years at diagnosis. The phenotype autosomal dominant or sporadic sleep-related hypermotor epilepsy (ADSHE) (n = 10, 71.4%) was most reported; the remaining 4 cases had either epilepsy of infancy with migrating focal seizures (EIMFS) or an unclassifiable developmental and epileptic encephalopathy (DEE). The birth incidence of KCNT1-related epilepsy was ≤1.1205 per 100,000 live births. The average annual incidence rates per 100,000 persons per year during the study period were 0.0431 (95% confidence interval [CI]: 0.0236–0.0723; n = 14) in the overall population ≤ 50 years, 0.0568 (95% CI: 0.0209–0.1237; n = 6) in the population < 18 years, and 0.0365 (95% CI: 0.0157–0.0718; n = 8) in the population ≥ 18 and ≤ 50 years. There were 3 families with at least 2 cases diagnosed with KCNT1-related epilepsies (on average 3.3 cases per family), indicating 10 cases in total within the 3 families. All KCNT1 cases of ADSHE phenotype came from the 3 families. The higher incidence of older ages and ADSHE cases compared with previous KCNT1 studies is likely due to the capture of prevalent and familial previously undiagnosed cases. Excluding these family cases, the average annual incidence was 0.0123 (95% CI: 0.0034–0.0315, n = 4) per 100,000 persons per year in the population ≤ 50 years during 2012–2020. Conclusions AGS and KCNT1-related epilepsy are particularly rare diseases. The annual average incidence rate of AGS was 0.0539 per 100,000 persons per year in the population < 18 years and birth incidence was <0.7600 per 100,000 live births during 2010–2020. The average annual incidence rate of KCNT1-related epilepsy was 0.0431 per 100,000 persons per year in the population ≤ 50 years and the birth incidence was ≤1.1205 per 100,000 live births during 2012–2020. Given similar healthcare systems and genetic pools, these findings may provide insight on the incidence of these rare diseases in the Nordics.
Collapse
|
32
|
Xiaoshuai L, Qiushi W, Rui W. Advantages of CRISPR-Cas9 combined organoid model in the study of congenital nervous system malformations. Front Bioeng Biotechnol 2022; 10:932936. [PMID: 36118578 PMCID: PMC9478582 DOI: 10.3389/fbioe.2022.932936] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022] Open
Abstract
In the past 10 years, gene-editing and organoid culture have completely changed the process of biology. Congenital nervous system malformations are difficult to study due to their polygenic pathogenicity, the complexity of cellular and neural regions of the brain, and the dysregulation of specific neurodevelopmental processes in humans. Therefore, the combined application of CRISPR-Cas9 in organoid models may provide a technical platform for studying organ development and congenital diseases. Here, we first summarize the occurrence of congenital neurological malformations and discuss the different modeling methods of congenital nervous system malformations. After that, it focuses on using organoid to model congenital nervous system malformations. Then we summarized the application of CRISPR-Cas9 in the organoid platform to study the pathogenesis and treatment strategies of congenital nervous system malformations and finally looked forward to the future.
Collapse
Affiliation(s)
- Li Xiaoshuai
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wang Qiushi
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wang Rui
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang, China
- *Correspondence: Wang Rui,
| |
Collapse
|
33
|
Panza E, Meyyazhagan A, Orlacchio A. Hereditary spastic paraplegia: Genetic heterogeneity and common pathways. Exp Neurol 2022; 357:114203. [PMID: 35970204 DOI: 10.1016/j.expneurol.2022.114203] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 07/11/2022] [Accepted: 08/09/2022] [Indexed: 02/07/2023]
Abstract
Hereditary Spastic Paraplegias (HSPs) are a heterogeneous group of disease, mainly characterized by progressive spasticity and weakness of the lower limbs resulting from distal degeneration of corticospinal tract axons. Although HSPs represent rare or ultra-rare conditions, with reported cases of mutated genes found in single families, overall, with 87 forms described, they are an important health and economic problem for society and patients. In fact, they are chronic and life-hindering conditions, still lacking a specific therapy. Notwithstanding the number of forms described, and 73 causative genes identified, overall, the molecular diagnostic rate varies among 29% to 61.8%, based on recent published analysis, suggesting that more genes are involved in HSP and/or that different molecular diagnostic approaches are necessary. The accumulating data in this field highlight several peculiar features of HSPs, such as genetic heterogeneity, the discovery that different mutations in a single gene can be transmitted in dominant and recessive trait in families and allelic heterogeneity, resulting in the involvement of HSP-genes in other conditions. Based on the observation of protein functions, the activity of many different proteins encoded by HSP-related genes converges into some distinct pathophysiological mechanisms. This suggests that common pathways could be a potential target for a therapy, possibly addressing several forms at once. Furthermore, the overlap of HSP genes with other neurological conditions can further expand this concept.
Collapse
Affiliation(s)
- Emanuele Panza
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Arun Meyyazhagan
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| | - Antonio Orlacchio
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy; Laboratorio di Neurogenetica, Centro Europeo di Ricerca sul Cervello (CERC), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
34
|
Lee WF, Fan WL, Tseng MH, Yang HY, Huang JL, Wu CY. Characteristics and genetic analysis of patients suspected with early-onset systemic lupus erythematosus. Pediatr Rheumatol Online J 2022; 20:68. [PMID: 35964089 PMCID: PMC9375402 DOI: 10.1186/s12969-022-00722-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/24/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is rarely diagnosed before 5-years-old. Those with disease onset at a very young age are predicted by a higher genetic risk and a more severe phenotype. We performed whole-exome sequencing to survey the genetic etiologies and clinical manifestations in patients fulfilling 2012 SLICC SLE classification criteria before the age of 5. CASE PRESENTATION Among the 184 childhood-onset SLE patients regularly followed in a tertiary medical center in Taiwan, 7 cases (3.8%) of which onset ≦ 5 years of age were identified for characteristic review and genetic analysis. Compared to those onset at elder age, cases onset before the age of 5 are more likely to suffer from proliferative glomerulonephritis, renal thrombotic microangiopathy, neuropsychiatric disorder and failure to thrive. Causative genetic etiologies were identified in 3. In addition to the abundance of autoantibodies, patient with homozygous TREX1 (c.292_293 ins A) mutation presented with chilblain-like skin lesions, peripheral spasticity, endocrinopathy and experienced multiple invasive infections. Patient with SLC7A7 (c.625 + 1 G > A) mutation suffered from profound glomerulonephritis with full-house glomerular deposits as well as hyperammonemia, metabolic acidosis and episodic conscious disturbance. Two other cases harbored variants in lupus associating genes C1s, C2, DNASE1 and DNASE1L3 and another with CFHR4. Despite fulfilling the classification criteria for lupus, many of the patients required treatments beyond conventional therapy. CONCLUSIONS Genetic etiologies and lupus mimickers were found among a substantial proportion of patients suspected with early-onset SLE. Detail clinical evaluation and genetic testing are important for tailored care and personalized treatment.
