1
|
Kolstad J, Zoppo C, Johnston JM, D'Souza P, Kühn AL, Vardar Z, Peker A, Hader A, Celik H, Lewis CJ, Lindsay C, Rentiya ZS, Lebel C, Vedantham S, Vachha B, Gray-Edwards HL, Acosta MT, Tifft CJ, Shazeeb MS. Natural history progression of MRI brain volumetrics in type II late-infantile and juvenile GM1 gangliosidosis patients. Mol Genet Metab 2025; 144:109025. [PMID: 39874851 DOI: 10.1016/j.ymgme.2025.109025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 01/14/2025] [Indexed: 01/30/2025]
Abstract
OBJECTIVE GM1 gangliosidosis is a rare lysosomal storage disorder characterized by the accumulation of GM1 gangliosides in neuronal cells, resulting in severe neurodegeneration. Currently, limited data exists on the brain volumetric changes associated with this disease. This study focuses on the late-infantile and juvenile subtypes of type II GM1 gangliosidosis, aiming to quantify brain volumetric characteristics to track disease progression. METHODS Brain volumetric analysis was conducted on 56 MRI scans from 24 type II GM1 patients (8 late-infantile and 16 juvenile) and 19 healthy controls over multiple time points. The analysis included the use of semi-automated segmentation of the whole brain, ventricles, cerebellum, corpus callosum, thalamus, caudate, and lentiform nucleus. A generalized linear model was used to compare the volumetric measurements between the patient groups and healthy controls, accounting for age as a confounding factor. RESULTS Both late-infantile and juvenile GM1 patients exhibited significant whole-brain atrophy compared to healthy controls, even after adjusting for age. Notably, the late-infantile subtype displayed more pronounced atrophy in the cerebellum, thalamus, and corpus callosum compared to the juvenile subtype. Both late-infantile and juvenile subtypes showed significantly higher ventricular volumes and a significant reduction in all other structure volumes compared to the healthy controls. The volumetric measurements also correlated well with disease severity based on clinical metrics. CONCLUSIONS The findings underscore the distinct brain volumetrics of the late-infantile and juvenile subtypes of GM1 gangliosidosis compared to healthy controls. These quantifications can be used as reliable imaging biomarkers to track disease progression and evaluate responses to therapeutic interventions.
Collapse
Affiliation(s)
- Josephine Kolstad
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA; McLean Hospital, Belmont, MA, USA
| | - Christopher Zoppo
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jean M Johnston
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Precilla D'Souza
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anna Luisa Kühn
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Zeynep Vardar
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Asma Hader
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Hakki Celik
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Connor J Lewis
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Clifford Lindsay
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Zubir S Rentiya
- Department of Radiation Oncology & Radiology, University of Virginia, Charlottesville, VA, USA
| | - Catherine Lebel
- Department of Radiology, University of Calgary, Alberta, Canada
| | | | - Behroze Vachha
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Division of Neuroradiology, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Heather L Gray-Edwards
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Maria T Acosta
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Cynthia J Tifft
- Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mohammed Salman Shazeeb
- Image Processing & Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
2
|
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]
|
3
|
Zoppo C, Kolstad J, Johnston J, D'Souza P, Kühn AL, Vardar Z, Peker A, Lindsay C, Rentiya ZS, King R, Gray-Edwards H, Vachha B, Acosta MT, Tifft CJ, Shazeeb MS. Quantitative reliability assessment of brain MRI volumetric measurements in type II GM1 gangliosidosis patients. FRONTIERS IN NEUROIMAGING 2024; 3:1410848. [PMID: 39350771 PMCID: PMC11440193 DOI: 10.3389/fnimg.2024.1410848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/22/2024] [Indexed: 10/04/2024]
Abstract
Purpose GM1-gangliosidosis (GM1) leads to extensive neurodegenerative changes and atrophy that precludes the use of automated MRI segmentation techniques for generating brain volumetrics. We developed a standardized segmentation protocol for brain MRIs of patients with type II GM1 and then assessed the inter- and intra-rater reliability of this methodology. The volumetric data may be used as a biomarker of disease burden and progression, and standardized methodology may support research into the natural history of the disease which is currently lacking in the literature. Approach Twenty-five brain MRIs were included in this study from 22 type II GM1 patients of which 8 were late-infantile subtype and 14 were juvenile subtype. The following structures were segmented by two rating teams on a slice-by-slice basis: whole brain, ventricles, cerebellum, lentiform nucleus, thalamus, corpus callosum, and caudate nucleus. The inter- and intra-rater reliability of the segmentation method was assessed with an intraclass correlation coefficient as well as Sorensen-Dice and Jaccard coefficients. Results Based on the Sorensen-Dice and Jaccard coefficients, the inter- and intra-rater reliability of the segmentation method was significantly better for the juvenile patients compared to late-infantile (p < 0.01). In addition, the agreement between the two rater teams and within themselves can be considered good with all p-values < 0.05. Conclusions The standardized segmentation approach described here has good inter- and intra-rater reliability and may provide greater accuracy and reproducibility for neuromorphological studies in this group of patients and help to further expand our understanding of the natural history of this disease.
Collapse
Affiliation(s)
- Christopher Zoppo
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Josephine Kolstad
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Jean Johnston
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Precilla D'Souza
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Anna Luisa Kühn
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Zeynep Vardar
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Ahmet Peker
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Clifford Lindsay
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Zubir S. Rentiya
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Robert King
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Heather Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Behroze Vachha
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
- Division of Neuroradiology, Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Maria T. Acosta
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Cynthia J. Tifft
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mohammed Salman Shazeeb
- Image Processing and Analysis Core (iPAC), Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, United States
| |
Collapse
|
4
|
D'Souza P, Farmer C, Johnston JM, Han ST, Adams D, Hartman AL, Zein W, Huryn LA, Solomon B, King K, Jordan CP, Myles J, Nicoli ER, Rothermel CE, Mojica Algarin Y, Huang R, Quimby R, Zainab M, Bowden S, Crowell A, Buckley A, Brewer C, Regier DS, Brooks BP, Acosta MT, Baker EH, Vézina G, Thurm A, Tifft CJ. GM1 gangliosidosis type II: Results of a 10-year prospective study. Genet Med 2024; 26:101144. [PMID: 38641994 PMCID: PMC11348282 DOI: 10.1016/j.gim.2024.101144] [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: 01/10/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024] Open
Abstract
PURPOSE GM1 gangliosidosis (GM1) a lysosomal disorder caused by pathogenic variants in GLB1, is characterized by relentless neurodegeneration. There are no approved treatments. METHODS Forty-one individuals with type II (late-infantile and juvenile) GM1 participated in a single-site prospective observational study. RESULTS Classification of 37 distinct variants using American College of Medical Genetics and Genomics criteria resulted in the upgrade of 6 and the submission of 4 new variants. In contrast to type I infantile disease, children with type II had normal or near normal hearing and did not have cherry-red maculae or hepatosplenomegaly. Some older children with juvenile onset disease developed thickened aortic and/or mitral valves. Serial magnetic resonance images demonstrated progressive brain atrophy, more pronounced in late infantile patients. Magnetic resonance spectroscopy showed worsening elevation of myo-inositol and deficit of N-acetyl aspartate that were strongly correlated with scores on the Vineland Adaptive Behavior Scale, progressing more rapidly in late infantile compared with juvenile onset disease. CONCLUSION Serial phenotyping of type II GM1 patients expands the understanding of disease progression and clarifies common misconceptions about type II patients; these are pivotal steps toward more timely diagnosis and better supportive care. The data amassed through this 10-year effort will serve as a robust comparator for ongoing and future therapeutic trials.
Collapse
Affiliation(s)
- Precilla D'Souza
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Cristan Farmer
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, Bethesda, MD
| | - Jean M Johnston
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Sangwoo T Han
- Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - David Adams
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD
| | - Adam L Hartman
- Division of Clinical Research, National Institute of Neurological Disorders and Stroke, Bethesda, MD
| | - Wadih Zein
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, MD
| | - Laryssa A Huryn
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, MD
| | - Beth Solomon
- Rehabilitation Medicine Department, Warren C. Magnuson Clinical Research Center, Bethesda, MD
| | - Kelly King
- Neurology Branch, National Institute on Deafness and Other Communication Disorders, Bethesda, MD
| | | | - Jennifer Myles
- Nutrition Department, Warren C. Magnuson Clinical Research Center, Bethesda, MD
| | - Elena-Raluca Nicoli
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Caroline E Rothermel
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Yoliann Mojica Algarin
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Reyna Huang
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Rachel Quimby
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Mosufa Zainab
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Sarah Bowden
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Anna Crowell
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD
| | - Ashura Buckley
- Sleep and Neurodevelopment Service, National Institute of Mental Health, Bethesda, MD
| | - Carmen Brewer
- Neurology Branch, National Institute on Deafness and Other Communication Disorders, Bethesda, MD
| | - Debra S Regier
- Genetics and Metabolism, Children's National Hospital, Washington, DC
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, Bethesda, MD
| | - Maria T Acosta
- Undiagnosed Disease Program, National Human Genome Research Institute, Bethesda, MD
| | - Eva H Baker
- Department of Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, MD
| | - Gilbert Vézina
- Program in Neuroradiology and Program in Radiology, Children's National Hospital, Washington, DC; Radiology and Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, Bethesda, MD
| | - Cynthia J Tifft
- Office of the Clinical Director, National Human Genome Research Institute, Bethesda, MD; Medical Genetics Branch, National Human Genome Research Institute, Bethesda, MD.
| |
Collapse
|
5
|
Jerves Serrano T, Gold J, Cooper JA, Church HJ, Tylee KL, Wu HY, Kim SY, Stepien KM. Hepatomegaly and Splenomegaly: An Approach to the Diagnosis of Lysosomal Storage Diseases. J Clin Med 2024; 13:1465. [PMID: 38592278 PMCID: PMC10932313 DOI: 10.3390/jcm13051465] [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: 02/04/2024] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 04/10/2024] Open
Abstract
Clinical findings of hepatomegaly and splenomegaly, the abnormal enlargement of the liver and spleen, respectively, should prompt a broad differential diagnosis that includes metabolic, congestive, neoplastic, infectious, toxic, and inflammatory conditions. Among the metabolic diseases, lysosomal storage diseases (LSDs) are a group of rare and ultrarare conditions with a collective incidence of 1 in 5000 live births. LSDs are caused by genetic variants affecting the lysosomal enzymes, transporters, or integral membrane proteins. As a result, abnormal metabolites accumulate in the organelle, leading to dysfunction. Therapeutic advances, including early diagnosis and disease-targeted management, have improved the life expectancy and quality of life of people affected by certain LSDs. To access these new interventions, LSDs must be considered in patients presenting with hepatomegaly and splenomegaly throughout the lifespan. This review article navigates the diagnostic approach for individuals with hepatosplenomegaly particularly focusing on LSDs. We provide hints in the history, physical exam, laboratories, and imaging that may identify LSDs. Additionally, we discuss molecular testing, arguably the preferred confirmatory test (over biopsy), accompanied by enzymatic testing when feasible.
