1
|
Osher E, Anis Y, Singer-Shapiro R, Urshanski N, Unger T, Albeck S, Bogin O, Weisinger G, Kohen F, Valevski A, Fattal-Valevski A, Sagi L, Weitman M, Shenberger Y, Sagiv N, Navon R, Wilchek M, Stern N. Treating late-onset Tay Sachs disease: Brain delivery with a dual trojan horse protein. Mol Ther Methods Clin Dev 2024; 32:101300. [PMID: 39211733 PMCID: PMC11357852 DOI: 10.1016/j.omtm.2024.101300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/13/2024] [Indexed: 09/04/2024]
Abstract
Tay-Sachs (TS) disease is a neurodegenerative disease resulting from mutations in the gene encoding the α-subunit (HEXA) of lysosomal β-hexosaminidase A (HexA). We report that (1) recombinant HEXA alone increased HexA activity and decreased GM2 content in human TS glial cells and peripheral mononuclear blood cells; 2) a recombinant chimeric protein composed of HEXA linked to two blood-brain barrier (BBB) entry elements, a transferrin receptor binding sequence and granulocyte-colony stimulating factor, associates with HEXB in vitro; reaches human cultured TS cells lysosomes and mouse brain cells, especially neurons, in vivo; lowers GM2 in cultured human TS cells; lowers whole brain GM2 concentration by approximately 40% within 6 weeks, when injected intravenously (IV) to adult TS-mutant mice mimicking the slow course of late-onset TS; and increases forelimbs grip strength. Hence, a chimeric protein equipped with dual BBB entry elements can transport a large protein such as HEXA to the brain, decrease the accumulation of GM2, and improve muscle strength, thereby providing potential treatment for late-onset TS.
Collapse
Affiliation(s)
- Esther Osher
- The Sagol Center for Epigenetics and Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yossi Anis
- The Sagol Center for Epigenetics and Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center, Tel Aviv, Israel
| | - Ruth Singer-Shapiro
- The Sagol Center for Epigenetics and Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center, Tel Aviv, Israel
| | - Nataly Urshanski
- The Sagol Center for Epigenetics and Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center, Tel Aviv, Israel
| | - Tamar Unger
- Department of Structural Proteomics, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Albeck
- Department of Structural Proteomics, Weizmann Institute of Science, Rehovot, Israel
| | - Oren Bogin
- The Sagol Center for Epigenetics and Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center, Tel Aviv, Israel
| | - Gary Weisinger
- The Sagol Center for Epigenetics and Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center, Tel Aviv, Israel
| | - Fortune Kohen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Liora Sagi
- Pediatric Neurology Unit, Tel Aviv-Sourasky Medical Center, Tel Aviv, Israel
| | - Michal Weitman
- The Chemistry Department, Bar Ian University, Ramat Gan, Israel
| | | | - Nadav Sagiv
- The Sagol Center for Epigenetics and Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center, Tel Aviv, Israel
| | - Ruth Navon
- Department of Human Molecular Genetics & Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Meir Wilchek
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Naftali Stern
- The Sagol Center for Epigenetics and Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv-Sourasky Medical Center, Tel Aviv, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
2
|
Metovic J, Li Y, Gong Y, Eichler F. Gene therapy for the leukodystrophies: From preclinical animal studies to clinical trials. Neurotherapeutics 2024; 21:e00443. [PMID: 39276676 PMCID: PMC11418141 DOI: 10.1016/j.neurot.2024.e00443] [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: 06/11/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/17/2024] Open
Abstract