Collapse
Affiliation(s)
- Wan-Fang Lee
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, No.5 Fu-Hsing St. Kuei Shan Hsiang, Taoyuan, Taoyuan Hsien, Taiwan
| | - Wen-Lang Fan
- Genomic Medicine Research Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Min-Hua Tseng
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Nephrology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Huang-Yu Yang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jing-Long Huang
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, No.5 Fu-Hsing St. Kuei Shan Hsiang, Taoyuan, Taoyuan Hsien, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei city, Taiwan.
| | - Chao-Yi Wu
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, No.5 Fu-Hsing St. Kuei Shan Hsiang, Taoyuan, Taoyuan Hsien, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
35
|
Adang L. Leukodystrophies. Continuum (Minneap Minn) 2022; 28:1194-1216. [PMID: 35938662 PMCID: PMC11320896 DOI: 10.1212/con.0000000000001130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
PURPOSE OF REVIEW This article reviews the most common leukodystrophies and is focused on diagnosis, clinical features, and emerging therapeutic options. RECENT FINDINGS In the past decade, the recognition of leukodystrophies has exponentially increased, and now this class includes more than 30 distinct disorders. Classically recognized as progressive and fatal disorders affecting young children, it is now understood that leukodystrophies are associated with an increasing spectrum of neurologic trajectories and can affect all ages. Next-generation sequencing and newborn screening allow the opportunity for the recognition of presymptomatic and atypical cases. These new testing opportunities, in combination with growing numbers of natural history studies and clinical consensus guidelines, have helped improve diagnosis and clinical care. Additionally, a more granular understanding of disease outcomes informs clinical trial design and has led to several recent therapeutic advances. This review summarizes the current understanding of the clinical manifestations of disease and treatment options for the most common leukodystrophies. SUMMARY As early testing becomes more readily available through next-generation sequencing and newborn screening, neurologists will better understand the true incidence of the leukodystrophies and be able to diagnose children within the therapeutic window. As targeted therapies are developed, it becomes increasingly imperative that this broad spectrum of disorders is recognized and diagnosed. This work summarizes key advances in the leukodystrophy field.
Collapse
|
36
|
Cetin Gedik K, Lamot L, Romano M, Demirkaya E, Piskin D, Torreggiani S, Adang LA, Armangue T, Barchus K, Cordova DR, Crow YJ, Dale RC, Durrant KL, Eleftheriou D, Fazzi EM, Gattorno M, Gavazzi F, Hanson EP, Lee-Kirsch MA, Montealegre Sanchez GA, Neven B, Orcesi S, Ozen S, Poli MC, Schumacher E, Tonduti D, Uss K, Aletaha D, Feldman BM, Vanderver A, Brogan PA, Goldbach-Mansky R. The 2021 European Alliance of Associations for Rheumatology/American College of Rheumatology Points to Consider for Diagnosis and Management of Autoinflammatory Type I Interferonopathies: CANDLE/PRAAS, SAVI, and AGS. Arthritis Rheumatol 2022; 74:735-751. [PMID: 35315249 DOI: 10.1002/art.42087] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/11/2022] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Autoinflammatory type I interferonopathies, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature/proteasome-associated autoinflammatory syndrome (CANDLE/PRAAS), stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy (SAVI), and Aicardi-Goutières syndrome (AGS) are rare and clinically complex immunodysregulatory diseases. With emerging knowledge of genetic causes and targeted treatments, a Task Force was charged with the development of "points to consider" to improve diagnosis, treatment, and long-term monitoring of patients with these rare diseases. METHODS Members of a Task Force consisting of rheumatologists, neurologists, an immunologist, geneticists, patient advocates, and an allied health care professional formulated research questions for a systematic literature review. Then, based on literature, Delphi questionnaires, and consensus methodology, "points to consider" to guide patient management were developed. RESULTS The Task Force devised consensus and evidence-based guidance of 4 overarching principles and 17 points to consider regarding the diagnosis, treatment, and long-term monitoring of patients with the autoinflammatory interferonopathies, CANDLE/PRAAS, SAVI, and AGS. CONCLUSION These points to consider represent state-of-the-art knowledge to guide diagnostic evaluation, treatment, and management of patients with CANDLE/PRAAS, SAVI, and AGS and aim to standardize and improve care, quality of life, and disease outcomes.