Collapse
Affiliation(s)
| | - Jessica Gold
- Division of Genetics, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - James A. Cooper
- Willink Biochemical Genetics Laboratory, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK; (J.A.C.); (H.J.C.); (K.L.T.); (H.Y.W.)
| | - Heather J. Church
- Willink Biochemical Genetics Laboratory, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK; (J.A.C.); (H.J.C.); (K.L.T.); (H.Y.W.)
| | - Karen L. Tylee
- Willink Biochemical Genetics Laboratory, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK; (J.A.C.); (H.J.C.); (K.L.T.); (H.Y.W.)
| | - Hoi Yee Wu
- Willink Biochemical Genetics Laboratory, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK; (J.A.C.); (H.J.C.); (K.L.T.); (H.Y.W.)
| | - Sun Young Kim
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH 45219, USA;
| | - Karolina M. Stepien
- Salford Royal Organization, Northern Care Alliance NHS Foundation Trust, Adult Inherited Metabolic Diseases Department, Salford M6 8HD, UK
- Division of Cardiovascular Sciences, University of Manchester, Manchester M13 9PL, UK
| |
Collapse
|
6
|
Májovská J, Nestrašil I, Ahmed A, Bondy MT, Klempíř J, Jahnová H, Schneider SA, Horáková D, Krásenský J, Ješina P, Vaneckova M, Nascene DR, Whitley CB, Jarnes JR, Magner M, Dušek P. Quantitative brain morphometry identifies cerebellar, cortical, and subcortical gray and white matter atrophy in late-onset Tay-Sachs disease. J Inherit Metab Dis 2024; 47:327-339. [PMID: 38112342 PMCID: PMC10947897 DOI: 10.1002/jimd.12700] [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: 09/04/2023] [Revised: 11/08/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Cerebellar atrophy is a characteristic sign of late-onset Tay-Sachs disease (LOTS). Other structural neuroimaging abnormalities are inconsistently reported. Our study aimed to perform a detailed whole-brain analysis and quantitatively characterize morphometric changes in LOTS patients. Fourteen patients (8 M/6F) with LOTS from three centers were included in this retrospective study. For morphometric brain analyses, we used deformation-based morphometry, voxel-based morphometry, surface-based morphometry, and spatially unbiased cerebellar atlas template. The quantitative whole-brain morphometric analysis confirmed the finding of profound pontocerebellar atrophy with most affected cerebellar lobules V and VI in LOTS patients. Additionally, the atrophy of structures mainly involved in motor control, including bilateral ventral and lateral thalamic nuclei, primary motor and sensory cortex, supplementary motor area, and white matter regions containing corticospinal tract, was present. The atrophy of the right amygdala, hippocampus, and regions of occipital, parietal and temporal white matter was also observed in LOTS patients in contrast with controls (p < 0.05, FWE corrected). Patients with dysarthria and those initially presenting with ataxia had more severe cerebellar atrophy. Our results show predominant impairment of cerebellar regions responsible for speech and hand motor function in LOTS patients. Widespread morphological changes of motor cortical and subcortical regions and tracts in white matter indicate abnormalities in central motor circuits likely coresponsible for impaired speech and motor function.
Collapse
Affiliation(s)
- Jitka Májovská
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital Prague, Czech Republic
| | - Igor Nestrašil
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Alia Ahmed
- Advanced Therapies Program, Division of Genetics and Metabolism, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Monica T Bondy
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Jiří Klempíř
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Helena Jahnová
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital Prague, Czech Republic
| | | | - Dana Horáková
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Jan Krásenský
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Pavel Ješina
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital Prague, Czech Republic
| | - Manuela Vaneckova
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - David R Nascene
- Department of Neuroradiology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Chester B Whitley
- Advanced Therapies Program, Division of Genetics and Metabolism, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
- Gene Therapy and Diagnostic Laboratory, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
- Department of Experimental and Clinical Pharmacology, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Jeanine R Jarnes
- Advanced Therapies Program, Division of Genetics and Metabolism, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, MN, USA
- Department of Experimental and Clinical Pharmacology, University of Minnesota, College of Pharmacy, Minneapolis, MN, USA
| | - Martin Magner
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital Prague, Czech Republic
| | - Petr Dušek
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| |
Collapse
|
7
|
D'Souza P, Farmer C, Johnston J, Han ST, Adams D, Hartman AL, Zein W, Huryn LA, Solomon B, King K, Jordan C, Myles J, Nicoli ER, Rothermel CE, Algarin YM, Huang R, Quimby R, Zainab M, Bowden S, Crowell A, Buckley A, Brewer C, Regier D, Brooks B, Baker E, Vézina G, Thurm A, Tifft CJ. GM1 Gangliosidosis Type II: Results of a 10-Year Prospective Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.04.24300778. [PMID: 38313286 PMCID: PMC10836125 DOI: 10.1101/2024.01.04.24300778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Purpose GM1 gangliosidosis (GM1) is an ultra-rare lysosomal storage disease caused by pathogenic variants in galactosidase beta 1 (GLB1; NM_000404), primarily characterized by neurodegeneration, often in children. There are no approved treatments for GM1, but clinical trials using gene therapy (NCT03952637, NCT04713475) and small molecule substrate inhibitors (NCT04221451) are ongoing. Understanding the natural history of GM1 is essential for timely diagnosis, facilitating better supportive care, and contextualizing the results of therapeutic trials. Methods Forty-one individuals with type II GM1 (n=17 late infantile and n=24 juvenile onset) participated in a single-site prospective observational study. Here, we describe the results of extensive multisystem assessment batteries, including clinical labs, neuroimaging, physiological exams, and behavioral assessments. Results Classification of 37 distinct variants in this cohort was performed according to ACMG criteria and resulted in the upgrade of six and the submission of four new variants to pathogenic or likely pathogenic. In contrast to type I infantile, children with type II disease exhibited normal or near normal hearing and did not have cherry red maculae or significant hepatosplenomegaly. Some older children with juvenile onset developed thickened aortic and/or mitral valves with regurgitation. Serial MRIs demonstrated progressive brain atrophy that were more pronounced in those with late infantile onset. MR spectroscopy showed worsening elevation of myo-inositol and deficit of N-acetyl aspartate that were strongly correlated with scores on the Vineland Adaptive Behavior Scale and progress more rapidly in late infantile than juvenile onset disease. Conclusion The comprehensive serial phenotyping of type II GM1 patients expands the understanding of disease progression and clarifies some common misconceptions about type II patients. Findings from this 10-year endeavor are a pivotal step toward more timely diagnosis and better supportive care for patients. The wealth of data amassed through this effort will serve as a robust comparator for ongoing and future therapeutic trials.