Leukodystrophies are progressive single gene disorders affecting the white matter of the brain. Several gene therapy trials are in progress to address the urgent unmet need for this patient population. We performed a comprehensive literature review of all gene therapy clinical trials listed in www.clinicaltrials.gov through August 2024, and the relevant preclinical studies that enabled clinical translation. Of the approximately 50 leukodystrophies described to date, only eight have existing gene therapy clinical trials: metachromatic leukodystrophy, X-linked adrenoleukodystrophy, globoid cell leukodystrophy, Canavan disease, giant axonal neuropathy, GM2 gangliosidoses, Alexander disease and Pelizaeus-Merzbacher disease. What led to the emergence of gene therapy trials for these specific disorders? What preclinical data or disease context was enabling? For each of these eight disorders, we first describe its pathophysiology and clinical presentation. We discuss the impact of gene therapy delivery route, targeted cell type, delivery modality, dosage, and timing on therapeutic efficacy. We note that use of allogeneic hematopoietic stem cell transplantation in some leukodystrophies allowed for an accelerated path to clinic even in the absence of available animal models. In other leukodystrophies, small and large animal model studies enabled clinical translation of experimental gene therapies. Human clinical trials for the leukodystrophies include ex vivo lentiviral gene delivery, in vivo AAV-mediated gene delivery, and intrathecal antisense oligonucleotide approaches. We outline adverse events associated with each modality focusing specifically on genotoxicity and immunotoxicity. We review monitoring and management of events related to insertional mutagenesis and immune responses. The data presented in this review show that gene therapy, while promising, requires systematic monitoring to account for the precarious disease biology and the adverse events associated with new technology.
Collapse
Affiliation(s)
- Jasna Metovic
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yedda Li
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yi Gong
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
3
|
Ryckman AE, Deschenes NM, Quinville BM, Osmon KJ, Mitchell M, Chen Z, Gray SJ, Walia JS. Intrathecal delivery of a bicistronic AAV9 vector expressing β-hexosaminidase A corrects Sandhoff disease in a murine model: A dosage study. Mol Ther Methods Clin Dev 2024; 32:101168. [PMID: 38205442 PMCID: PMC10777117 DOI: 10.1016/j.omtm.2023.101168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 11/30/2023] [Indexed: 01/12/2024]
Abstract
The pathological accumulation of GM2 ganglioside associated with Tay-Sachs disease (TSD) and Sandhoff disease (SD) occurs in individuals who possess mutant forms of the heterodimer β-hexosaminidase A (Hex A) because of mutation of the HEXA and HEXB genes, respectively. With a lack of approved therapies, patients experience rapid neurological decline resulting in early death. A novel bicistronic vector carrying both HEXA and HEXB previously demonstrated promising results in mouse models of SD following neonatal intravenous administration, including significant reduction in GM2 accumulation, increased levels of Hex A, and a 2-fold extension of survival. The aim of the present study was to identify an optimal dose of the bicistronic vector in 6-week-old SD mice by an intrathecal route of administration along with transient immunosuppression, to inform possible clinical translation. Three doses of the bicistronic vector were tested: 2.5e11, 1.25e11, and 0.625e11 vector genomes per mouse. The highest dose provided the greatest increase in biochemical and behavioral parameters, such that treated mice lived to a median age of 56 weeks (>3 times the lifespan of the SD controls). These results have direct implications in deciding a human equivalent dose for TSD/SD and have informed the approval of a clinical trial application (NCT04798235).