Collapse
Affiliation(s)
- Kader Cetin Gedik
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | - Lovro Lamot
- University of Zagreb School of Medicine, Zagreb, Croatia
| | - Micol Romano
- University of Western Ontario, London, Ontario, Canada
| | | | - David Piskin
- University of Western Ontario, London Health Sciences Center, and Lawson Health Research Institute, London, Ontario, Canada
| | - Sofia Torreggiani
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, and UOC Pediatria a Media Intensità di Cura, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura A Adang
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Thais Armangue
- Sant Joan de Deu Children's Hospital and IDIBAPS-Hospital Clinic; University of Barcelona, Barcelona, Spain
| | - Kathe Barchus
- Autoinflammatory Alliance, San Francisco, California
| | - Devon R Cordova
- Aicardi-Goutieres Syndrome Americas Association, Manhattan Beach, California
| | - Yanick J Crow
- University of Edinburgh, Edinburgh, UK, and Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, University of Paris, Paris, France
| | - Russell C Dale
- University of Sydney, Sydney, New South Wales, Australia
| | - Karen L Durrant
- Autoinflammatory Alliance and Kaiser San Francisco Hospital, San Francisco, California
| | | | - Elisa M Fazzi
- ASST Civil Hospital and University of Brescia, Brescia, Italy
| | | | - Francesco Gavazzi
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, and University of Brescia, Brescia, Italy
| | - Eric P Hanson
- Riley Hospital for Children and Indiana University School of Medicine, Indianapolis
| | | | | | - Bénédicte Neven
- Necker Children's Hospital, AP-HP, Institut Imagine Institut des Maladies Genetiques, University of Paris, Paris, France
| | - Simona Orcesi
- IRCCS Mondino Foundation and University of Pavia, Pavia, Italy
| | - Seza Ozen
- Hacettepe University, Ankara, Turkey
| | | | | | | | - Katsiaryna Uss
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland
| | | | - Brian M Feldman
- Hospital for Sick Children and University of Toronto Institute of Health Policy Management and Evaluation, Toronto, Ontario, Canada
| | - Adeline Vanderver
- Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia
| | | | | |
Collapse
|
37
|
Cetin Gedik K, Lamot L, Romano M, Demirkaya E, Piskin D, Torreggiani S, Adang LA, Armangue T, Barchus K, Cordova DR, Crow YJ, Dale RC, Durrant KL, Eleftheriou D, Fazzi EM, Gattorno M, Gavazzi F, Hanson EP, Lee-Kirsch MA, Montealegre Sanchez GA, Neven B, Orcesi S, Ozen S, Poli MC, Schumacher E, Tonduti D, Uss K, Aletaha D, Feldman BM, Vanderver A, Brogan PA, Goldbach-Mansky R. The 2021 EULAR and ACR points to consider for diagnosis and management of autoinflammatory type I interferonopathies: CANDLE/PRAAS, SAVI and AGS. Ann Rheum Dis 2022; 81:601-613. [PMID: 35086813 PMCID: PMC9036471 DOI: 10.1136/annrheumdis-2021-221814] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/11/2022] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Autoinflammatory type I interferonopathies, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature/proteasome-associated autoinflammatory syndrome (CANDLE/PRAAS), stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy (SAVI) and Aicardi-Goutières syndrome (AGS) are rare and clinically complex immunodysregulatory diseases. With emerging knowledge of genetic causes and targeted treatments, a Task Force was charged with the development of 'points to consider' to improve diagnosis, treatment and long-term monitoring of patients with these rare diseases. METHODS Members of a Task Force consisting of rheumatologists, neurologists, an immunologist, geneticists, patient advocates and an allied healthcare professional formulated research questions for a systematic literature review. Then, based on literature, Delphi questionnaires and consensus methodology, 'points to consider' to guide patient management were developed. RESULTS The Task Force devised consensus and evidence-based guidance of 4 overarching principles and 17 points to consider regarding the diagnosis, treatment and long-term monitoring of patients with the autoinflammatory interferonopathies, CANDLE/PRAAS, SAVI and AGS. CONCLUSION These points to consider represent state-of-the-art knowledge to guide diagnostic evaluation, treatment and management of patients with CANDLE/PRAAS, SAVI and AGS and aim to standardise and improve care, quality of life and disease outcomes.
Collapse
Affiliation(s)
- Kader Cetin Gedik
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lovro Lamot
- Department of Pediatrics, University Hospital Centre Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Micol Romano
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Erkan Demirkaya
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - David Piskin
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada.,London Health Sciences Center, Lawson Health Research Institute, London, Ontario, Canada
| | - Sofia Torreggiani
- 1Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA.,UOC Pediatria a Media Intensità di Cura, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Lombardia, Italy
| | - Laura A Adang
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Thais Armangue
- Pediatric Neuroimmunology Unit, Neurology Service, Sant Joan de Deu Children's Hospital, and IDIBAPS-Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Kathe Barchus
- Autoinflammatory Alliance, San Francisco, California, USA
| | - Devon R Cordova
- Aicardi-Goutieres Syndrome Americas Association, Manhattan Beach, California, USA
| | - Yanick J Crow
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburg, Edinburg, UK.,Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Université de Paris, Paris, Île-de-France, France
| | - Russell C Dale
- Kids Neuroscience Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Karen L Durrant
- Autoinflammatory Alliance, San Francisco, California, USA.,Kaiser San Francisco Hospital, San Francisco, California, USA
| | - Despina Eleftheriou
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Elisa M Fazzi
- Child Neurology and Psychiatry Unit, Department of Clinical and Experimental Sciences ASST Civil Hospital, University of Brescia, Brescia, Italy
| | - Marco Gattorno
- Center for Autoinflammatory diseases and Immunodeficiencies, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Francesco Gavazzi
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Eric P Hanson
- Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Min Ae Lee-Kirsch
- Department of Pediatrics, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Gina A Montealegre Sanchez
- Intramural Clinical Management and Operations Branch (ICMOB), Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Bénédicte Neven
- Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, Institut Imagine Institut des Maladies Genetiques, Paris, Île-de-France, France
| | - Simona Orcesi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy, Italy.,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Lombardia, Italy
| | - Seza Ozen
- Pediatric Rheumatology, Hacettepe University, Ankara, Turkey
| | - M Cecilia Poli
- Department of Pediatrics, Facultad de Medicina Clinica Alemana Universidad del Desarrollo, Santiago, Chile
| | | | - Davide Tonduti
- Child Neurology Unit, COALA (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children's Hospital, Milano, Italy
| | - Katsiaryna Uss