Collapse
Affiliation(s)
- Precilla D'Souza
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Cristan Farmer
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, 10 Center Drive, Bethesda MD USA
| | - Jean Johnston
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Sangwoo T Han
- Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - David Adams
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Adam L Hartman
- Division of Clinical Research, National Institute of Neurological Disorders and Stroke, 6001 Executive Blvd, Rockville, MD, USA
| | - Wadih Zein
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, 10 Center Drive, Bethesda MD, USA
| | - Laryssa A Huryn
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, 10 Center Drive, Bethesda MD, USA
| | - Beth Solomon
- Speech Language Pathology Section, Rehabilitation Medicine Department, Warren Grant Magnuson Clinical Research Center, 10 Center Drive Bethesda MD USA
| | - Kelly King
- Neurotology Branch, Division of Intramural Research, National Institute on Deafness and Other Communication Disorders, 10 Center Drive, Bethesda, MD USA
| | - Christopher Jordan
- Inova Children's Cardiology, 8260 Willow Oaks Corporate Drive; suite 400; Fairfax, VA, 22031
| | - Jennifer Myles
- Nutrition Department, Warren Grant Magnuson Clinical Research Center, 10 Center Drive Bethesda MD USA
| | - Elena-Raluca Nicoli
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Caroline E Rothermel
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Yoliann Mojica Algarin
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Reyna Huang
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Rachel Quimby
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Mosufa Zainab
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Sarah Bowden
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Anna Crowell
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| | - Ashura Buckley
- Sleep and Neurodevelopment Service, National Institute of Mental Health, 10 Center Drive, Bethesda MD USA
| | - Carmen Brewer
- Neurotology Branch, Division of Intramural Research, National Institute on Deafness and Other Communication Disorders, 10 Center Drive, Bethesda, MD USA
| | - Deborah Regier
- Genetics and Metabolism, Children's National Hospital, 111 Michigan Avenue NW, Washington DC USA
| | - Brian Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, 10 Center Drive, Bethesda MD, USA
| | - Eva Baker
- Department of Radiology and Imaging Sciences, Warren Grant Magnuson Clinical Research Center, 10 Center Drive, Bethesda, MD, USA
| | - Gilbert Vézina
- Program in Neuroradiology, Children's National Hospital, 111 Michigan Avenue NW, Washington DC USA; Radiology and Pediatrics, The George Washington University School of Medicine and Health Sciences, 2300 I St NW, Washington DC USA
| | - Audrey Thurm
- Neurodevelopmental and Behavioral Phenotyping Service, National Institute of Mental Health, 10 Center Drive, Bethesda MD USA
| | - Cynthia J Tifft
- Office of the Clinical Director and Medical Genetics Branch, National Human Genome Research Institute, 10 Center Drive, Bethesda MD USA
| |
Collapse
|
8
|
Kumar S, Panda SP. Targeting GM2 Ganglioside Accumulation in Dementia: Current Therapeutic Approaches and Future Directions. Curr Mol Med 2024; 24:1329-1345. [PMID: 37877564 DOI: 10.2174/0115665240264547231017110613] [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: 05/30/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 10/26/2023]
Abstract
Dementia in neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and dementia with Lewy bodies (DLB) is a progressive neurological condition affecting millions worldwide. The amphiphilic molecule GM2 gangliosides are abundant in the human brain and play important roles in neuronal development, intercellular recognition, myelin stabilization, and signal transduction. GM2 ganglioside's degradation requires hexosaminidase A (HexA), a heterodimer composed of an α subunit encoded by HEXA and a β subunit encoded by HEXB. The hydrolysis of GM2 also requires a non-enzymatic protein, the GM2 activator protein (GM2-AP), encoded by GM2A. Pathogenic mutations of HEXA, HEXB, and GM2A are responsible for autosomal recessive diseases known as GM2 gangliosidosis, caused by the excessive intralysosomal accumulation of GM2 gangliosides. In AD, PD and DLB, GM2 ganglioside accumulation is reported to facilitate Aβ and α-synuclein aggregation into toxic oligomers and plaques through activation of downstream signaling pathways, such as protein kinase C (PKC) and oxidative stress factors. This review explored the potential role of GM2 ganglioside alteration in toxic protein aggregations and its related signaling pathways leading to neurodegenerative diseases. Further review explored potential therapeutic approaches, which include synthetic and phytomolecules targeting GM2 ganglioside accumulation in the brain, holding a promise for providing new and effective management for dementia.
Collapse
Affiliation(s)
- Sanjesh Kumar
- Institute of Pharmaceutical Research, GLA University Mathura, Uttara Pradesh-281406, India
| | - Siva Prasad Panda
- Institute of Pharmaceutical Research, GLA University Mathura, Uttara Pradesh-281406, India
| |
Collapse
|
9
|
Leuzzi V, Galosi S. Experimental pharmacology: Targeting metabolic pathways. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 169:259-315. [PMID: 37482395 DOI: 10.1016/bs.irn.2023.05.005] [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: 07/25/2023]
Abstract
Since the discovery of the treatment for Wilson disease a growing number of treatable inherited dystonias have been identified and their search and treatment have progressively been implemented in the clinics of patients with dystonia. While waiting for gene therapy to be more widely and adequately translated into the clinical setting, the efforts to divert the natural course of dystonia reside in unveiling its pathogenesis. Specific metabolic treatments can rewrite the natural history of the disease by preventing neurotoxic metabolite accumulation or interfering with the cell accumulation of damaging metabolites, restoring energetic cell fuel, supplementing defective metabolites, and supplementing the defective enzyme. A metabolic derangement of cell homeostasis is part of the progression of many non-metabolic genetic lesions and could be the target for possible metabolic approaches. In this chapter, we provided an update on treatment strategies for treatable inherited dystonias and an overview of genetic dystonias with new experimental therapeutic approaches available or close to clinical translation.
Collapse
Affiliation(s)
- Vincenzo Leuzzi
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Serena Galosi
- Department of Human Neuroscience, Sapienza University, Rome, Italy.
| |
Collapse
|
10
|
Mignani L, Guerra J, Corli M, Capoferri D, Presta M. Zebra-Sphinx: Modeling Sphingolipidoses in Zebrafish. Int J Mol Sci 2023; 24:ijms24054747. [PMID: 36902174 PMCID: PMC10002607 DOI: 10.3390/ijms24054747] [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: 01/20/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Sphingolipidoses are inborn errors of metabolism due to the pathogenic mutation of genes that encode for lysosomal enzymes, transporters, or enzyme cofactors that participate in the sphingolipid catabolism. They represent a subgroup of lysosomal storage diseases characterized by the gradual lysosomal accumulation of the substrate(s) of the defective proteins. The clinical presentation of patients affected by sphingolipid storage disorders ranges from a mild progression for some juvenile- or adult-onset forms to severe/fatal infantile forms. Despite significant therapeutic achievements, novel strategies are required at basic, clinical, and translational levels to improve patient outcomes. On these bases, the development of in vivo models is crucial for a better understanding of the pathogenesis of sphingolipidoses and for the development of efficacious therapeutic strategies. The teleost zebrafish (Danio rerio) has emerged as a useful platform to model several human genetic diseases owing to the high grade of genome conservation between human and zebrafish, combined with precise genome editing and the ease of manipulation. In addition, lipidomic studies have allowed the identification in zebrafish of all of the main classes of lipids present in mammals, supporting the possibility to model diseases of the lipidic metabolism in this animal species with the advantage of using mammalian lipid databases for data processing. This review highlights the use of zebrafish as an innovative model system to gain novel insights into the pathogenesis of sphingolipidoses, with possible implications for the identification of more efficacious therapeutic approaches.
Collapse
|
11
|
Nicoli ER, Huebecker M, Han ST, Garcia K, Munasinghe J, Lizak M, Latour Y, Yoon R, Glase B, Tyrlik M, Peiravi M, Springer D, Baker EH, Priestman D, Sidhu R, Kell P, Jiang X, Kolstad J, Kuhn AL, Shazeeb MS, Acosta MT, Proia RL, Platt FM, Tifft CJ. Glb1 knockout mouse model shares natural history with type II GM1 gangliosidosis patients. Mol Genet Metab 2023; 138:107508. [PMID: 36709532 PMCID: PMC10617618 DOI: 10.1016/j.ymgme.2023.107508] [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: 12/07/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
GM1 gangliosidosis is a rare lysosomal storage disorder affecting multiple organ systems, primarily the central nervous system, and is caused by functional deficiency of β-galactosidase (GLB1). Using CRISPR/Cas9 genome editing, we generated a mouse model to evaluate characteristics of the disease in comparison to GM1 gangliosidosis patients. Our Glb1-/- mice contain small deletions in exons 2 and 6, producing a null allele. Longevity is approximately 50 weeks and studies demonstrated that female Glb1-/- mice die six weeks earlier than male Glb1-/- mice. Gait analyses showed progressive abnormalities including abnormal foot placement, decreased stride length and increased stance width, comparable with what is observed in type II GM1 gangliosidosis patients. Furthermore, Glb1-/- mice show loss of motor skills by 20 weeks assessed by adhesive dot, hanging wire, and inverted grid tests, and deterioration of motor coordination by 32 weeks of age when evaluated by rotarod testing. Brain MRI showed progressive cerebellar atrophy in Glb1-/- mice as seen in some patients. In addition, Glb1-/- mice also show significantly increased levels of a novel pentasaccharide biomarker in urine and plasma which we also observed in GM1 gangliosidosis patients. Glb1-/- mice also exhibit accumulation of glycosphingolipids in the brain with increases in GM1 and GA1 beginning by 8 weeks. Surprisingly, despite being a null variant, this Glb1-/- mouse most closely models the less severe type II disease and will guide the development of new therapies for patients with the disorder.