Collapse
Affiliation(s)
- Alex E. Ryckman
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Natalie M. Deschenes
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Brianna M. Quinville
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Karlaina J.L. Osmon
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Melissa Mitchell
- Medical Genetics/Departments of Pediatrics, Queen’s University, Kingston, ON K7L 2V7, Canada
| | - Zhilin Chen
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Steven J. Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jagdeep S. Walia
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
- Medical Genetics/Departments of Pediatrics, Queen’s University, Kingston, ON K7L 2V7, Canada
| |
Collapse
|
4
|
Wang A, Chen C, Mei C, Liu S, Xiang C, Fang W, Zhang F, Xu Y, Chen S, Zhang Q, Bai X, Lin A, Neculai D, Xia B, Ye C, Zou J, Liang T, Feng XH, Li X, Shen C, Xu P. Innate immune sensing of lysosomal dysfunction drives multiple lysosomal storage disorders. Nat Cell Biol 2024; 26:219-234. [PMID: 38253667 DOI: 10.1038/s41556-023-01339-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/15/2023] [Indexed: 01/24/2024]
Abstract
Lysosomal storage disorders (LSDs), which are characterized by genetic and metabolic lysosomal dysfunctions, constitute over 60 degenerative diseases with considerable health and economic burdens. However, the mechanisms driving the progressive death of functional cells due to lysosomal defects remain incompletely understood, and broad-spectrum therapeutics against LSDs are lacking. Here, we found that various gene abnormalities that cause LSDs, including Hexb, Gla, Npc1, Ctsd and Gba, all shared mutual properties to robustly autoactivate neuron-intrinsic cGAS-STING signalling, driving neuronal death and disease progression. This signalling was triggered by excessive cytoplasmic congregation of the dsDNA and DNA sensor cGAS in neurons. Genetic ablation of cGAS or STING, digestion of neuronal cytosolic dsDNA by DNase, and repair of neuronal lysosomal dysfunction alleviated symptoms of Sandhoff disease, Fabry disease and Niemann-Pick disease, with substantially reduced neuronal loss. We therefore identify a ubiquitous mechanism mediating the pathogenesis of a variety of LSDs, unveil an inherent connection between lysosomal defects and innate immunity, and suggest a uniform strategy for curing LSDs.
Collapse
Affiliation(s)
- Ailian Wang
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, China
| | - Chen Chen
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chen Mei
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, China
| | - Shengduo Liu
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Cong Xiang
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Wen Fang
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Fei Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yifan Xu
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Shasha Chen
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, China
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, China
| | - Aifu Lin
- MOE Laboratory of Biosystem Homeostasis and Protection, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Dante Neculai
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Xia
- Department of Thoracic Cancer, Affiliated Hangzhou Cancer Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Cunqi Ye
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Jian Zou
- Eye Center of the Second Affiliated Hospital, Institutes of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Xin-Hua Feng
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Xinran Li
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China.
| | - Chengyong Shen
- Department of Neurobiology of The First Affiliated Hospital, Institute of Translational Medicine, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Pinglong Xu
- MOE Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- Institute of Intelligent Medicine, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou, China.
- Cancer Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
5
|
Allende ML, Lee YT, Byrnes C, Li C, Tuymetova G, Bakir JY, Nicoli ER, James VK, Brodbelt JS, Tifft CJ, Proia RL. Sialidase NEU3 action on GM1 ganglioside is neuroprotective in GM1 gangliosidosis. J Lipid Res 2023; 64:100463. [PMID: 37871851 PMCID: PMC10694597 DOI: 10.1016/j.jlr.2023.100463] [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: 09/13/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023] Open
Abstract
GM1 gangliosidosis is a neurodegenerative disorder caused by mutations in the GLB1 gene, which encodes lysosomal β-galactosidase. The enzyme deficiency blocks GM1 ganglioside catabolism, leading to accumulation of GM1 ganglioside and asialo-GM1 ganglioside (GA1 glycolipid) in brain. This disease can present in varying degrees of severity, with the level of residual β-galactosidase activity primarily determining the clinical course. Glb1 null mouse models, which completely lack β-galactosidase expression, exhibit a less severe form of the disease than expected from the comparable deficiency in humans, suggesting a potential species difference in the GM1 ganglioside degradation pathway. We hypothesized this difference may involve the sialidase NEU3, which acts on GM1 ganglioside to produce GA1 glycolipid. To test this hypothesis, we generated Glb1/Neu3 double KO (DKO) mice. These mice had a significantly shorter lifespan, increased neurodegeneration, and more severe ataxia than Glb1 KO mice. Glb1/Neu3 DKO mouse brains exhibited an increased GM1 ganglioside to GA1 glycolipid ratio compared with Glb1 KO mice, indicating that NEU3 mediated GM1 ganglioside to GA1 glycolipid conversion in Glb1 KO mice. The expression of genes associated with neuroinflammation and glial responses were enhanced in Glb1/Neu3 DKO mice compared with Glb1 KO mice. Mouse NEU3 more efficiently converted GM1 ganglioside to GA1 glycolipid than human NEU3 did. Our findings highlight NEU3's role in ameliorating the consequences of Glb1 deletion in mice, provide insights into NEU3's differential effects between mice and humans in GM1 gangliosidosis, and offer a potential therapeutic approach for reducing toxic GM1 ganglioside accumulation in GM1 gangliosidosis patients.