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Aletaha
- Department of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Brian M Feldman
- Division of Rheumatology, Hospital for Sick Children, Toronto, Ontario, Canada.,30Department of Pediatrics, Faculty of Medicine, University of Toronto Institute of Health Policy Management and Evaluation, Toronto, Ontario, Canada
| | - Adeline Vanderver
- Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul A Brogan
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
38
|
Gavazzi F, Fraser JL, Bloom M, Tochen L, Rhee J, Kwan M, Victoria T, Teachey DT, Ho CY, Vanderver A, Linn RL. Hodgkin lymphoma in an individual with TREX1-mediated Aicardi Goutières syndrome. Pediatr Blood Cancer 2022; 69:e29322. [PMID: 34490982 PMCID: PMC11348674 DOI: 10.1002/pbc.29322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Francesco Gavazzi
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jamie L. Fraser
- Rare Disease Institute, Division of Genetics and Metabolism, Children’s National Hospital, Washington, District of Columbia, USA
| | - Miriam Bloom
- Department of Pediatrics, Children’s National Hospital, Washington, District of Columbia, USA
| | - Laura Tochen
- Department of Neurology, Children’s National Hospital, Washington, District of Columbia, USA
| | - Jullie Rhee
- Department of Neurology, Children’s National Hospital, Washington, District of Columbia, USA
| | - Megan Kwan
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Teresa Victoria
- Division of Radiology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - David T. Teachey
- Divisions of Hematology and Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Divisions of Hematology and Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Cheng-Ying Ho
- Department of Pathology and Neurology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Adeline Vanderver
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rebecca L. Linn
- Department of Pathology and Lab Medicine at Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Gavazzi F, Adang L, Waldman A, Jan AK, Liu G, Lorch SA, DeMauro SB, Shults J, Pierce SR, Ballance E, Kornafel T, Harrington A, Glanzman AM, Vanderver A. Reliability of the Telemedicine Application of the Gross Motor Function Measure-88 in Patients With Leukodystrophy. Pediatr Neurol 2021; 125:34-39. [PMID: 34624609 PMCID: PMC8629609 DOI: 10.1016/j.pediatrneurol.2021.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Leukodystrophies are a rare class of disorders characterized by severe neuromotor disability. There is a strong need for research regarding the functional status of people with leukodystrophy which is limited by the need for in-person assessments of mobility. The purpose of this study is to assess the reliability of the Gross Motor Function Measure-88 (GMFM-88) using telemedicine compared with standard in-person assessments in patients with leukodystrophy. METHODS A total of 21 subjects with a diagnosis of leukodystrophy (age range = 1.79-52.82 years) were evaluated by in-person and by telemedicine evaluations with the GMFM-88 by physical therapists. Inter-rater reliability was assessed through evaluation of the same subject by two independent raters within a three-week period (n = 10 encounters), and intrarater reliability was assessed through blinded rescoring of video-recorded assessments after a one-week time interval (n = 6 encounters). RESULTS Remote assessments were performed by caregivers in all 21 subjects using resources found in the home with remote guidance. There was agreement between all paired in-person and remote measurements (Lin's concordance correlation ≥0.995). The Bland-Altman analysis indicated that the paired differences were within ±5%. Intrarater and inter-rater reliability demonstrated an intraclass correlation coefficient of >0.90. CONCLUSIONS These results support that remote application of the GMFM-88 is a feasible and reliable approach to assess individuals with leukodystrophy. Telemedicine application of outcome measures may be of particular value in rare diseases and those with severe neurologic disability that impacts the ability to travel.
Collapse
Affiliation(s)
- Francesco Gavazzi
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Laura Adang
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Amy Waldman
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amanda K. Jan
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Geraldine Liu
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Scott A. Lorch
- Department of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Sara B. DeMauro
- Department of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Justine Shults
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Samuel R. Pierce
- Departmen of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Elizabeth Ballance
- Departmen of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Tracy Kornafel
- Departmen of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Ann Harrington
- Departmen of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Allan M. Glanzman
- Departmen of Physical Therapy, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Adeline Vanderver
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
40
|
Gavazzi F, Charsar BA, Williams C, Shults J, Alves CA, Adang L, Vanderver A. Acquisition of Developmental Milestones in Hypomyelination With Atrophy of the Basal Ganglia and Cerebellum and Other TUBB4A-Related Leukoencephalopathy. J Child Neurol 2021; 36:805-811. [PMID: 34514881 PMCID: PMC8505576 DOI: 10.1177/08830738211000977] [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] [Indexed: 01/22/2023]
Abstract
Mutations in TUBB4A are associated with a spectrum of neurologic disorders categorized as TUBB4A-related leukoencephalopathy. Affected children can present with global developmental delay or normal early development, followed by a variable loss of skills over time. Further research is needed to characterize the factors associated with the divergent developmental trajectories in this rare monogenic disorder because this phenotypic spectrum is not fully explained by genotype alone.To characterize early psychomotor features, developmental milestones and age of disease onset were collected from medical records (n=54 individuals). Three subcohorts were identified: individuals with the common p.Asp249Asn variant vs all other genotypes with either early (<12 months of age) or late onset of presentation. Individuals with the p.Asp249Asn variant or those with non-p.Asp249Asn genotypes with later disease onset attained key milestones, including head control, sitting, and independent walking. Subjects with early-onset, non-p.Asp249Asn-associated disease were less likely to achieve developmental milestones. Next, we defined the developmental severity as the percentage of milestones attained by age 2 years. The mild form was defined as attaining at least 75% of key developmental milestones. Among cohort categorized as mild, individuals with p.Asp249Asn variant were more likely to lose acquired abilities when compared with non-p.Asp249Asn individuals.Our results suggest multiple influences on developmental trajectory, including a strong contribution from genotype and age of onset. Further studies are needed to identify additional factors that influence overall outcomes to better counsel families and to design clinical trials with appropriate clinical endpoints.
