1
|
Bregant E, Betto E, Dal Secco C, Zucco J, Baldan F, Allegri L, Lonigro IR, Faletra F, Verriello L, Damante G, Mio C. The molecular landscape of hereditary ataxia: a single-center study. Hum Genet 2025; 144:545-557. [PMID: 40208338 PMCID: PMC12033174 DOI: 10.1007/s00439-025-02744-y] [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: 10/23/2024] [Accepted: 04/02/2025] [Indexed: 04/11/2025]
Abstract
Hereditary ataxia (HA) is a heterogeneous group of complex neurological disorders, which represent a diagnostic challenge due to their diverse phenotypes and genetic etiologies. Next-generation sequencing (NGS) has revolutionized the field of neurogenetics, improving the identification of ataxia-associated genes. Notwithstanding, repeat expansions analysis remains a cornerstone in the diagnostic workflow of these diseases. Here we describe the molecular characterization of a consecutive single-center series of 70 patients with genetically uncharacterized HA. Patients' samples were analyzed for known HA-associated repeat expansions as first tier and negative ones were analyzed by whole exome sequencing (WES) as second tier. Overall, we identified pathogenic/likely pathogenic variants in 40% (n = 28/70) and variants of unknown significance (VUS) in 20% (n = 14/70) of cases. In particular, 10 patients (14.3%, n = 10/70) presented pathogenic repeat expansions while 18 cases (30%, n = 18/60) harbored at least a single nucleotide variant (SNV) or a copy number variant (CNV) in HA or HSP-related genes. WES allowed assessing complex neurological diseases (i.e., leukodystrophies, cerebrotendinous xanthomatosis and atypical xeroderma pigmentosum), which are not usually referred as pure genetic ataxias. Our data suggests that the combined use of repeat expansion analysis and WES, coupled to detailed clinical phenotyping, is able to detect the molecular alteration underpinning ataxia in almost 50% cases, regardless of the hereditary pattern. Indeed, NGS-based tests are fundamental to acknowledge novel HA-associated genes useful to explain the remaining wide fraction of negative tests. Nowadays, this gap is problematic since these patients could not benefit from an etiological diagnosis of their disease that allows prognostic trajectories and prenatal/preimplantation diagnosis.
Collapse
Affiliation(s)
- Elisa Bregant
- Institute of Medical Genetics, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Elena Betto
- Institute of Medical Genetics, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Chiara Dal Secco
- Department of Medicine (DMED), University of Udine, Via Chiusaforte ingresso E, 33100, Udine, Italy
| | - Jessica Zucco
- Institute of Medical Genetics, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Federica Baldan
- Department of Medicine (DMED), University of Udine, Via Chiusaforte ingresso E, 33100, Udine, Italy
| | - Lorenzo Allegri
- Department of Medicine (DMED), University of Udine, Via Chiusaforte ingresso E, 33100, Udine, Italy
| | | | - Flavio Faletra
- Institute of Medical Genetics, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
- Department of Medicine (DMED), University of Udine, Via Chiusaforte ingresso E, 33100, Udine, Italy
| | - Lorenzo Verriello
- Neurology Unit, Department of Neurosciences, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
| | - Giuseppe Damante
- Institute of Medical Genetics, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC), Udine, Italy
- Department of Medicine (DMED), University of Udine, Via Chiusaforte ingresso E, 33100, Udine, Italy
| | - Catia Mio
- Department of Medicine (DMED), University of Udine, Via Chiusaforte ingresso E, 33100, Udine, Italy.
| |
Collapse
|
2
|
Zhou L, Wang Y, Xu Y, Zhang Y, Zhu C. Advances in AAV-mediated gene replacement therapy for pediatric monogenic neurological disorders. Mol Ther Methods Clin Dev 2024; 32:101357. [PMID: 39559557 PMCID: PMC11570947 DOI: 10.1016/j.omtm.2024.101357] [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] [Indexed: 11/20/2024]
Abstract
Pediatric monogenetic diseases encompass a spectrum of debilitating neurological disorders that affect infants and children, often resulting in profound cognitive and motor impairments. Gene replacement therapy holds immense promise in addressing the underlying genetic defects responsible for these conditions. Adeno-associated virus (AAV) vectors have emerged as a leading platform for delivering therapeutic genes due to their safety profile and ability to transduce various cell types, including neurons. This review highlights recent advancements in AAV-mediated gene replacement therapy for pediatric monogenetic diseases, focusing on key preclinical and clinical studies. We discuss various strategies to enhance transduction efficiency, target specificity, and safety. Furthermore, we explore challenges such as immune responses, along with innovative approaches to overcome these obstacles. Moreover, we examine the clinical outcomes and safety profiles of AAV-based gene therapies in pediatric patients, providing insights into the feasibility and efficacy of these interventions. Finally, we discuss future directions and potential avenues for further research to optimize the therapeutic potential of AAV-delivered gene replacement therapy for pediatric encephalopathies, ultimately aiming to improve the quality of life for affected individuals and their families.