Collapse
Affiliation(s)
- Elena-Raluca Nicoli
- Glycosphingolipid and Glycoprotein Disorders Unit, Medical Genetic Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mylene Huebecker
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Sangwoo T Han
- Glycosphingolipid and Glycoprotein Disorders Unit, Medical Genetic Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Karolyn Garcia
- Glycosphingolipid and Glycoprotein Disorders Unit, Medical Genetic Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Jeeva Munasinghe
- Mouse Imaging Facility, National Institute of Neurological Disorder and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Martin Lizak
- Mouse Imaging Facility, National Institute of Neurological Disorder and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Yvonne Latour
- Glycosphingolipid and Glycoprotein Disorders Unit, Medical Genetic Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Robin Yoon
- Glycosphingolipid and Glycoprotein Disorders Unit, Medical Genetic Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Brianna Glase
- Glycosphingolipid and Glycoprotein Disorders Unit, Medical Genetic Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Michal Tyrlik
- Glycosphingolipid and Glycoprotein Disorders Unit, Medical Genetic Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States; Phenotyping Core (D.A.S.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Morteza Peiravi
- Phenotyping Core (D.A.S.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Danielle Springer
- Phenotyping Core (D.A.S.), National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Eva H Baker
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - David Priestman
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Rohini Sidhu
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Pamela Kell
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Xuntian Jiang
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| | - Josephine Kolstad
- Image Processing and Analysis Core (iPAC), Department of Radiology, UMass Chan Medical School, Worcester, MA, United States
| | - Anna Luisa Kuhn
- Image Processing and Analysis Core (iPAC), Department of Radiology, UMass Chan Medical School, Worcester, MA, United States
| | - Mohammed Salman Shazeeb
- Image Processing and Analysis Core (iPAC), Department of Radiology, UMass Chan Medical School, Worcester, MA, United States
| | - Maria T Acosta
- Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, United States
| | - Richard L Proia
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Cynthia J Tifft
- Glycosphingolipid and Glycoprotein Disorders Unit, Medical Genetic Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States; Undiagnosed Diseases Program, Common Fund, Office of the Director, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
12
|
Muschol N, Koehn A, von Cossel K, Okur I, Ezgu F, Harmatz P, de Castro Lopez MJ, Couce ML, Lin SP, Batzios S, Cleary M, Solano M, Nestrasil I, Kaufman B, Shaywitz AJ, Maricich SM, Kuca B, Kovalchin J, Zanelli E. A phase I/II study on intracerebroventricular tralesinidase alfa in patients with Sanfilippo syndrome type B. J Clin Invest 2023; 133:165076. [PMID: 36413418 PMCID: PMC9843052 DOI: 10.1172/jci165076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
BackgroundSanfilippo type B is a mucopolysaccharidosis (MPS) with a major neuronopathic component characterized by heparan sulfate (HS) accumulation due to mutations in the NAGLU gene encoding alfa-N-acetyl-glucosaminidase. Enzyme replacement therapy for neuronopathic MPS requires efficient enzyme delivery throughout the brain in order to normalize HS levels, prevent brain atrophy, and potentially delay cognitive decline.MethodsIn this phase I/II open-label study, patients with MPS type IIIB (n = 22) were treated with tralesinidase alfa administered i.c.v. The patients were monitored for drug exposure; total HS and HS nonreducing end (HS-NRE) levels in both cerebrospinal fluid (CSF) and plasma; anti-drug antibody response; brain, spleen, and liver volumes as measured by MRI; and cognitive development as measured by age-equivalent (AEq) scores.ResultsIn the Part 1 dose escalation (30, 100, and 300 mg) phase, a 300 mg dose of tralesinidase alfa was necessary to achieve normalization of HS and HS-NRE levels in the CSF and plasma. In Part 2, 300 mg tralesinidase alfa sustained HS and HS-NRE normalization in the CSF and stabilized cortical gray matter volume (CGMV) over 48 weeks of treatment. Resolution of hepatomegaly and a reduction in spleen volume were observed in most patients. Significant correlations were also established between the change in cognitive AEq score and plasma drug exposure, plasma HS-NRE levels, and CGMV.ConclusionAdministration of tralesinidase alfa i.c.v. effectively normalized HS and HS-NRE levels as a prerequisite for clinical efficacy. Peripheral drug exposure data suggest a role for the glymphatic system in altering tralesinidase alfa efficacy.Trial registrationClinicaltrials.gov NCT02754076.FUNDINGBioMarin Pharmaceutical Inc. and Allievex Corporation.
Collapse
Affiliation(s)
- Nicole Muschol
- University Medical Center Hamburg-Eppendorf, International Center for Lysosomal Disorders (ICLD), Hamburg, Germany
| | - Anja Koehn
- University Medical Center Hamburg-Eppendorf, International Center for Lysosomal Disorders (ICLD), Hamburg, Germany
| | - Katharina von Cossel
- University Medical Center Hamburg-Eppendorf, International Center for Lysosomal Disorders (ICLD), Hamburg, Germany
| | - Ilyas Okur
- Gazi University Faculty of Medicine, Departments of Pediatric Metabolism and Genetics, Ankara, Turkey
| | - Fatih Ezgu
- Gazi University Faculty of Medicine, Departments of Pediatric Metabolism and Genetics, Ankara, Turkey
| | - Paul Harmatz
- UCSF Benioff Children’s Hospital Oakland, Oakland, California, USA
| | - Maria J. de Castro Lopez
- Hospital Clínico Universitario de Santiago, University of Santiago de Compostela, IDIS, CIBERER, MetabERN, A Coruña, Spain
| | - Maria Luz Couce
- Hospital Clínico Universitario de Santiago, University of Santiago de Compostela, IDIS, CIBERER, MetabERN, A Coruña, Spain
| | | | | | | | | | - Igor Nestrasil
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, and Masonic Institute for the Developing Brain, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brian Kaufman
- CLB Consulting, Falls of Neuse, Raleigh, North Carolina, USA
| | | | | | - Bernice Kuca
- Allievex Corporation, Marblehead, Massachusetts, USA
| | | | - Eric Zanelli
- Allievex Corporation, Marblehead, Massachusetts, USA
| |
Collapse
|
13
|
Ries M, Mendoza G, Arash-Kaps L, Amraoui Y, Quack F, Hardt B, Diederich S, Beck M, Mengel E. Quantitative longitudinal natural history of 8 gangliosidoses-conceptual framework and baseline data of the German 8-in-1 disease registry. A cross-sectional analysis. Genet Med 2022; 24:2434-2443. [PMID: 36194207 DOI: 10.1016/j.gim.2022.09.001] [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: 04/15/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Gangliosidoses are a group of inherited neurogenetic autosomal recessive lysosomal storage disorders usually presenting with progressive macrocephaly, developmental delay, and regression, leading to significant morbidity and premature death. A quantitative definition of the natural history would support and enable clinical development of specific therapies. METHODS Single disease registry of 8 gangliosidoses (NCT04624789). Cross-sectional analysis of baseline data in N = 26 patients. Primary end point: disease severity assessed by the 8-in-1 score. Secondary end points: first neurologic sign or symptom observed (1) by parents and (2) by physicians, diagnostic delay, as well as phenotypical characterization. Tertiary end points: neurologic outcomes (development, ataxia, dexterity) and disability. RESULTS The 8-in-1 score quantitatively captured severity of disease. Parents recognized initial manifestations (startle reactions) earlier than physicians (motor developmental delay and hypotonia). Median diagnostic delay was 3.16 (interquartile range 0.69-6.25) years. In total, 8 patients presented with late-infantile phenotypes. CONCLUSION Data in this registry raise awareness of these rare and fatal conditions to accelerate diagnosis, inform counseling of afflicted families, define quantitative end points for clinical trials, and can serve as historical controls for future therapeutic studies. We provide further insight into the rare late-infantile phenotype for GM2-gangliosidosis. Longitudinal follow up is planned.
Collapse
Affiliation(s)
- Markus Ries
- Pediatric Neurology and Metabolic Medicine, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany; Center for Rare Disorders, Heidelberg University Hospital, Heidelberg, Germany; Center for Virtual Patients, Heidelberg Faculty of Medicine, Heidelberg, Germany.
| | - Grecia Mendoza
- SphinCS - Institute for Clinical Research in Lysosomal Storage Disorders, Hochheim, Germany
| | - Laila Arash-Kaps
- SphinCS - Institute for Clinical Research in Lysosomal Storage Disorders, Hochheim, Germany
| | - Yasmina Amraoui
- SphinCS - Institute for Clinical Research in Lysosomal Storage Disorders, Hochheim, Germany
| | - Folker Quack
- Hand in Hand against Tay-Sachs and Sandhoff disease in Germany, Höchberg, Germany
| | - Brigitte Hardt
- Hand in Hand against Tay-Sachs and Sandhoff disease in Germany, Höchberg, Germany
| | - Stefan Diederich
- Institute of Human Genetics, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael Beck
- SphinCS - Institute for Clinical Research in Lysosomal Storage Disorders, Hochheim, Germany
| | - Eugen Mengel
- SphinCS - Institute for Clinical Research in Lysosomal Storage Disorders, Hochheim, Germany
| |
Collapse
|
14
|
Okur I, Ezgu F, Giugliani R, Muschol N, Koehn A, Amartino H, Harmatz P, de Castro Lopez MJ, Couce ML, Lin SP, Batzios S, Cleary M, Solano M, Peters H, Lee J, Nestrasil I, Shaywitz AJ, Maricich SM, Kuca B, Kovalchin J, Zanelli E. Longitudinal Natural History of Pediatric Subjects Affected with Mucopolysaccharidosis IIIB. J Pediatr 2022; 249:50-58.e2. [PMID: 35709957 DOI: 10.1016/j.jpeds.2022.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To characterize the longitudinal natural history of disease progression in pediatric subjects affected with mucopolysaccharidosis (MPS) IIIB. STUDY DESIGN Sixty-five children with a confirmed diagnosis of MPS IIIB were enrolled into 1 of 2 natural history studies and followed for up to 4 years. Cognitive and adaptive behavior functions were analyzed in all subjects, and volumetric magnetic resonance imaging analysis of liver, spleen, and brain, as well as levels of heparan sulfate (HS) and heparan sulfate nonreducing ends (HS-NRE), were measured in a subset of subjects. RESULTS The majority of subjects with MPS IIIB achieved an apex on both cognition and adaptive behavior age equivalent scales between age 3 and 6 years. Development quotients for both cognition and adaptive behavior follow a linear trajectory by which subjects reach a nadir with a score <25 for an age equivalent of 24 months by age 8 years on average and by 13.5 years at the latest. All tested subjects (n = 22) had HS and HS-NRE levels above the normal range in cerebrospinal fluid and plasma, along with signs of hepatomegaly. Subjects lost an average of 26 mL of brain volume (-2.7%) over 48 weeks, owing entirely to a loss of cortical gray matter (32 mL; -6.5%). CONCLUSIONS MPS IIIB exists along a continuum based on cognitive decline and cortical gray matter atrophy. Although a few individuals with MPS IIIB have an attenuated phenotype, the majority follow predicted trajectories for both cognition and adaptive behavior. TRIAL REGISTRATION ClinicalTrials.gov identifiers NCT02493998, NCT03227042, and NCT02754076.