Collapse
Affiliation(s)
- Maria L Allende
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Y Terry Lee
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Colleen Byrnes
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cuiling Li
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Galina Tuymetova
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jenna Y Bakir
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Elena-Raluca Nicoli
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Virginia K James
- Department of Chemistry, University of Texas at Austin, Austin, TX, USA
| | | | - Cynthia J Tifft
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Richard L Proia
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Deschenes NM, Cheng C, Khanal P, Quinville BM, Ryckman AE, Mitchell M, Pshezhetsky AV, Walia JS. Characterization of a phenotypically severe animal model for human AB-Variant GM2 gangliosidosis. Front Mol Neurosci 2023; 16:1242814. [PMID: 38098938 PMCID: PMC10720325 DOI: 10.3389/fnmol.2023.1242814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/23/2023] [Indexed: 12/17/2023] Open
Abstract
AB-Variant GM2 gangliosidosis (ABGM2) is a rare and lethal genetic disorder caused by mutations in the GM2A gene that lead to fatal accumulation of GM2 gangliosides (GM2) in neurons of the central nervous system (CNS). GM2A encodes a transport protein known as GM2 activator (GM2A) protein, which is essential for degrading GM2 into their GM3 form. ABGM2 presents in infantile-, juvenile-, and adult-onset forms; of the three, the infantile-onset is the most prominent, and by far the most severe, as evidenced by high levels of GM2 accumulation, widespread neurodegeneration, and death by the age of 4. Gm2a-/- mice are commonly used as a model of ABGM2. These mice are characterized by phenotypes most representative of predicted adult-onset form of ABGM2, which include moderate GM2 accumulation and mild neurological defects. This mild phenotype has been attributed to compensation by alternative GM2 degradation pathways mediated by sialidase, neuraminidase 3 (NEU3), a pathway that is more prominent in mice than humans. To assess the extent to which NEU3 contributes to GM2 degradation, we generated double knock-out (Gm2a-/-Neu3-/-) mice. Compellingly, these mice present with a clinical phenotype resembling that of a more severe ABGM2, including ataxia, reduced mobility and coordination, weight loss, poor body scores, and lethality by 6-7 months. Furthermore, these phenotypes correlate with a dramatic increase in GM2 accumulation in the CNS compared to levels observed in either Gm2a-/- or Neu3-/- mice. Taken together, these studies, for the first-time, confirm that the mild neurological phenotype of Gm2a-/- mice is due to compensatory activity on GM2 catabolism through an alternate breakdown pathway involving NEU3. These studies support the use of double knockout mice as a novel and highly relevant model for pre-clinical drug studies in a more severe form of ABGM2.