Collapse
Affiliation(s)
- Francesco Gavazzi
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Department of Molecular and Translational Medicine, University of Brescia, Italy
| | | | - Catherine Williams
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Justine Shults
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Cesar A. Alves
- Division of Neuroradiology, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Laura Adang
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Adeline Vanderver
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
41
|
Maletzko A, Key J, Wittig I, Gispert S, Koepf G, Canet-Pons J, Torres-Odio S, West AP, Auburger G. Increased presence of nuclear DNAJA3 and upregulation of cytosolic STAT1 and of nucleic acid sensors trigger innate immunity in the ClpP-null mouse. Neurogenetics 2021; 22:297-312. [PMID: 34345994 PMCID: PMC8426249 DOI: 10.1007/s10048-021-00657-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 07/08/2021] [Indexed: 11/29/2022]
Abstract
Mitochondrial dysfunction may activate innate immunity, e.g. upon abnormal handling of mitochondrial DNA in TFAM mutants or in altered mitophagy. Recent reports showed that also deletion of mitochondrial matrix peptidase ClpP in mice triggers transcriptional upregulation of inflammatory factors. Here, we studied ClpP-null mouse brain at two ages and mouse embryonal fibroblasts, to identify which signaling pathways are responsible, employing mass spectrometry, subcellular fractionation, immunoblots, and reverse transcriptase polymerase chain reaction. Several mitochondrial unfolded protein response factors showed accumulation and altered migration in blue-native gels, prominently the co-chaperone DNAJA3. Its mitochondrial dysregulation increased also its extra-mitochondrial abundance in the nucleus, a relevant observation given that DNAJA3 modulates innate immunity. Similar observations were made for STAT1, a putative DNAJA3 interactor. Elevated expression was observed not only for the transcription factors Stat1/2, but also for two interferon-stimulated genes (Ifi44, Gbp3). Inflammatory responses were strongest for the RLR pattern recognition receptors (Ddx58, Ifih1, Oasl2, Trim25) and several cytosolic nucleic acid sensors (Ifit1, Ifit3, Oas1b, Ifi204, Mnda). The consistent dysregulation of these factors from an early age might influence also human Perrault syndrome, where ClpP loss-of-function leads to early infertility and deafness, with subsequent widespread neurodegeneration.
Collapse
Affiliation(s)
- Antonia Maletzko
- Experimental Neurology, Medical School, Goethe University, 60590, Frankfurt, Germany
| | - Jana Key
- Experimental Neurology, Medical School, Goethe University, 60590, Frankfurt, Germany.,Faculty of Biosciences, Goethe University, Altenhöferallee 1, 60438, Frankfurt, Germany
| | - Ilka Wittig
- Functional Proteomics, Faculty of Medicine, Goethe University, 60590, Frankfurt, Germany
| | - Suzana Gispert
- Experimental Neurology, Medical School, Goethe University, 60590, Frankfurt, Germany
| | - Gabriele Koepf
- Experimental Neurology, Medical School, Goethe University, 60590, Frankfurt, Germany
| | - Júlia Canet-Pons
- Experimental Neurology, Medical School, Goethe University, 60590, Frankfurt, Germany
| | - Sylvia Torres-Odio
- Experimental Neurology, Medical School, Goethe University, 60590, Frankfurt, Germany.,Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M, University Health Science Center, Bryan, TX, 77807, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M, University Health Science Center, Bryan, TX, 77807, USA
| | - Georg Auburger
- Experimental Neurology, Medical School, Goethe University, 60590, Frankfurt, Germany.
| |
Collapse
|
42
|
Mura E, Masnada S, Antonello C, Parazzini C, Izzo G, Garau J, Sproviero D, Cereda C, Orcesi S, Veggiotti P, Zuccotti G, Dilillo D, Penagini F, Tonduti D. Ruxolitinib in Aicardi-Goutières syndrome. Metab Brain Dis 2021; 36:859-863. [PMID: 33721182 DOI: 10.1007/s11011-021-00716-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/10/2021] [Indexed: 12/31/2022]
Abstract
Aicardi-Goutières Syndrome (AGS) is a monogenic leukodystrophy with pediatric onset, clinically characterized by a variable degree of neurologic impairment. It belongs to a group of condition called type I interferonopathies that are characterized by abnormal overproduction of interferon alpha, an inflammatory cytokine which action is mediated by the activation of two of the four human Janus Kinases. Thanks to an ever-increasing knowledge of the molecular basis and pathogenetic mechanisms of the disease, Janus Kinase inhibitors (JAKIs) have been proposed as a treatment option for selected interferonopathies. Here we reported the 24 months follow-up of the fifth AGS patient treated with ruxolitinib described so far in literature. The treatment was globally well tolerated; clinical examinations and radiological images demonstrated a progressively improving course. It is however to note that patients presenting with mild and spontaneously improving course have been reported. Large natural history studies on AGS spectrum are strongly required in order to get a better understanding of the results emerging from ongoing therapeutic trials on such rare disease.
Collapse
Affiliation(s)
- Eleonora Mura
- Department of Pediatric Neurology, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
| | - Silvia Masnada
- Department of Pediatric Neurology, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
| | - Clara Antonello
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- Department of Pediatric Orthopedics, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
| | - Cecilia Parazzini
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- Department of Pediatric Radiology and Neuroradiology, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
| | - Giana Izzo
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- Department of Pediatric Radiology and Neuroradiology, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
| | - Jessica Garau
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
| | - Daisy Sproviero
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100, Pavia, Italy
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100, Pavia, Italy
| | - Simona Orcesi
- Department of Brain and Behavioral Sciences, University of Pavia, Via Bassi 21, 27100, Pavia, Italy
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Via Mondino 2, 27100, Pavia, Italy
| | - Pierangelo Veggiotti
- Department of Pediatric Neurology, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- Department of Biomedical and Clinical Science "L. Sacco", University of Milan, Via Giovanni Battista Grassi 74, 20157, Milan, Italy
| | - Gianvincenzo Zuccotti
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- Department of Biomedical and Clinical Science "L. Sacco", University of Milan, Via Giovanni Battista Grassi 74, 20157, Milan, Italy
- Department of Pediatrics, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
| | - Dario Dilillo
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- Department of Pediatrics, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
| | - Francesca Penagini
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
- Department of Pediatrics, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy
| | - Davide Tonduti
- Department of Pediatric Neurology, V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy.