Collapse
Affiliation(s)
- Livia Zhou
- Henan Neurodevelopment Engineering Research Center for Children, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital Zhengzhou Children’s Hospital, Zhengzhou 450018, China
| | - Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for Children, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital Zhengzhou Children’s Hospital, Zhengzhou 450018, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yaodong Zhang
- Henan Neurodevelopment Engineering Research Center for Children, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital Zhengzhou Children’s Hospital, Zhengzhou 450018, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
3
|
Peng H, Lam YW, Zhou Z, Herdt AR, Gelb MH, Lee CW. Expression study of Krabbe Disease GALC missense variants - Insights from quantification profiles of residual enzyme activity, secretion and psychosine levels. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618938. [PMID: 39464077 PMCID: PMC11507934 DOI: 10.1101/2024.10.17.618938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Krabbe disease (KD) is an autosomal recessive lysosomal storage disorder caused by loss-of-function mutations in the GALC gene, which encodes for the enzyme galactosylceramidase (GALC). GALC is crucial for myelin metabolism. Functional deficiency of GALC leads to toxic accumulation of psychosine, dysfunction and death of oligodendrocytes, and eventual brain demyelination. To date, 46 clinically-relevant, pathogenic GALC missense mutations (MMs) have been identified in KD patients. These MMs are present in ∼70% of KD cases reported over 8 published studies between 1996 - 2019. However, the mechanisms by which these MMs lead to GALC functional deficiency and their correlations with clinical phenotype remain poorly understood. To address this, we generated a GALC -knockout human oligodendrocytic cell line (MO3.13/ GALC -KO) using CRISPR-Cas9 method to assess GALC function and GALC secretion. We evaluated 5 polymorphic and 31 clinically-relevant MM variants (MMVs) using transient expression assays. Our results showed that 26 MMVs, including 10 co-variants with p.I562T, reduced GALC activity by 92% - 100% compared to wild-type GALC (WT-GALC). MMVs from infantile-onset KD patients produced < 2% of WT activity, whereas those associated with juvenile- and adult-onset cases retained up to 7% of WT activity. Residual GALC activity was correlated with mature, lysosomal GALC protein levels (Pearson r = 0.93, P<0.0001). Many low-activity MMVs did not correspondingly impair GALC secretion. Twenty-one of the 26 low-activity MMVs showed a 21% - 100% reduction in sec-GALC levels, indicating varying degrees of GALC mis-trafficking among these variants. Importantly, GALC activity among MMVs strongly correlates with clinical disease severity, based on the age of symptom onset in patients with either homozygous MM (Pearson r = 0.98, P<0.0001, n = 7) or compound heterozygous (Pearson r = 0.94, P<0.0001, n = 12) MM-null mutation genotypes. Thus, our data suggests that GALC activity could serve as a prognostic disease indicator under specific experimental conditions. We further investigated the impact of pathogenic MMVs on psychosine accumulation, a key biomarker for KD. Psychosine levels were 21-fold higher in mock control cells compared to WT-GALC transfected cells (mock = 0.349 pmol/mg, WT-GALC = 0.016 pmol/mg), but negatively correlated with GALC activity among pathogenic MMVs (Pearson r = -0.63, P < 0.01, n = 15). Although psychosine levels were higher in most MMVs associated with infantile-onset KD, no significant correlations with clinical onset were detected. Overall, our study provides a comprehensive quantitative analysis of the functional deficits and mis-trafficking associated with clinically-relevant GALC MMVs, enhancing our understanding of the molecular genetics and genotype-phenotype correlations of the GALC gene in Krabbe disease.