Collapse
Affiliation(s)
| | - Camilyn Cheng
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON, Canada
| | - Prem Khanal
- Department of Pediatrics, Queen’s University, Kingston, ON, Canada
| | | | - Alex E. Ryckman
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON, Canada
| | - Melissa Mitchell
- Department of Pediatrics, Queen’s University, Kingston, ON, Canada
| | - Alexey V. Pshezhetsky
- Centre Hospitalier Universitaire Sainte-Justine Research Centre, Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Jagdeep S. Walia
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON, Canada
- Department of Pediatrics, Queen’s University, Kingston, ON, Canada
| |
Collapse
|
7
|
Vyas M, Deschenes NM, Osmon KJL, Chen Z, Ahmad I, Kot S, Thompson P, Richmond C, Gray SJ, Walia JS. Efficacy of Adeno-Associated Virus Serotype 9-Mediated Gene Therapy for AB-Variant GM2 Gangliosidosis. Int J Mol Sci 2023; 24:14611. [PMID: 37834060 PMCID: PMC10572999 DOI: 10.3390/ijms241914611] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/06/2023] [Accepted: 09/16/2023] [Indexed: 10/15/2023] Open
Abstract
GM2 gangliosidoses are a group of neurodegenerative lysosomal storage disorders that are characterized by the accumulation of GM2 gangliosides (GM2), leading to rapid neurological decline and death. The hydrolysis of GM2 requires the specific synthesis, processing, and combination of products of three genes-HEXA, HEXB, and GM2A-within the cell's lysosomes. Mutations in these genes result in Tay-Sachs disease, Sandhoff disease, or AB-variant GM2 gangliosidosis (ABGM2), respectively. ABGM2, the rarest of the three types, is characterized by a mutation in the GM2A gene, which encodes the GM2 activator (GM2A) protein. Being a monogenic disease, gene therapy is a plausible and likely effective method of treatment for ABGM2. This study aimed at assessing the effects of administering a one-time intravenous treatment of single-stranded Adeno-associated virus serotype 9 (ssAAV9)-GM2A viral vector at a dose of 1 × 1014 vector genomes (vg) per kilogram per mouse in an ABGM2 mouse model (Gm2a-/-). ssAAV9-GM2A was administered at 1-day (neonatal) or 6-weeks of age (adult-stage). The results demonstrated that, in comparison to Gm2a-/- mice that received a vehicle injection, the treated mice had reduced GM2 accumulation within the central nervous system and had long-term persistence of vector genomes in the brain and liver. This proof-of-concept study is a step forward towards the development of a clinically therapeutic approach for the treatment of patients with ABGM2.
Collapse
Affiliation(s)
- Meera Vyas
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Natalie M. Deschenes
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Karlaina J. L. Osmon
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Zhilin Chen
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (Z.C.)
| | - Imtiaz Ahmad
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Shalini Kot
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (Z.C.)
| | - Patrick Thompson
- Department of Pediatrics, Queen’s University, Kingston, ON K7L 2V7, Canada;
| | - Chris Richmond
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (Z.C.)
| | - Steven J. Gray
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jagdeep S. Walia
- Centre for Neuroscience Studies, Queen’s University, Kingston, ON K7L 3N6, Canada
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (Z.C.)
- Department of Pediatrics, Queen’s University, Kingston, ON K7L 2V7, Canada;
| |
Collapse
|
8
|
Carvelli L, Hermo L, O’Flaherty C, Oko R, Pshezhetsky AV, Morales CR. Effects of Heparan sulfate acetyl-CoA: Alpha-glucosaminide N-acetyltransferase (HGSNAT) inactivation on the structure and function of epithelial and immune cells of the testis and epididymis and sperm parameters in adult mice. PLoS One 2023; 18:e0292157. [PMID: 37756356 PMCID: PMC10529547 DOI: 10.1371/journal.pone.0292157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Heparan sulfate (HS), an abundant component of the apical cell surface and basement membrane, belongs to the glycosaminoglycan family of carbohydrates covalently linked to proteins called heparan sulfate proteoglycans. After endocytosis, HS is degraded in the lysosome by several enzymes, including heparan-alpha-glucosaminide N-acetyltransferase (HGSNAT), and in its absence causes Mucopolysaccharidosis III type C (Sanfilippo type C). Since endocytosis occurs in epithelial cells of the testis and epididymis, we examined the morphological effects of Hgsnat inactivation in these organs. In the testis, Hgsnat knockout (Hgsnat-Geo) mice revealed statistically significant decrease in tubule and epithelial profile area of seminiferous tubules. Electron microscopy (EM) analysis revealed cross-sectional tubule profiles with normal and moderately to severely altered appearances. Abnormalities in Sertoli cells and blood-testis barrier and the absence of germ cells in some tubules were noted along with altered morphology of sperm, sperm motility parameters and a reduction in fertilization rates in vitro. Along with quantitatively increased epithelial and tubular profile areas in the epididymis, EM demonstrated significant accumulations of electrolucent lysosomes in the caput-cauda regions that were reactive for cathepsin D and prosaposin antibodies. Lysosomes with similar storage materials were also found in basal, clear and myoid cells. In the mid/basal region of the epithelium of caput-cauda regions of KO mice, large vacuolated cells, unreactive for cytokeratin 5, a basal cell marker, were identified morphologically as epididymal mononuclear phagocytes (eMPs). The cytoplasm of the eMPs was occupied by a gigantic lysosome suggesting an active role of these cells in removing debris from the epithelium. Some eMPs were found in proximity to T-lymphocytes, a feature of dendritic cells. Taken together, our results reveal that upon Hgsnat inactivation, morphological alterations occur to the testis affecting sperm morphology and motility parameters and abnormal lysosomes in epididymal epithelial cells, indicative of a lysosomal storage disease.