- C.O.A.L.A (Center for diagnosis and treatment of leukodystrophies), V. Buzzi Children's Hospital, Via Castelvetro 32, 20154, Milan, Italy.
| |
Collapse
|
43
|
Gavazzi F, Charsar BA, Williams C, Shults J, Alves CA, Adang L, Vanderver A. Acquisition of Developmental Milestones in Hypomyelination With Atrophy of the Basal Ganglia and Cerebellum and Other TUBB4A-Related Leukoencephalopathy. J Child Neurol 2021. [PMID: 33843299 DOI: 10.1177/0883073821000977] [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] [Indexed: 11/16/2022]
Abstract
Mutations in TUBB4A are associated with a spectrum of neurologic disorders categorized as TUBB4A-related leukoencephalopathy. Affected children can present with global developmental delay or normal early development, followed by a variable loss of skills over time. Further research is needed to characterize the factors associated with the divergent developmental trajectories in this rare monogenic disorder because this phenotypic spectrum is not fully explained by genotype alone.To characterize early psychomotor features, developmental milestones and age of disease onset were collected from medical records (n=54 individuals). Three subcohorts were identified: individuals with the common p.Asp249Asn variant vs all other genotypes with either early (<12 months of age) or late onset of presentation. Individuals with the p.Asp249Asn variant or those with non-p.Asp249Asn genotypes with later disease onset attained key milestones, including head control, sitting, and independent walking. Subjects with early-onset, non-p.Asp249Asn-associated disease were less likely to achieve developmental milestones. Next, we defined the developmental severity as the percentage of milestones attained by age 2 years. The mild form was defined as attaining at least 75% of key developmental milestones. Among cohort categorized as mild, individuals with p.Asp249Asn variant were more likely to lose acquired abilities when compared with non-p.Asp249Asn individuals.Our results suggest multiple influences on developmental trajectory, including a strong contribution from genotype and age of onset. Further studies are needed to identify additional factors that influence overall outcomes to better counsel families and to design clinical trials with appropriate clinical endpoints.
Collapse
Affiliation(s)
- Francesco Gavazzi
- Division of Neurology, 6567Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Brittany A Charsar
- Sidney Kimmel Medical College, 23217Jefferson University, Philadelphia, PA, USA
| | - Catherine Williams
- Division of Neurology, 6567Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Justine Shults
- Department of Pediatrics, 6567Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cesar A Alves
- Division of Neuroradiology, 6567Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laura Adang
- Division of Neurology, 6567Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adeline Vanderver
- Division of Neurology, 6567Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
44
|
Teke Kisa P, Arslan N. Inborn errors of immunity and metabolic disorders: current understanding, diagnosis, and treatment approaches. J Pediatr Endocrinol Metab 2021; 34:277-294. [PMID: 33675210 DOI: 10.1515/jpem-2020-0277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/19/2020] [Indexed: 12/31/2022]
Abstract
Inborn errors of metabolism consist of a heterogeneous group of disorders with various organ systems manifestations, and some metabolic diseases also cause immunological disorders or dysregulation. In this review, metabolic diseases that affect the immunological system and particularly lead to primary immune deficiency will be reviewed. In a patient with frequent infections and immunodeficiency, the presence of symptoms such as growth retardation, abnormal facial appearance, heart, skeletal, lung deformities, skin findings, arthritis, motor developmental retardation, seizure, deafness, hepatomegaly, splenomegaly, impairment of liver function tests, the presence of anemia, thrombocytopenia and eosinophilia in hematological examinations should suggest metabolic diseases for the underlying cause. In some patients, these phenotypic findings may appear before the immunodeficiency picture. Metabolic diseases leading to immunological disorders are likely to be rare but probably underdiagnosed. Therefore, the presence of recurrent infections or autoimmune findings in a patient with a suspected metabolic disease should suggest that immune deficiency may also accompany the picture, and diagnostic examinations in this regard should be deepened.
Collapse
Affiliation(s)
- Pelin Teke Kisa
- Division of Pediatric Metabolism and Nutrition, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Nur Arslan
- Division of Pediatric Metabolism and Nutrition, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| |
Collapse
|
45
|
De Giorgis V, Varesio C, Viri M, Giordano L, La Piana R, Tonduti D, Roncarolo F, Masnada S, Pichiecchio A, Veggiotti P, Fazzi E, Orcesi S. The epileptology of Aicardi-Goutières syndrome: electro-clinical-radiological findings. Seizure 2021; 86:197-209. [PMID: 33589296 DOI: 10.1016/j.seizure.2020.11.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVE Although epileptic seizures occur in approximately a quarter of patients with Aicardi-Goutières syndrome (AGS), their phenotypic and electrophysiological characterization remains elusive. The aim of our study was to characterize epilepsy phenotypes and electroencephalographic (EEG) patterns in AGS and look for possible correlations with clinical, genetic and neuroradiological features. METHODS We selected patients with an established AGS diagnosis followed at three Italian reference centers. Medical records, EEGs and MRI/CT findings were reviewed. EEGs were independently and blindly reviewed by three board-certified pediatric epileptologists. Chi square and Fisher's exact tests were used to test associations between epilepsy and EEG feature categories and clinical, radiological and genetic variables. RESULTS Twenty-seven patients were enrolled. We reviewed 63 EEGs and at least one brain MRI scan per patient. Epilepsy, mainly in the form of epileptic spasms and focal seizures, was present in 37 % of the cohort; mean age at epilepsy onset was 9.5 months (range 1-36). The presence of epilepsy was associated with calcification severity (p = 0.016) and startle reactions (p = 0.05). Organization of EEG electrical activity appeared to be disrupted or markedly disrupted in 73 % of cases. Severe EEG disorganization correlated with microcephaly (p < 0.001) and highly abnormal MRI T2-weighted signal intensity in white matter (p = 0.022). Physiological organization of the EEG was found to be better preserved during sleep (87 %) than wakefulness (38 %). Focal slow activity was recorded in more than one third of cases. Fast activity, either diffuse or with frontal location, was more frequent in the awake state (78 %) than in sleep (50 %). Interictal epileptiform discharges (IEDs) were present in 33 % of awake and 45 % of sleep recordings. IEDs during sleep were associated with a higher risk of a epileptic seizures (p = 0.008). SIGNIFICANCE The hallmarks of EEG recordings in AGS were found to be: disruption of electrical organization, the presence of focal slow and fast activity, and the presence of IEDs, both in patients with and in those without epilepsy. The associations between epilepsy and calcification and between EEG pattern and the finding of a highly abnormal white matter T2 signal intensity suggest a common anatomical correlate. However, the complex anatomical-electroclinical basis of AGS-related epilepsy still requires further elucidation.