Collapse
|
4
|
Takahashi H, Perez-Canamas A, Lee CW, Ye H, Han X, Strittmatter SM. Lysosomal TMEM106B interacts with galactosylceramidase to regulate myelin lipid metabolism. Commun Biol 2024; 7:1088. [PMID: 39237682 PMCID: PMC11377756 DOI: 10.1038/s42003-024-06810-5] [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: 07/21/2023] [Accepted: 08/30/2024] [Indexed: 09/07/2024] Open
Abstract
TMEM106B is an endolysosomal transmembrane protein not only associated with multiple neurological disorders including frontotemporal dementia, Alzheimer's disease, and hypomyelinating leukodystrophy but also potentially involved in COVID-19. Additionally, recent studies have identified amyloid fibrils of C-terminal TMEM106B in both aged healthy and neurodegenerative brains. However, so far little is known about physiological functions of TMEM106B in the endolysosome and how TMEM106B is involved in a wide range of human conditions at molecular levels. Here, we performed lipidomic analysis of the brain of TMEM106B-deficient mice. We found that TMEM106B deficiency significantly decreases levels of two major classes of myelin lipids, galactosylceramide and its sulfated derivative sulfatide. Subsequent co-immunoprecipitation assay showed that TMEM106B physically interacts with galactosylceramidase. We also found that galactosylceramidase activity was significantly increased in TMEM106B-deficient brains. Thus, our results suggest that TMEM106B interacts with galactosylceramidase to regulate myelin lipid metabolism and have implications for TMEM106B-associated diseases.
Collapse
Affiliation(s)
- Hideyuki Takahashi
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Azucena Perez-Canamas
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Chris W Lee
- Biomedical Research Institute of New Jersey (BRInj), Cedar Knolls, NJ, 07927, USA
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ, 07960, USA
- Atlantic Health System, Morristown, NJ, 07960, USA
| | - Hongping Ye
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA.
| |
Collapse
|
5
|
Saldivia N, Heller G, Zelada D, Whitehair J, Venkat N, Konjeti A, Savitzky R, Samano S, Simchuk D, van Breemen R, Givogri MI, Bongarzone ER. Deficiency of galactosyl-ceramidase in adult oligodendrocytes worsens disease severity during chronic experimental allergic encephalomyelitis. Mol Ther 2024; 32:3163-3176. [PMID: 38937968 PMCID: PMC11403238 DOI: 10.1016/j.ymthe.2024.06.035] [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: 12/14/2023] [Revised: 04/24/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024] Open
Abstract
Galactosyl-ceramidase (GALC) is a ubiquitous lysosomal enzyme crucial for the correct myelination of the mammalian nervous system during early postnatal development. However, the physiological consequence of GALC deficiency in the adult brain remains unknown. In this study, we found that mice with conditional ablation of GALC activity in post-myelinating oligodendrocytes were lethally sensitized when challenged with chronic experimental allergic encephalomyelitis (EAE), in contrast with the non-lethal dysmyelination observed in Galc-ablated mice without the EAE challenge. Mechanistically, we found strong inflammatory demyelination without remyelination and an impaired fusion of lysosomes and autophagosomes with accumulation of myelin debris after a transcription factor EB-dependent increase in the lysosomal autophagosome flux. These results indicate that the physiological impact of GALC deficiency is highly influenced by the cell context (oligodendroglial vs. global expression), the presence of inflammation, and the developmental time when it happens (pre-myelination vs. post-myelination). We conclude that Galc expression in adult oligodendrocytes is crucial for the maintenance of adult central myelin and to decrease vulnerability to additional demyelinating insults.
Collapse
Affiliation(s)
- Natalia Saldivia
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| | - Gregory Heller
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Diego Zelada
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jason Whitehair
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Nikhil Venkat
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ashna Konjeti
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Reina Savitzky
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Shayla Samano
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Daniel Simchuk
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | | | - Maria I Givogri
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
6
|
Ahmad A, Khan JM, Paray BA, Rashid K, Parvez A. Endolysosomal trapping of therapeutics and endosomal escape strategies. Drug Discov Today 2024; 29:104070. [PMID: 38942071 DOI: 10.1016/j.drudis.2024.104070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/31/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
Internalizing therapeutic molecules or genes into cells and safely delivering them to the target tissue where they can perform the intended tasks is one of the key characteristics of the smart gene/drug delivery vector. Despite much research in this field, endosomal escape continues to be a significant obstacle to the development of effective gene/drug delivery systems. In this review, we discuss in depth the several types of endocytic pathways involved in the endolysosomal trapping of therapeutic agents. In addition, we describe numerous mechanisms involved in nanoparticle endosomal escape. Furthermore, many other techniques are employed to increase endosomal escape to minimize entrapment of therapeutic compounds within endolysosomes, which have been reviewed at length in this study.