Collapse
Affiliation(s)
- Lorena Carvelli
- IHEM-CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Louis Hermo
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Cristian O’Flaherty
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
- Department of Surgery (Urology Division), McGill University, Montréal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Richard Oko
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Canada
| | - Alexey V. Pshezhetsky
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
- Sainte-Justine University Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Carlos R. Morales
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Deschenes NM, Cheng C, Ryckman AE, Quinville BM, Khanal P, Mitchell M, Chen Z, Sangrar W, Gray SJ, Walia JS. Biochemical Correction of GM2 Ganglioside Accumulation in AB-Variant GM2 Gangliosidosis. Int J Mol Sci 2023; 24:ijms24119217. [PMID: 37298170 DOI: 10.3390/ijms24119217] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 06/12/2023] Open
Abstract
GM2 gangliosidosis is a group of genetic disorders that result in the accumulation of GM2 ganglioside (GM2) in brain cells, leading to progressive central nervous system (CNS) atrophy and premature death in patients. AB-variant GM2 gangliosidosis (ABGM2) arises from loss-of-function mutations in the GM2 activator protein (GM2AP), which is essential for the breakdown of GM2 in a key catabolic pathway required for CNS lipid homeostasis. In this study, we show that intrathecal delivery of self-complementary adeno-associated virus serotype-9 (scAAV9) harbouring a functional human GM2A transgene (scAAV9.hGM2A) can prevent GM2 accumulation in in GM2AP-deficient mice (Gm2a-/- mice). Additionally, scAAV9.hGM2A efficiently distributes to all tested regions of the CNS within 14 weeks post-injection and remains detectable for the lifespan of these animals (up to 104 weeks). Remarkably, GM2AP expression from the transgene scales with increasing doses of scAAV9.hGM2A (0.5, 1.0 and 2.0 × 1011 vector genomes (vg) per mouse), and this correlates with dose-dependent correction of GM2 accumulation in the brain. No severe adverse events were observed, and comorbidities in treated mice were comparable to those in disease-free cohorts. Lastly, all doses yielded corrective outcomes. These data indicate that scAAV9.hGM2A treatment is relatively non-toxic and tolerable, and biochemically corrects GM2 accumulation in the CNS-the main cause of morbidity and mortality in patients with ABGM2. Importantly, these results constitute proof-of-principle for treating ABGM2 with scAAV9.hGM2A by means of a single intrathecal administration and establish a foundation for future preclinical research.