Collapse
Affiliation(s)
- Ilyas Okur
- Department of Pediatric Metabolism, Gazi University Faculty of Medicine, Ankara, Turkey; Department of Genetics, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Fatih Ezgu
- Department of Pediatric Metabolism, Gazi University Faculty of Medicine, Ankara, Turkey; Department of Genetics, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Roberto Giugliani
- Medical Genetics Service and DR Brasil, HCPA, Department of Genetics, UFRGS, DASA, and INAGEMP, Porto Alegre, Brazil
| | - Nicole Muschol
- International Center for Lysosomal Disorders, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anja Koehn
- International Center for Lysosomal Disorders, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Paul Harmatz
- UCSF Benioff Children's Hospital Oakland, Oakland, CA
| | - Maria J de Castro Lopez
- Hospital Clínico Universitario de Santiago, University of Santiago de Compostela, IDIS, CIBERER, MetabERN, A Coruña, Spain
| | - Maria Luz Couce
- Hospital Clínico Universitario de Santiago, University of Santiago de Compostela, IDIS, CIBERER, MetabERN, A Coruña, Spain
| | | | | | | | | | | | - Joy Lee
- Royal Children's Hospital, Melbourne, Australia
| | - Igor Nestrasil
- Division of Clinical Behavioral Neuroscience and Masonic Institute for the Developing Brain, Department of Pediatrics, University of Minnesota, Minneapolis, MN
| | | | | | | | | | | |
Collapse
|
15
|
Nowacki JC, Fields AM, Fu MM. Emerging cellular themes in leukodystrophies. Front Cell Dev Biol 2022; 10:902261. [PMID: 36003149 PMCID: PMC9393611 DOI: 10.3389/fcell.2022.902261] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
Leukodystrophies are a broad spectrum of neurological disorders that are characterized primarily by deficiencies in myelin formation. Clinical manifestations of leukodystrophies usually appear during childhood and common symptoms include lack of motor coordination, difficulty with or loss of ambulation, issues with vision and/or hearing, cognitive decline, regression in speech skills, and even seizures. Many cases of leukodystrophy can be attributed to genetic mutations, but they have diverse inheritance patterns (e.g., autosomal recessive, autosomal dominant, or X-linked) and some arise from de novo mutations. In this review, we provide an updated overview of 35 types of leukodystrophies and focus on cellular mechanisms that may underlie these disorders. We find common themes in specialized functions in oligodendrocytes, which are specialized producers of membranes and myelin lipids. These mechanisms include myelin protein defects, lipid processing and peroxisome dysfunction, transcriptional and translational dysregulation, disruptions in cytoskeletal organization, and cell junction defects. In addition, non-cell-autonomous factors in astrocytes and microglia, such as autoimmune reactivity, and intercellular communication, may also play a role in leukodystrophy onset. We hope that highlighting these themes in cellular dysfunction in leukodystrophies may yield conceptual insights on future therapeutic approaches.
Collapse
|
16
|
Blumenreich S, Nehushtan T, Barav OB, Saville JT, Dingjan T, Hardy J, Fuller M, Futerman AH. Elevation of gangliosides in four brain regions from Parkinson's disease patients with a GBA mutation. NPJ Parkinsons Dis 2022; 8:99. [PMID: 35933559 PMCID: PMC9357011 DOI: 10.1038/s41531-022-00363-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022] Open
Abstract
A number of genetic risk factors have been identified over the past decade for Parkinson's Disease (PD), with variants in GBA prominent among them. GBA encodes the lysosomal enzyme that degrades the glycosphingolipid, glucosylceramide (GlcCer), with the activity of this enzyme defective in Gaucher disease. Based on the ill-defined relationship between glycosphingolipid metabolism and PD, we now analyze levels of various lipids by liquid chromatography/electrospray ionization-tandem mass spectrometry in four brain regions from age- and sex-matched patient samples, including idiopathic PD, PD patients with a GBA mutation and compare both to control brains (n = 21 for each group) obtained from individuals who died from a cause unrelated to PD. Of all the glycerolipids, sterols, and (glyco)sphingolipids (251 lipids in total), the only lipid class which showed significant differences were the gangliosides (sialic acid-containing complex glycosphingolipids), which were elevated in 3 of the 4 PD-GBA brain regions. There was no clear correlation between levels of individual gangliosides and the genetic variant in Gaucher disease [9 samples of severe (neuronopathic), 4 samples of mild (non-neuronopathic) GBA variants, and 8 samples with low pathogenicity variants which have a higher risk for development of PD]. Most brain regions, i.e. occipital cortex, cingulate gyrus, and striatum, did not show a statistically significant elevation of GlcCer in PD-GBA. Only one region, the middle temporal gyrus, showed a small, but significant elevation in GlcCer concentration in PD-GBA. We conclude that changes in ganglioside, but not in GlcCer levels, may contribute to the association between PD and GBA mutations.
Collapse
Affiliation(s)
- Shani Blumenreich
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Tamar Nehushtan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Or B Barav
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Jennifer T Saville
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital and Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Tamir Dingjan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Dementia Research Institute, University College London, London, WC1N 3BG, UK
| | - Maria Fuller
- Genetics and Molecular Pathology, SA Pathology at Women's and Children's Hospital and Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel.
- The Joseph Meyerhof Professor of Biochemistry at the Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
17
|
Intracranial Volumes of Healthy Children in the First 3 Years of Life: An Analysis of 270 Magnetic Resonance Imaging Scans. Plast Reconstr Surg 2022; 150:136e-144e. [PMID: 35575631 DOI: 10.1097/prs.0000000000009188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND There is a paucity of data on normal intracranial volumes for healthy children during the first few years of life, when cranial growth velocity is greatest. The aim of this study was to generate a normative predictive model of intracranial volumes based on brain magnetic resonance imaging from a large sample of healthy children to serve as a reference tool for future studies on craniosynostosis. METHODS Structural magnetic resonance imaging data for healthy children up to 3 years of age was acquired from the National Institutes of Health Pediatric MRI Data Repository. Intracranial volumes were calculated using T1-weighted scans with FreeSurfer (version 6.0.0). Mean intracranial volumes were calculated and best-fit logarithmic curves were generated. Results were compared to previously published intracranial volume curves. RESULTS Two-hundred seventy magnetic resonance imaging scans were available: 118 were collected in the first year of life, 97 were collected between years 1 and 2, and 55 were collected between years 2 and 3. A best-fit logarithmic growth curve was generated for male and female patients. The authors' regression models showed that male patients had significantly greater intracranial volumes than female patients after 1 month of age. Predicted intracranial volumes were also greater in male and female patients in the first 6 months of life as compared to previously published intracranial volume curves. CONCLUSIONS To the authors' knowledge, this is the largest series of demographically representative magnetic resonance imaging-based intracranial volumes for children aged 3 years and younger. The model generated in this study can be used by investigators as a reference for evaluating craniosynostosis patients.
Collapse
|
18
|
Pardridge WM. Blood-brain barrier delivery for lysosomal storage disorders with IgG-lysosomal enzyme fusion proteins. Adv Drug Deliv Rev 2022; 184:114234. [PMID: 35307484 DOI: 10.1016/j.addr.2022.114234] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/25/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
The majority of lysosomal storage diseases affect the brain. Treatment of the brain with intravenous enzyme replacement therapy is not successful, because the recombinant lysosomal enzymes do not cross the blood-brain barrier (BBB). Biologic drugs, including lysosomal enzymes, can be re-engineered for BBB delivery as IgG-enzyme fusion proteins. The IgG domain of the fusion protein is a monoclonal antibody directed against an endogenous receptor-mediated transporter at the BBB, such as the insulin receptor or the transferrin receptor. This receptor transports the IgG across the BBB, in parallel with the endogenous receptor ligand, and the IgG acts as a molecular Trojan horse to ferry into brain the lysosomal enzyme genetically fused to the IgG. The IgG-enzyme fusion protein is bi-functional and retains both high affinity binding for the BBB receptor, and high lysosomal enzyme activity. IgG-lysosomal enzymes are presently in clinical trials for treatment of the brain in Mucopolysaccharidosis.
Collapse
|
19
|
Welford RW, Farine H, Steiner M, Garzotti M, Dobrenis K, Sievers C, Strasser DS, Amraoui Y, Groenen PM, Giugliani R, Mengel E. Plasma neurofilament light, glial fibrillary acidic protein and lysosphingolipid biomarkers for pharmacodynamics and disease monitoring of GM2 and GM1 gangliosidoses patients. Mol Genet Metab Rep 2022; 30:100843. [PMID: 35242574 PMCID: PMC8856936 DOI: 10.1016/j.ymgmr.2022.100843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 12/03/2022] Open
Abstract
GM2 and GM1 gangliosidoses are genetic, neurodegenerative lysosomal sphingolipid storage disorders. The earlier the age of onset, the more severe the clinical presentation and progression, with infantile, juvenile and late-onset presentations broadly delineated into separate phenotypic subtypes. Gene and substrate reduction therapies, both of which act directly on sphingolipidosis are entering clinical trials for treatment of these disorders. Simple to use biomarkers for disease monitoring are urgently required to support and expedite these clinical trials. Here, lysosphingolipid and protein biomarkers of sphingolipidosis and neuropathology respectively, were assessed in plasma samples from 33 GM2 gangliosidosis patients, 13 GM1 gangliosidosis patients, and compared to 66 controls. LysoGM2 and lysoGM1 were detectable in 31/33 GM2 gangliosidosis and 12/13 GM1 gangliosidosis patient samples respectively, but not in any controls. Levels of the axonal damage marker Neurofilament light (NF-L) were highly elevated in both GM2 and GM1 gangliosidosis patient plasma samples, with no overlap with controls. Levels of the astrocytosis biomarker Glial fibrillary acidic protein (GFAP) were also elevated in samples from both patient populations, albeit with some overlap with controls. In GM2 gangliosidosis patient plasma NF-L, Tau, GFAP and lysoGM2 were all most highly elevated in infantile onset patients, indicating a relationship to severity and phenotype. Plasma NF-L and liver lysoGM2 were also elevated in a GM2 gangliosidosis mouse model, and were lowered by treatment with a drug that slowed disease progression. These results indicate that lysosphingolipids and NF-L/GFAP have potential to monitor pharmacodynamics and pathogenic processes respectively in GM2 and GM1 gangliosidoses patients.