Collapse
Affiliation(s)
- Aqeel Ahmad
- Department of Medical Biochemistry, College of Medicine, Shaqra University, Shaqra 11961, Saudi Arabia.
| | - Javed Masood Khan
- Department of Food Science and Nutrition, Faculty of Food and Agricultural Sciences, King Saud University, 2460, Riyadh 11451, Saudi Arabia
| | - Bilal Ahamad Paray
- Department of Zoology, College of Science, King Saud University, PO Box 2455, Riyadh 11451, Saudi Arabia
| | - Khalid Rashid
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ashib Parvez
- Department of Community Medicine, F.H. Medical College, Atal Bihari Vajpayee Medical University, Etmadpur, Agra, India
| |
Collapse
|
7
|
Ketata I, Ellouz E. From pathological mechanisms in Krabbe disease to cutting-edge therapy: A comprehensive review. Neuropathology 2024; 44:255-277. [PMID: 38444347 DOI: 10.1111/neup.12967] [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: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/07/2024]
Abstract
Since its initial documentation by Knud Krabbe in 1916, numerous studies have scrutinized the characteristics of Krabbe disease (KD) until the identification of the mutation in the GALC gene. In alignment with that, we investigated the natural history of KD spanning eight decades to gain a deeper understanding of the evolutionary trajectory of its mechanisms. Through our comprehensive analysis, we unearthed additional novel elements in molecular biology involving the micropathological mechanism of the disease. This review offers an updated perspective on the metabolic disorder that defines KD. Recently, extracellular vesicles (EVs), autophagy impairment, and α-synuclein have emerged as pivotal players in the neuropathological processes. EVs might serve as a cellular mechanism to avoid or alleviate the detrimental impacts of excessive toxic psychosine levels, and extracting EVs could contribute to synapse dysfunction. Autophagy impairment was found to be independent of psychosine and reliant on AKT and B-cell lymphoma 2. Additionally, α-synuclein has been recognized for inducing cellular death and dysfunction in common biological pathways. Our objective is to assess the effectiveness of advanced therapies in addressing this particular condition. While hematopoietic stem cells have been a primary treatment, its administration proves challenging, particularly in the presymptomatic phase. In this review, we have compiled information from over 10 therapy trials, comparing them based on their benefits and disadvantage.
Collapse
Affiliation(s)
- Imen Ketata
- Neurology Department, University Hospital of Gabes, Gabes, Tunisia
- Sfax University, Sfax Faculty of Medicine, Sfax, Tunisia
| | - Emna Ellouz
- Neurology Department, University Hospital of Gabes, Gabes, Tunisia
- Sfax University, Sfax Faculty of Medicine, Sfax, Tunisia
| |
Collapse
|
8
|
Sevin C, Mochel F. Hematopoietic stem cell transplantation in leukodystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:355-366. [PMID: 39322389 DOI: 10.1016/b978-0-323-99209-1.00017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
More than 50 leukodystrophies have been described. This group of inherited disorders affects myelin development and/or maintenance and can manifest from birth to adulthood. Neuroinflammation is a hallmark of some leukodystrophies, explaining in part the therapeutic benefit of hematopoietic stem cell transplantation (HSCT). Indeed, in addition to supplying the CNS with myelomonocyte donor cells expressing the deficient protein or enzyme, HSCT allows the restoration of normal microglia function, which may act on neuroinflammation. In this chapter, we explore the rationale, indication, and outcome of HSCT in Cerebral Adrenoleukodystrophy (CALD), Metachromatic Leukodystrophy (MLD), Krabbe Disease (KD), and Adult-onset Leukoencephalopathy with Axonal Spheroids and Pigmented Glia (ALSP), which are among the most frequent leukodystrophies. For these leukodystrophies, HSCT may modify notably the natural history and improve CNS-related deficits, provided that the procedure is performed early into the disease course. In addition, we discuss the recent development of ex vivo gene therapy for CALD and MLD as a promising alternative to allograft.
Collapse
Affiliation(s)
- Caroline Sevin
- AP-HP, Kremlin-Bicêtre University Hospital, Department of Neuropediatrics, Reference Center for Pediatric Leukodystrophies, Paris, France; INSERM U 1127, CNRS UMR 7225, Sorbonne Université, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau, ICM, Paris, France
| | - Fanny Mochel
- INSERM U 1127, CNRS UMR 7225, Sorbonne Université, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau, ICM, Paris, France; AP-HP, Pitié-Salpêtrière University Hospital, Department of Medical Genetics, Reference Centers for Adult Neurometabolic Diseases and Adult Leukodystrophies, Paris, France.
| |
Collapse
|
9
|
Maghazachi AA. Globoid Cell Leukodystrophy (Krabbe Disease): An Update. Immunotargets Ther 2023; 12:105-111. [PMID: 37928748 PMCID: PMC10625317 DOI: 10.2147/itt.s424622] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023] Open
Abstract
Globoid cell leukodystrophy or Krabbe is a disease that affects children as well as adults who have mutations in the gene encoding the enzyme galactosylceramidase/galctocerebrosidase (GALC), resulting in the deposition of the toxic lipid D-galactosyl-beta1-1' sphingosine (GalSph or psychosine). Several therapeutic modalities were used to treat patients with Krabbe disease, including hematopoietic stem cell transplantation, enzyme replacement therapy, autophagy activators, intravenous immunoglobulin, and inhibitors of the Pyroptosis process, among many other approaches. In this article, I will briefly discuss the disease in both human and animal model, describe recent clinical observations as well as methods utilizing genetic analysis for diagnosis, and finally review recent advances in treating this rare and devastating disease.