Collapse
Affiliation(s)
- Natalie M Deschenes
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Camilyn Cheng
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Alex E Ryckman
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Brianna M Quinville
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Prem Khanal
- Department of Pediatrics, Queen's University, Kingston, ON K7L 2V7, Canada
| | - Melissa Mitchell
- Department of Pediatrics, Queen's University, Kingston, ON K7L 2V7, Canada
| | - Zhilin Chen
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Waheed Sangrar
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Steven J Gray
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jagdeep S Walia
- Centre for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
- Department of Pediatrics, Queen's University, Kingston, ON K7L 2V7, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
10
|
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
|
11
|
A patient advocating for transparent science in rare disease research. Orphanet J Rare Dis 2023; 18:14. [PMID: 36658594 PMCID: PMC9854194 DOI: 10.1186/s13023-022-02557-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/14/2022] [Accepted: 10/12/2022] [Indexed: 01/20/2023] Open
Abstract
300 million people live with at least one of 6,000 rare diseases worldwide. However, rare disease research is not always reviewed with scrutiny, making it susceptible to what the author refers to as nontransparent science. Nontransparent science can obscure animal model flaws, misguide medicine regulators and drug developers, delay or frustrate orphan drug development, or waste limited resources for rare disease research. Flawed animal models not only lack pharmacologic relevance, but also give rise to issue of clinical translatability. Sadly, these consequences and risks are grossly overlooked. Nontransparency in science can take many forms, such as premature publication of animal models without clinically significant data, not providing corrections when flaws to the model are discovered, lack of warning of critical study limitations, missing critical control data, questionable data quality, surprising results without a sound explanation, failure to rule out potential factors which may affect study conclusions, lack of sufficient detail for others to replicate the study, dubious authorship and study accountability. Science has no boarders, neither does nontransparent science. Nontransparent science can happen irrespective of the researcher's senority, institutional affiliation or country. As a patient-turned researcher suffering from Bietti crystalline dystrophy (BCD), I use BCD as an example to analyze various forms of nontransparent science in rare disease research. This article analyzes three papers published by different research groups on Cyp4v3-/-, high-fat diet (HFD)-Cyp4v3-/-, and Exon1-Cyp4v3-/- mouse models of BCD. As the discussion probes various forms of nontransparent science, the flaws of these knockout mouse models are uncovered. These mouse models do not mimic BCD in humans nor do they address the lack of Cyp4v3 (murine ortholog of human CYP4V2) expression in wild type (WT) mouse retina which is markedly different from CYP4V2 expression in human retina. Further, this article discusses the impact of nontransparent science on drug development which can lead to significant delays ultimately affecting the patients. Lessons from BCD research can be helpful to all those suffering from rare diseases. As a patient, I call for transparent science in rare disease research.
Collapse
|
12
|
Sandhoff R, Sandhoff K. Neuronal Ganglioside and Glycosphingolipid (GSL) Metabolism and Disease : Cascades of Secondary Metabolic Errors Can Generate Complex Pathologies (in LSDs). ADVANCES IN NEUROBIOLOGY 2023; 29:333-390. [PMID: 36255681 DOI: 10.1007/978-3-031-12390-0_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Glycosphingolipids (GSLs) are a diverse group of membrane components occurring mainly on the surfaces of mammalian cells. They and their metabolites have a role in intercellular communication, serving as versatile biochemical signals (Kaltner et al, Biochem J 476(18):2623-2655, 2019) and in many cellular pathways. Anionic GSLs, the sialic acid containing gangliosides (GGs), are essential constituents of neuronal cell surfaces, whereas anionic sulfatides are key components of myelin and myelin forming oligodendrocytes. The stepwise biosynthetic pathways of GSLs occur at and lead along the membranes of organellar surfaces of the secretory pathway. After formation of the hydrophobic ceramide membrane anchor of GSLs at the ER, membrane-spanning glycosyltransferases (GTs) of the Golgi and Trans-Golgi network generate cell type-specific GSL patterns for cellular surfaces. GSLs of the cellular plasma membrane can reach intra-lysosomal, i.e. luminal, vesicles (ILVs) by endocytic pathways for degradation. Soluble glycoproteins, the glycosidases, lipid binding and transfer proteins and acid ceramidase are needed for the lysosomal catabolism of GSLs at ILV-membrane surfaces. Inherited mutations triggering a functional loss of glycosylated lysosomal hydrolases and lipid binding proteins involved in GSL degradation cause a primary lysosomal accumulation of their non-degradable GSL substrates in lysosomal storage diseases (LSDs). Lipid binding proteins, the SAPs, and the various lipids of the ILV-membranes regulate GSL catabolism, but also primary storage compounds such as sphingomyelin (SM), cholesterol (Chol.), or chondroitin sulfate can effectively inhibit catabolic lysosomal pathways of GSLs. This causes cascades of metabolic errors, accumulating secondary lysosomal GSL- and GG- storage that can trigger a complex pathology (Breiden and Sandhoff, Int J Mol Sci 21(7):2566, 2020).