Collapse
|
20
|
The GM2 gangliosidoses: Unlocking the mysteries of pathogenesis and treatment. Neurosci Lett 2021; 764:136195. [PMID: 34450229 PMCID: PMC8572160 DOI: 10.1016/j.neulet.2021.136195] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/28/2022]
|
21
|
Rowe OE, Rangaprakash D, Weerasekera A, Godbole N, Haxton E, James PF, Stephen CD, Barry RL, Eichler FS, Ratai EM. Magnetic resonance imaging and spectroscopy in late-onset GM2-gangliosidosis. Mol Genet Metab 2021; 133:386-396. [PMID: 34226107 PMCID: PMC8289742 DOI: 10.1016/j.ymgme.2021.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Our study aimed to quantify structural changes in relation to metabolic abnormalities in the cerebellum, thalamus, and parietal cortex of patients with late-onset GM2-gangliosidosis (LOGG), which encompasses late-onset Tay-Sachs disease (LOTS) and Sandhoff disease (LOSD). METHODS We enrolled 10 patients with LOGG (7 LOTS, 3 LOSD) who underwent a neurological assessment battery and 7 age-matched controls. Structural MRI and MRS were performed on a 3 T scanner. Structural volumes were obtained from FreeSurfer and normalized by total intracranial volume. Quantified metabolites included N-acetylaspartate (NAA), choline (Cho), myo-inositol (mI), creatine (Cr), and combined glutamate-glutamine (Glx). Metabolic concentrations were corrected for partial volume effects. RESULTS Structural analyses revealed significant cerebellar atrophy in the LOGG cohort, which was primarily driven by LOTS patients. NAA was lower and mI higher in LOGG, but this was also significantly driven by the LOTS patients. Clinical ataxia deficits (via the Scale for the Assessment and Rating of Ataxia) were associated with neuronal injury (via NAA), neuroinflammation (via mI), and volumetric atrophy in the cerebellum. INTERPRETATION The decrease of NAA in the cerebellum suggests that, in addition to cerebellar atrophy, there is ongoing impaired neuronal function and/or loss, while an increase in mI indicates possible neuroinflammation in LOGG (more so within the LOTS subvariant). Quantifying cerebellar atrophy in relation to neurometabolic differences in LOGG may lead to improvements in assessing disease severity, progression, and pharmacological efficacy. Lastly, additional neuroimaging studies in LOGG are required to contrast LOTS and LOSD more accurately.
Collapse
Affiliation(s)
- Olivia E Rowe
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - D Rangaprakash
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Akila Weerasekera
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Neha Godbole
- Leukodystrophy Clinic, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Rare Neurological Diseases, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth Haxton
- Leukodystrophy Clinic, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Rare Neurological Diseases, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter F James
- Leukodystrophy Clinic, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Rare Neurological Diseases, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Christopher D Stephen
- Center for Rare Neurological Diseases, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Movement Disorders Division and Ataxia Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Robert L Barry
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Harvard-Massachusetts Institute of Technology Health Sciences & Technology, Cambridge, MA, USA
| | - Florian S Eichler
- Leukodystrophy Clinic, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Center for Rare Neurological Diseases, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eva-Maria Ratai
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA; Division of Neuroradiology, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Sheth J, Nair A. Treatment for Lysosomal Storage Disorders. Curr Pharm Des 2021; 26:5110-5118. [PMID: 33059565 DOI: 10.2174/1381612826666201015154932] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/22/2020] [Indexed: 12/31/2022]
Abstract
Lysosomal storage disorders comprise a group of approximately 70 types of inherited diseases resulting due to lysosomal gene defects. The outcome of the defect is a deficiency in either of the three: namely, lysosomal enzymes, activator protein, or transmembrane protein, as a result of which there is an unwanted accumulation of biomolecules inside the lysosomes. The pathophysiology of these conditions is complex affecting several organ systems and nervous system involvement in a majority of cases. Several research studies have well elucidated the mechanism underlying the disease condition leading to the development in devising the treatment strategies for the same. Currently, these approaches aim to reduce the severity of symptoms or delay the disease progression but do not provide a complete cure. The main treatment methods include Enzyme replacement therapy, Bone marrow transplantation, Substrate reduction therapy, use of molecular chaperones, and Gene therapy. This review article presents an elaborate description of these strategies and discusses the ongoing studies for the same.
Collapse
Affiliation(s)
- Jayesh Sheth
- Foundation for Research in Genetics and Endocrinology, Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad, Gujarat, India
| | - Aadhira Nair
- Foundation for Research in Genetics and Endocrinology, Institute of Human Genetics, FRIGE House, Jodhpur Gam Road, Satellite, Ahmedabad, Gujarat, India
| |
Collapse
|
23
|
Rha AK, Maguire AS, Martin DR. GM1 Gangliosidosis: Mechanisms and Management. Appl Clin Genet 2021; 14:209-233. [PMID: 33859490 PMCID: PMC8044076 DOI: 10.2147/tacg.s206076] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/15/2021] [Indexed: 01/10/2023] Open
Abstract
The lysosomal storage disorder, GM1 gangliosidosis (GM1), is a neurodegenerative condition resulting from deficiency of the enzyme β-galactosidase (β-gal). Mutation of the GLB1 gene, which codes for β-gal, prevents cleavage of the terminal β-1,4-linked galactose residue from GM1 ganglioside. Subsequent accumulation of GM1 ganglioside and other substrates in the lysosome impairs cell physiology and precipitates dysfunction of the nervous system. Beyond palliative and supportive care, no FDA-approved treatments exist for GM1 patients. Researchers are critically evaluating the efficacy of substrate reduction therapy, pharmacological chaperones, enzyme replacement therapy, stem cell transplantation, and gene therapy for GM1. A Phase I/II clinical trial for GM1 children is ongoing to evaluate the safety and efficacy of adeno-associated virus-mediated GLB1 delivery by intravenous injection, providing patients and families with hope for the future.
Collapse
Affiliation(s)
- Allisandra K Rha
- Scott-Ritchey Research Center, Auburn University, Auburn, AL, 36849, USA
| | - Anne S Maguire
- Scott-Ritchey Research Center, Auburn University, Auburn, AL, 36849, USA
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, 36849, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Auburn University, Auburn, AL, 36849, USA
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, 36849, USA
| |
Collapse
|
24
|
King KE, Kim S, Whitley CB, Jarnes-Utz JR. The juvenile gangliosidoses: A timeline of clinical change. Mol Genet Metab Rep 2020; 25:100676. [PMID: 33240792 PMCID: PMC7674119 DOI: 10.1016/j.ymgmr.2020.100676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Background The gangliosidoses are rare inherited diseases that result in pathologic accumulation of gangliosides in the central nervous system and other tissues, leading to severe and progressive neurological impairment and early death in the childhood forms. No treatments are currently approved for the gangliosidoses, and development of treatments is impaired by limited understanding of the natural history of these diseases. Objective The objective of this study is to improve understanding of the juvenile gangliosidoses phenotypes and the late-infantile phenotypic subtype. Methods Through a prospective natural history study of subjects with juvenile GM1- and GM2-gangliosidosis, a timeline of clinical changes was developed for the classic juvenile phenotypes and the late-infantile phenotypes and results of serial neurodevelopmental testing was analyzed. Results Several candidate ‘outcome measures’ were identified: changes in ambulation and verbalization skills, the communication domain from neurodevelopmental testing and the caregiver-reported socialization domain. Conclusions The most common symptoms leading caregivers to seek a genetic diagnosis were changes in ambulation and verbalization.
Collapse
Affiliation(s)
- Kelly E King
- Department of Pediatrics, University of Minnesota, 2450 Riverside Avenue, Minneapolis, MN 55454-1450, USA
| | - Sarah Kim
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 420 Delaware St SE, MMC 446, Minneapolis, MN 55455-0341, USA.,Advanced Therapies Program, University of Minnesota (UMMC) and Fairview Hospitals, Minneapolis, MN 55454, USA
| | - Chester B Whitley
- Department of Pediatrics, University of Minnesota, 2450 Riverside Avenue, Minneapolis, MN 55454-1450, USA.,Gene Therapy Center, University of Minnesota, 420 Delaware St SE, MMC 391, Minneapolis, MN 55455-0341, USA.,Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 420 Delaware St SE, MMC 446, Minneapolis, MN 55455-0341, USA.,Advanced Therapies Program, University of Minnesota (UMMC) and Fairview Hospitals, Minneapolis, MN 55454, USA
| | - Jeanine R Jarnes-Utz
- Department of Pediatrics, University of Minnesota, 2450 Riverside Avenue, Minneapolis, MN 55454-1450, USA.,Gene Therapy Center, University of Minnesota, 420 Delaware St SE, MMC 391, Minneapolis, MN 55455-0341, USA.,Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 420 Delaware St SE, MMC 446, Minneapolis, MN 55455-0341, USA.,Advanced Therapies Program, University of Minnesota (UMMC) and Fairview Hospitals, Minneapolis, MN 55454, USA
| |
Collapse
|
25
|
Pant DC, Aguilera-Albesa S, Pujol A. Ceramide signalling in inherited and multifactorial brain metabolic diseases. Neurobiol Dis 2020; 143:105014. [PMID: 32653675 DOI: 10.1016/j.nbd.2020.105014] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/13/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
In recent years, research on sphingolipids, particularly ceramides, has attracted increased attention, revealing the important roles and many functions of these molecules in several human neurological disorders. The nervous system is enriched with important classes of sphingolipids, e.g., ceramide and its derivatives, which compose the major portion of this group, particularly in the form of myelin. Ceramides have also emerged as important nodes for lipid signalling, both inside the cell and between cells. Until recently, knowledge about ceramides in the nervous system was limited, but currently, multiple links between ceramide signalling and neurological diseases have been reported. Alterations in the regulation of ceramide pathobiology have been shown to influence the risk of developing neurometabolic diseases. Thus, these molecules are critically important in the maintenance and development of the nervous system and are culprits or major contributors to the development of brain disorders, either inherited or multifactorial. In the present review, we highlight the critical role of ceramide signalling in several different neurological disorders as well as the effects of their perturbations and discuss how this emerging class of bioactive sphingolipids has attracted interest in the field of neurological diseases.