Collapse
|
10
|
Sutter PA, Ménoret A, Jellison ER, Nicaise AM, Bradbury AM, Vella AT, Bongarzone ER, Crocker SJ. CD8+ T cell depletion prevents neuropathology in a mouse model of globoid cell leukodystrophy. J Exp Med 2023; 220:e20221862. [PMID: 37310382 PMCID: PMC10266545 DOI: 10.1084/jem.20221862] [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: 10/31/2022] [Revised: 04/10/2023] [Accepted: 05/26/2023] [Indexed: 06/14/2023] Open
Abstract
Globoid cell leukodystrophy (GLD) or Krabbe's disease is a fatal genetic demyelinating disease of the central nervous system caused by loss-of-function mutations in the galactosylceramidase (galc) gene. While the metabolic basis for disease is known, the understanding of how this results in neuropathology is not well understood. Herein, we report that the rapid and protracted elevation of CD8+ cytotoxic T lymphocytes occurs coincident with clinical disease in a mouse model of GLD. Administration of a function-blocking antibody against CD8α effectively prevented disease onset, reduced morbidity and mortality, and prevented CNS demyelination in mice. These data indicate that subsequent to the genetic cause of disease, neuropathology is driven by pathogenic CD8+ T cells, thus offering novel therapeutic potential for treatment of GLD.
Collapse
Affiliation(s)
- Pearl A. Sutter
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Antoine Ménoret
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Evan R. Jellison
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Alexandra M. Nicaise
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Clinical Neuroscience and National Institute for Health Research Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Allison M. Bradbury
- Department of Pediatrics, Nationwide Children's Hospital, Ohio State University, Columbus, OH, USA
| | - Anthony T. Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Ernesto R. Bongarzone
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Stephen J. Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
11
|
Herdt AR, Peng H, Dickson DW, Golde TE, Eckman EA, Lee CW. Brain Targeted AAV1-GALC Gene Therapy Reduces Psychosine and Extends Lifespan in a Mouse Model of Krabbe Disease. Genes (Basel) 2023; 14:1517. [PMID: 37628569 PMCID: PMC10454254 DOI: 10.3390/genes14081517] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Krabbe disease (KD) is a progressive and devasting neurological disorder that leads to the toxic accumulation of psychosine in the white matter of the central nervous system (CNS). The condition is inherited via biallelic, loss-of-function mutations in the galactosylceramidase (GALC) gene. To rescue GALC gene function in the CNS of the twitcher mouse model of KD, an adeno-associated virus serotype 1 vector expressing murine GALC under control of a chicken β-actin promoter (AAV1-GALC) was administered to newborn mice by unilateral intracerebroventricular injection. AAV1-GALC treatment significantly improved body weight gain and survival of the twitcher mice (n = 8) when compared with untreated controls (n = 5). The maximum weight gain after postnatal day 10 was significantly increased from 81% to 217%. The median lifespan was extended from 43 days to 78 days (range: 74-88 days) in the AAV1-GALC-treated group. Widespread expression of GALC protein and alleviation of KD neuropathology were detected in the CNS of the treated mice when examined at the moribund stage. Functionally, elevated levels of psychosine were completely normalized in the forebrain region of the treated mice. In the posterior region, which includes the mid- and the hindbrain, psychosine was reduced by an average of 77% (range: 53-93%) compared to the controls. Notably, psychosine levels in this region were inversely correlated with body weight and lifespan of AAV1-GALC-treated mice, suggesting that the degree of viral transduction of posterior brain regions following ventricular injection determined treatment efficacy on growth and survivability, respectively. Overall, our results suggest that viral vector delivery via the cerebroventricular system can partially correct psychosine accumulation in brain that leads to slower disease progression in KD.
Collapse
Affiliation(s)
- Aimee R. Herdt
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Hui Peng
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Todd E. Golde
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA 30322, USA
- Department of Neurology, Emory University, Atlanta, GA 30322, USA
- Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Elizabeth A. Eckman
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Chris W. Lee
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| |
Collapse
|