Collapse
Affiliation(s)
- Valentina De Giorgis
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy
| | - Costanza Varesio
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy.
| | - Maurizio Viri
- Department of Child Neurology and Psychiatry, AOU Maggiore della Carità Novara, Novara, Italy
| | - Lucio Giordano
- Child Neurology and Psychiatry Unit, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Roberta La Piana
- Department of Neuroradiology and Laboratory of Neurogenetics of Motion, Neurological Institute and Hospital, McGill University, Montreal, QC H3A2B4, Canada
| | - Davide Tonduti
- Pediatric Neurology Unit - COALA (Center for Diagnosis and Treatment of Leukodystrophies) -V. Buzzi Children's Hospital, Milan, Italy
| | - Federico Roncarolo
- Institute of Public Health Research of University of Montreal (IRSPUM), University of Montreal, Montreal, QC, Canada
| | - Silvia Masnada
- Pediatric Neurology Unit - COALA (Center for Diagnosis and Treatment of Leukodystrophies) -V. Buzzi Children's Hospital, Milan, Italy
| | - Anna Pichiecchio
- Neuroradiology Unit, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Pierangelo Veggiotti
- Pediatric Neurology Unit - COALA (Center for Diagnosis and Treatment of Leukodystrophies) -V. Buzzi Children's Hospital, Milan, Italy; Biomedical and Clinical Sciences Department, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Elisa Fazzi
- Child Neurology and Psychiatry Unit, ASST Spedali Civili di Brescia, Brescia, Italy; Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Simona Orcesi
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | | |
Collapse
|
46
|
Piccoli C, Bronner N, Gavazzi F, Dubbs H, De Simone M, De Giorgis V, Orcesi S, Fazzi E, Galli J, Masnada S, Tonduti D, Varesio C, Vanderver A, Vossough A, Adang L. Late-Onset Aicardi-Goutières Syndrome: A Characterization of Presenting Clinical Features. Pediatr Neurol 2021; 115:1-6. [PMID: 33307271 PMCID: PMC7856674 DOI: 10.1016/j.pediatrneurol.2020.10.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Aicardi-Goutières syndrome (AGS) is a genetic interferonopathy characterized by early onset of severe neurological injury with intracranial calcifications, leukoencephalopathy, and systemic inflammation. Increasingly, a spectrum of neurological dysfunction and presentation beyond the infantile period is being recognized in AGS. The aim of this study was to characterize late-infantile and juvenile-onset AGS. METHODS We conducted a multi-institution review of individuals with AGS who were older than one year at the time of presentation, including medical history, imaging characteristics, and suspected diagnoses at presentation. RESULTS Thirty-four individuals were identified, all with pathogenic variants in RNASEH2B, SAMHD1, ADAR1, or IFIH1. Most individuals had a history of developmental delay and/or systemic symptoms, such as sterile pyrexias and chilblains, followed by a prodromal period associated with increasing symptoms. This was followed by an abrupt onset of neurological decline (fulminant phase), with a median onset at 1.33 years (range 1.00 to 17.68 years). Most individuals presented with a change in gross motor skills (97.0%), typically with increased tone (78.8%). Leukodystrophy was the most common magnetic resonance imaging finding (40.0%). Calcifications were less common (12.9%). CONCLUSIONS This is the first study to characterize the presentation of late-infantile and juvenile onset AGS and its phenotypic spectrum. Late-onset AGS can present insidiously and lacks classical clinical and neuroimaging findings. Signs of early systemic dysfunction before fulminant disease onset and loss of motor symptoms were common. We strongly recommend genetic testing when there is concern for sustained inflammation of unknown origins or changes in motor skills in children older than one year.
Collapse
Affiliation(s)
- Cara Piccoli
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Nowa Bronner
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Holly Dubbs
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Micaela De Simone
- ASST Spedali Civili di Brescia, Azienda Socio Sanitaria Territoriale degli Spedali Civili di Brescia, Brescia, Italy
| | | | | | - Elisa Fazzi
- ASST Spedali Civili di Brescia, Azienda Socio Sanitaria Territoriale degli Spedali Civili di Brescia, Brescia, Italy
| | - Jessica Galli
- ASST Spedali Civili di Brescia, Azienda Socio Sanitaria Territoriale degli Spedali Civili di Brescia, Brescia, Italy
| | - Silvia Masnada
- Vittore Buzzi Children’s Hospital, Ospedale dei Bambini Vittore Buzzi, Milan, Italy
| | - Davide Tonduti
- Vittore Buzzi Children’s Hospital, Ospedale dei Bambini Vittore Buzzi, Milan, Italy
| | | | | | | | - Laura Adang
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
| |
Collapse
|
47
|
Wu D, Fang L, Huang T, Ying S. Case Report: Aicardi-Goutières Syndrome Caused by Novel TREX1 Variants. Front Pediatr 2021; 9:634281. [PMID: 33996686 PMCID: PMC8113616 DOI: 10.3389/fped.2021.634281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 02/18/2021] [Indexed: 11/13/2022] Open
Abstract
TREX1 (three prime repair exonuclease 1) gene encodes DNA 3' end repair exonuclease that plays an important role in DNA repair. Mutations in TREX1 gene have been identified as the cause of a rare autoimmune neurological disease, Aicardi-Goutières syndrome (AGS). Here, we report an AGS case of a 6-month-old Chinese girl with novel TREX1 variants. The patient had mild rashes on the face and legs, increased muscle tensions in the limbs, and positive cervical correction reflex. Cranial magnetic resonance imaging showed that there were patches of slightly longer T1 and T2 signals in the bilateral cerebral hemisphere and brainstem white matter, mainly in the frontotemporal lobe, together with decreased white matter volume, enlarged ventricles, and widened sulcus fissure. Total exon sequencing showed that the TREX1 gene of the child had mutations of c.137_138insC and c.292_293insA, which had not been reported before. In addition, elevated type I interferons were detected by using enzyme-linked immunosorbent assay in the patient's serum. Together, our study demonstrated that novel TREX1 variants (c.137_138insC and c.292_293insA) cause AGS for the first time.