Collapse
Affiliation(s)
- Devesh C Pant
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Sergio Aguilera-Albesa
- Pediatric Neurology Unit, Department of Pediatrics, Navarra Health Service Hospital, Irunlarrea 4, 310620 Pamplona, Spain; Navarrabiomed-Miguel Servet Research Foundation, Pamplona, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, IDIBELL, Hospital Duran i Reynals, Gran Via 199, 08908, L'Hospitalet de Llobregat, Barcelona, Spain; Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain; Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain.
| |
Collapse
|
26
|
Lang FM, Korner P, Harnett M, Karunakara A, Tifft CJ. The natural history of Type 1 infantile GM1 gangliosidosis: A literature-based meta-analysis. Mol Genet Metab 2020; 129:228-235. [PMID: 31937438 PMCID: PMC7093236 DOI: 10.1016/j.ymgme.2019.12.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/26/2019] [Accepted: 12/29/2019] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Type 1 GM1 gangliosidosis is an ultra-rare, rapidly fatal lysosomal storage disorder, with life expectancy of <3 years of age. To date, only one prospective natural history study of limited size has been reported. Thus, there is a need for additional research to provide a better understanding of the progression of this disease. We have leveraged the past two decades of medical literature to conduct the first comprehensive retrospective study characterizing the natural history of Type 1 GM1 gangliosidosis. OBJECTIVES The objectives of this study were to establish a large sample of patients from the literature in order to identify: 1) clinically distinguishing factors between Type 1 and Type 2 GM1 gangliosidosis, 2) age at first symptom onset, first hospital admission, diagnosis, and death, 3) time to onset of common clinical findings, and 4) timing of developmental milestone loss. METHODS PubMed was searched with the keyword "GM1 Gangliosidosis" and for articles from the year 2000 onwards. A preliminary review of these results was conducted to establish subtype classification criteria for inclusion of only Type 1 patients, resulting in 44 articles being selected to generate the literature dataset of 154 Type 1 GM1 gangliosidosis patients. Key clinical events of these patient cases were recorded from the articles. RESULTS Comprehensive subtyping criteria for Type 1 GM1 gangliosidosis were created, and clinical events, including onset, diagnosis, death, and symptomology, were mapped over time. In this dataset, average age of diagnosis was 8.7 months, and average age of death was 18.9 months. DISCUSSION This analysis demonstrates the predictable clinical course of this disease, as almost all patients experienced significant multi-organ system dysfunction and neurodevelopmental regression, particularly in the 6- to 18-month age range. Patients were diagnosed at a late age relative to disease progression, indicating the need for improved public awareness and screening. CONCLUSION This study highlights the significant burden of illness in this disease and provides critical natural history data to drive earlier diagnosis, inform clinical trial design, and facilitate family counseling.
Collapse
Affiliation(s)
- Frederick M Lang
- Axovant Sciences, a subsidiary of Axovant Gene Therapies (Axovant), United States of America
| | - Paul Korner
- Axovant Sciences, a subsidiary of Axovant Gene Therapies (Axovant), United States of America
| | - Mark Harnett
- Axovant Sciences, a subsidiary of Axovant Gene Therapies (Axovant), United States of America
| | - Ajith Karunakara
- Axovant Sciences, a subsidiary of Axovant Gene Therapies (Axovant), United States of America
| | - Cynthia J Tifft
- Office of the Clinical Director & Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health (NHGRI), United States of America.
| |
Collapse
|
27
|
Bi-functional IgG-lysosomal enzyme fusion proteins for brain drug delivery. Sci Rep 2019; 9:18632. [PMID: 31819150 PMCID: PMC6901507 DOI: 10.1038/s41598-019-55136-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/03/2019] [Indexed: 11/12/2022] Open
Abstract
Most lysosomal storage disorders affect the central nervous system. However, lysosomal enzymes do not cross the blood-brain barrier (BBB), and intravenous enzyme infusion is not effective for the brain. Lysosomal enzymes can be re-engineered for BBB transport as IgG-enzyme fusion proteins, where the IgG domain is a monoclonal antibody (MAb) against an endogenous BBB receptor/transporter, and which acts as a molecular Trojan horse to deliver the enzyme to brain. However, the problem is retention of high enzyme activity following enzyme fusion to the IgG. The present investigation shows this is possible with a versatile approach that employs fusion of the enzyme to either the IgG heavy chain or light chain using a long flexible linker. The model IgG is a chimeric monoclonal antibody (MAb) against the human insulin receptor (HIR). The enzyme activity of the HIRMAb-enzyme fusion protein is preserved for hexosaminidase A, which is mutated in Tay Sachs disease, for protein palmitoylthioesterase-1, which is mutated in Batten disease type 1, acid sphingomyelinase, which is mutated in Niemann Pick disease type A, and beta galactosidase-1, which is mutated in GM1 gangliosidosis.
Collapse
|
28
|
Taghian T, Marosfoi MG, Puri AS, Cataltepe OI, King RM, Diffie EB, Maguire AS, Martin DR, Fernau D, Batista AR, Kuchel T, Christou C, Perumal R, Chandra S, Gamlin PD, Bertrand SG, Flotte TR, McKenna-Yasek D, Tai PWL, Aronin N, Gounis MJ, Sena-Esteves M, Gray-Edwards HL. A Safe and Reliable Technique for CNS Delivery of AAV Vectors in the Cisterna Magna. Mol Ther 2019; 28:411-421. [PMID: 31813800 DOI: 10.1016/j.ymthe.2019.11.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 11/29/2022] Open
Abstract
Global gene delivery to the CNS has therapeutic importance for the treatment of neurological disorders that affect the entire CNS. Due to direct contact with the CNS, cerebrospinal fluid (CSF) is an attractive route for CNS gene delivery. A safe and effective route to achieve global gene distribution in the CNS is needed, and administration of genes through the cisterna magna (CM) via a suboccipital puncture results in broad distribution in the brain and spinal cord. However, translation of this technique to clinical practice is challenging due to the risk of serious and potentially fatal complications in patients. Herein, we report development of a gene therapy delivery method to the CM through adaptation of an intravascular microcatheter, which can be safely navigated intrathecally under fluoroscopic guidance. We examined the safety, reproducibility, and distribution/transduction of this method in sheep using a self-complementary adeno-associated virus 9 (scAAV9)-GFP vector. This technique was used to treat two Tay-Sachs disease patients (30 months old and 7 months old) with AAV gene therapy. No adverse effects were observed during infusion or post-treatment. This delivery technique is a safe and minimally invasive alternative to direct infusion into the CM, achieving broad distribution of AAV gene transfer to the CNS.
Collapse
Affiliation(s)
- Toloo Taghian
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Miklos G Marosfoi
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Ajit S Puri
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Neurological Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Oguz I Cataltepe
- Department of Neurological Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Robert M King
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Elise B Diffie
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA
| | - Anne S Maguire
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Auburn University, Auburn, AL 36849, USA; Department of Anatomy, Physiology and Pharmacology, Auburn University, AL 36849, USA
| | - Deborah Fernau
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ana Rita Batista
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Tim Kuchel
- South Australian Health and Medical Research Institute, Gillies Plains, SA 5086, Australia
| | - Chris Christou
- South Australian Health and Medical Research Institute, Gillies Plains, SA 5086, Australia
| | - Raj Perumal
- South Australian Health and Medical Research Institute, Gillies Plains, SA 5086, Australia
| | | | - Paul D Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stephanie G Bertrand
- Department of Environmental Population Health, Cummings Veterinary School at Tufts University, Grafton, MA 01536, USA
| | - Terence R Flotte
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Diane McKenna-Yasek
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Phillip W L Tai
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Neil Aronin
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Matthew J Gounis
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Miguel Sena-Esteves
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| |
Collapse
|
29
|
Chen JC, Luu AR, Wise N, Angelis RD, Agrawal V, Mangini L, Vincelette J, Handyside B, Sterling H, Lo MJ, Wong H, Galicia N, Pacheco G, Van Vleet J, Giaramita A, Fong S, Roy SM, Hague C, Lawrence R, Bullens S, Christianson TM, d'Azzo A, Crawford BE, Bunting S, LeBowitz JH, Yogalingam G. Intracerebroventricular enzyme replacement therapy with β-galactosidase reverses brain pathologies due to GM1 gangliosidosis in mice. J Biol Chem 2019; 295:13532-13555. [PMID: 31481471 PMCID: PMC7521651 DOI: 10.1074/jbc.ra119.009811] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/10/2019] [Indexed: 01/16/2023] Open
Abstract
Autosomal recessive mutations in the galactosidase β1 (GLB1) gene cause lysosomal β-gal deficiency, resulting in accumulation of galactose-containing substrates and onset of the progressive and fatal neurodegenerative lysosomal storage disease, GM1 gangliosidosis. Here, an enzyme replacement therapy (ERT) approach in fibroblasts from GM1 gangliosidosis patients with recombinant human β-gal (rhβ-gal) produced in Chinese hamster ovary cells enabled direct and precise rhβ-gal delivery to acidified lysosomes. A single, low dose (3 nm) of rhβ-gal was sufficient for normalizing β-gal activity and mediating substrate clearance for several weeks. We found that rhβ-gal uptake by the fibroblasts is dose-dependent and saturable and can be competitively inhibited by mannose 6-phosphate, suggesting cation-independent, mannose 6-phosphate receptor–mediated endocytosis from the cell surface. A single intracerebroventricularly (ICV) administered dose of rhβ-gal (100 μg) resulted in broad bilateral biodistribution of rhβ-gal to critical regions of pathology in a mouse model of GM1 gangliosidosis. Weekly ICV dosing of rhβ-gal for 8 weeks substantially reduced brain levels of ganglioside and oligosaccharide substrates and reversed well-established secondary neuropathology. Of note, unlike with the ERT approach, chronic lentivirus-mediated GLB1 overexpression in the GM1 gangliosidosis patient fibroblasts caused accumulation of a prelysosomal pool of β-gal, resulting in activation of the unfolded protein response and endoplasmic reticulum stress. This outcome was unsurprising in light of our in vitro biophysical findings for rhβ-gal, which include pH-dependent and concentration-dependent stability and dynamic self-association. Collectively, our results highlight that ICV-ERT is an effective therapeutic intervention for managing GM1 gangliosidosis potentially more safely than with gene therapy approaches.
Collapse
Affiliation(s)
- Joseph C Chen
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Amanda R Luu
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Nathan Wise
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Rolando De Angelis
- Process Sciences, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Vishal Agrawal
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Linley Mangini
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Jon Vincelette
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Britta Handyside
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Harry Sterling
- Process Sciences, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Melanie J Lo
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Hio Wong
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Nicole Galicia
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Glenn Pacheco
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Jeremy Van Vleet
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | | | - Sylvia Fong
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Sushmita M Roy
- Process Sciences, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Chuck Hague
- Process Sciences, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Roger Lawrence
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Sherry Bullens
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | | | - Alessandra d'Azzo
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105
| | - Brett E Crawford
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | - Stuart Bunting
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949
| | | | - Gouri Yogalingam
- Research, BioMarin Pharmaceutical, Inc., Novato, California 94949.
| |
Collapse
|
30
|
Labounek R, Mai K, Mueller B, Ellinwood NM, Dickson P, Nestrasil I. In-vivo cortical thickness estimation from high-resolution T 1w MRI scans in healthy and mucopolysaccharidosis affected dogs. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2019; 2019:2848-2851. [PMID: 31946486 PMCID: PMC11366836 DOI: 10.1109/embc.2019.8856826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cortical thickness measurement estimated from high-resolution anatomical MRI scans may serve as a marker of cortical atrophy in clinical research applications. Most of the working algorithms and pipelines are optimized for human in-vivo data analyses that offer robust and reproducible measures. As animal-models are widely utilized in many preclinical phases of clinical trials the need for an optimized automated MRI data analysis to yield reliable data is warranted. We present a processing pipeline optimized for cortical thickness estimation of canine brains in native and template spaces. Preliminary results of 5 healthy and 5 mucopolysaccharidosis (MPS) dogs demonstrate single-canine mean/median cortical thickness in range of 2.69-3.58mm in native space and 3.26-4.15mm in template space. Our MRI generated values exceed previous histological measurements (observed mean about 2mm) in limited literature reports. Randomly selected manual measures corroborated the ranges defined by estimated cortical thickness probability density functions. Geometric transformations between native and template spaces change absolute mean/median cortical thickness values, but do not change the data nature and properties since the Pearson correlation coefficients between different space estimates were 0.84 for mean values and 0.89 for median values. No significant difference in total cortical thickness between MPS and age-and gender-matched dogs was observed.
Collapse
|
31
|
Substrate Reduction Therapy for Sandhoff Disease through Inhibition of Glucosylceramide Synthase Activity. Mol Ther 2019; 27:1495-1506. [PMID: 31208914 DOI: 10.1016/j.ymthe.2019.05.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/10/2019] [Accepted: 05/19/2019] [Indexed: 12/12/2022] Open
Abstract
Neuronopathic glycosphingolipidoses are a sub-group of lysosomal storage disorders for which there are presently no effective therapies. Here, we evaluated the potential of substrate reduction therapy (SRT) using an inhibitor of glucosylceramide synthase (GCS) to decrease the synthesis of glucosylceramide (GL1) and related glycosphingolipids. The substrates that accumulate in Sandhoff disease (e.g., ganglioside GM2 and its nonacylated derivative, lyso-GM2) are distal to the drug target, GCS. Treatment of Sandhoff mice with a GCS inhibitor that has demonstrated CNS access (Genz-682452) reduced the accumulation of GL1 and GM2, as well as a variety of disease-associated substrates in the liver and brain. Concomitant with these effects was a significant decrease in the expression of CD68 and glycoprotein non-metastatic melanoma B protein (Gpnmb) in the brain, indicating a reduction in microgliosis in the treated mice. Moreover, using in vivo imaging, we showed that the monocytic biomarker translocator protein (TSPO), which was elevated in Sandhoff mice, was normalized following Genz-682452 treatment. These positive effects translated in turn into a delay (∼28 days) in loss of motor function and coordination, as measured by rotarod latency, and a significant increase in longevity (∼17.5%). Together, these results support the development of SRT for the treatment of gangliosidoses, particularly in patients with residual enzyme activity.
Collapse
|
32
|
Lan XR, Qiu JW, Li H, Cai XR, Song YZ. [Identification and pathogenicity prediction of a novel GLB1 variant c.101T>C (p.Ile34Thr) in an infant with GM1 gangliosidosis]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:71-76. [PMID: 30675867 PMCID: PMC7390173 DOI: 10.7499/j.issn.1008-8830.2019.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/10/2018] [Indexed: 06/09/2023]
Abstract
GM1 gangliosidosis is an autosomal recessive disorder caused by galactosidase beta1 (GLB1) gene variants which affect the activity of β-galactosidase (GLB). GLB dysfunction causes abnormalities in the degradation of GM1 and its accumulation in lysosome. This article reports the clinical and genetic features of a child with GM1 gangliosidosis. The girl, aged 2 years and 5 months, was referred to the hospital due to motor developmental regression for more than one year. Physical examination showed binocular deflection and horizontal nystagmus, but no abnormality was found on fundoscopy. The girl had increased muscular tone of the extremities, limitation of motion of the elbow, knee, and ankle joints, and hyperactive patellar tendon reflex. Blood biochemical examination showed a significant increase in aspartate aminotransferase. The 24-hour electroencephalographic monitoring detected frequent seizure attacks and diffuse θ wave activity, especially in the right hemisphere. Head magnetic resonance imaging showed thinner white matter in the periventricular region and diffuse high T2WI signal with unclear boundary. Three-dimensional reconstruction of white matter fiber tracts by diffusion tensor imaging showed smaller and thinner white matter fiber tracts, especially in the right hemisphere. Genetic analysis showed that the girl had compound heterozygous mutations of c.446C>T (p.Ser149Phe) and c.101T>C (p.Ile34Thr) in the GLB1 gene from her parents, among which c.101T>C (p.Ile34Thr) had not been reported in the literatures. The girl was finally diagnosed with GM1 gangliosidosis. Her conditions were not improved after antiepileptic treatment and rehabilitation training for 2 months.
Collapse
Affiliation(s)
- Xue-Rong Lan
- Department of Pediatrics, First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | | | | | | | | |
Collapse
|
33
|
Solovyeva VV, Shaimardanova AA, Chulpanova DS, Kitaeva KV, Chakrabarti L, Rizvanov AA. New Approaches to Tay-Sachs Disease Therapy. Front Physiol 2018; 9:1663. [PMID: 30524313 PMCID: PMC6256099 DOI: 10.3389/fphys.2018.01663] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
Tay-Sachs disease belongs to the group of autosomal-recessive lysosomal storage metabolic disorders. This disease is caused by β-hexosaminidase A (HexA) enzyme deficiency due to various mutations in α-subunit gene of this enzyme, resulting in GM2 ganglioside accumulation predominantly in lysosomes of nerve cells. Tay-Sachs disease is characterized by acute neurodegeneration preceded by activated microglia expansion, macrophage and astrocyte activation along with inflammatory mediator production. In most cases, the disease manifests itself during infancy, the “infantile form,” which characterizes the most severe disorders of the nervous system. The juvenile form, the symptoms of which appear in adolescence, and the most rare form with late onset of symptoms in adulthood are also described. The typical features of Tay-Sachs disease are muscle weakness, ataxia, speech, and mental disorders. Clinical symptom severity depends on residual HexA enzymatic activity associated with some mutations. Currently, Tay-Sachs disease treatment is based on symptom relief and, in case of the late-onset form, on the delay of progression. There are also clinical reports of substrate reduction therapy using miglustat and bone marrow or hematopoietic stem cell transplantation. At the development stage there are methods of Tay-Sachs disease gene therapy using adeno- or adeno-associated viruses as vectors for the delivery of cDNA encoding α and β HexA subunit genes. Effectiveness of this approach is evaluated in α or β HexA subunit defective model mice or Jacob sheep, in which Tay-Sachs disease arises spontaneously and is characterized by the same pathological features as in humans. This review discusses the possibilities of new therapeutic strategies in Tay-Sachs disease therapy aimed at preventing neurodegeneration and neuroinflammation.
Collapse
Affiliation(s)
- Valeriya V Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Alisa A Shaimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Daria S Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Kristina V Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Lisa Chakrabarti
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|