Collapse
Affiliation(s)
- De Wu
- Department of Paediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liwei Fang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Ting Huang
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Songcheng Ying
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
48
|
Wu CC, Peng SSF, Lee WT. Intracerebral large artery disease in Aicardi-Goutières syndrome with TREX1 mutation: a case report. Neurol Sci 2020; 41:3353-3356. [DOI: 10.1007/s10072-020-04516-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/04/2020] [Indexed: 11/30/2022]
|
49
|
Forrer Charlier C, Martins RAP. Protective Mechanisms Against DNA Replication Stress in the Nervous System. Genes (Basel) 2020; 11:E730. [PMID: 32630049 PMCID: PMC7397197 DOI: 10.3390/genes11070730] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/25/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
The precise replication of DNA and the successful segregation of chromosomes are essential for the faithful transmission of genetic information during the cell cycle. Alterations in the dynamics of genome replication, also referred to as DNA replication stress, may lead to DNA damage and, consequently, mutations and chromosomal rearrangements. Extensive research has revealed that DNA replication stress drives genome instability during tumorigenesis. Over decades, genetic studies of inherited syndromes have established a connection between the mutations in genes required for proper DNA repair/DNA damage responses and neurological diseases. It is becoming clear that both the prevention and the responses to replication stress are particularly important for nervous system development and function. The accurate regulation of cell proliferation is key for the expansion of progenitor pools during central nervous system (CNS) development, adult neurogenesis, and regeneration. Moreover, DNA replication stress in glial cells regulates CNS tumorigenesis and plays a role in neurodegenerative diseases such as ataxia telangiectasia (A-T). Here, we review how replication stress generation and replication stress response (RSR) contribute to the CNS development, homeostasis, and disease. Both cell-autonomous mechanisms, as well as the evidence of RSR-mediated alterations of the cellular microenvironment in the nervous system, were discussed.
Collapse
Affiliation(s)
| | - Rodrigo A. P. Martins
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro 21941-902, Brazil;
| |
Collapse
|
50
|
Adang LA, Gavazzi F, Jawad AF, Cusack SV, Kopin K, Peer K, Besnier C, De Simone M, De Giorgis V, Orcesi S, Fazzi E, Galli J, Shults J, Vanderver A. Development of a neurologic severity scale for Aicardi Goutières Syndrome. Mol Genet Metab 2020; 130:153-160. [PMID: 32279991 PMCID: PMC7366613 DOI: 10.1016/j.ymgme.2020.03.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND PURPOSE Aicardi Goutières Syndrome (AGS) is a severe, autoinflammatory leukodystrophy characterized by global neurologic dysfunction. Our goal was to create an easy-to-apply scale relevant to the unique developmental challenges associated with AGS. METHODS All individuals were recruited through our natural history study. Individuals were classified by AGS severity as mild, moderate, or severe, and clinical encounters were assigned a composite score for neurologic function calculated from the sum of three functional classification scales. Through expert consensus, we identified 11 key items to reflect the severity of AGS across gross motor, fine motor, and cognitive skills to create the AGS Scale. There was strong interrater reliability. The AGS scale was applied across available medical records to evaluate neurologic function over time. The AGS scale was compared to performance on a standard measure of gross motor function (Gross Motor Function Measure-88, GMFM-88) and a putative diagnostic biomarker of disease, the interferon signaling gene expression score (ISG). RESULTS The AGS scale score correlated with severity classifications and the composite neurologic function scores. When retrospectively applied across our natural history study, the majority of individuals demonstrated an initial decline in function followed by stable scores. Within the first 6 months of disease, the AGS score was the most dynamic. The AGS scale correlated with performance by the GMFM-88, but did not correlate with ISG levels. CONCLUSIONS This study demonstrates the utility of the AGS scale as a multimodal tool for the assessment of neurologic function in AGS. The AGS scale correlates with clinical severity and with a more labor-intensive tool, GMFM-88. This study underscores the limitations of the ISG score as a marker of disease severity. With the AGS scale, we found that AGS neurologic severity is the most dynamic early in disease. This novel AGS scale is a promising tool to longitudinally follow neurologic function in this unique population.
Collapse
Affiliation(s)
- Laura A Adang
- Division of Neurology, Children's Hospital of Philadelphia, United States.
| | - Francesco Gavazzi
- Division of Neurology, Children's Hospital of Philadelphia, United States
| | - Abbas F Jawad
- Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at University of Pennsylvania, United States
| | - Stacy V Cusack
- Division of Occupational Therapy, Children's Hospital of Philadelphia, United States
| | - Kimberly Kopin
- Division of Physical Therapy, Children's Hospital of Philadelphia, United States
| | - Kyle Peer
- Division of Neurology, Children's Hospital of Philadelphia, United States
| | - Constance Besnier
- Division of Neurology, Children's Hospital of Philadelphia, United States
| | - Micaela De Simone
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, Italy
| | - Valentina De Giorgis
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy
| | - Simona Orcesi
- Department of Child Neurology and Psychiatry, IRCCS Mondino Foundation, Pavia, Italy
| | - Elisa Fazzi
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, Italy
| | - Jessica Galli
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, Italy
| | - Justine Shults
- Department of Biostatistics, Perelman School of Medicine at University of Pennsylvania, United States
| | - Adeline Vanderver
- Division of Neurology, Children's Hospital of Philadelphia, United States
| |
Collapse
|