Collapse
Affiliation(s)
- Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Konrad Sandhoff
- LIMES, c/o Kekule-Institute for Organic Chemistry and Biochemistry, University of Bonn, Bonn, Germany.
| |
Collapse
|
13
|
Sengul T, Can M, Ateş N, Seyrantepe V. Autophagic flux is impaired in the brain tissue of Tay-Sachs disease mouse model. PLoS One 2023; 18:e0280650. [PMID: 36928510 PMCID: PMC10019743 DOI: 10.1371/journal.pone.0280650] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 01/05/2023] [Indexed: 03/18/2023] Open
Abstract
Tay-Sachs disease is a lethal lysosomal storage disorder caused by mutations in the HexA gene encoding the α subunit of the lysosomal β-hexosaminidase enzyme (HEXA). Abnormal GM2 ganglioside accumulation causes progressive deterioration in the central nervous system in Tay-Sachs patients. Hexa-/- mouse model failed to display abnormal phenotype. Recently, our group generated Hexa-/-Neu3-/- mouse showed severe neuropathological indications similar to Tay-Sachs patients. Despite excessive GM2 ganglioside accumulation in the brain and visceral organs, the regulation of autophagy has not been clarified yet in the Tay-Sachs disease mouse model. Therefore, we investigated distinct steps of autophagic flux using markers including LC3 and p62 in four different brain regions from the Hexa-/-Neu3-/- mice model of Tay-Sachs disease. Our data revealed accumulated autophagosomes and autophagolysosomes indicating impairment in autophagic flux in the brain. We suggest that autophagy might be a new therapeutic target for the treatment of devastating Tay-Sachs disease.
Collapse
Affiliation(s)
- Tugce Sengul
- İzmir Institute of Technology, Department of Molecular Biology and Genetics, İzmir, Turkey
| | - Melike Can
- İzmir Institute of Technology, Department of Molecular Biology and Genetics, İzmir, Turkey
| | - Nurselin Ateş
- İzmir Institute of Technology, Department of Molecular Biology and Genetics, İzmir, Turkey
| | - Volkan Seyrantepe
- İzmir Institute of Technology, Department of Molecular Biology and Genetics, İzmir, Turkey
- İzmir Institute of Technology, IYTEDEHAM, İzmir, Turkey
- * E-mail:
| |
Collapse
|
14
|
Gaudioso Á, Silva TP, Ledesma MD. Models to study basic and applied aspects of lysosomal storage disorders. Adv Drug Deliv Rev 2022; 190:114532. [PMID: 36122863 DOI: 10.1016/j.addr.2022.114532] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 08/05/2022] [Accepted: 09/04/2022] [Indexed: 01/24/2023]
Abstract
The lack of available treatments and fatal outcome in most lysosomal storage disorders (LSDs) have spurred research on pathological mechanisms and novel therapies in recent years. In this effort, experimental methodology in cellular and animal models have been developed, with aims to address major challenges in many LSDs such as patient-to-patient variability and brain condition. These techniques and models have advanced knowledge not only of LSDs but also for other lysosomal disorders and have provided fundamental insights into the biological roles of lysosomes. They can also serve to assess the efficacy of classical therapies and modern drug delivery systems. Here, we summarize the techniques and models used in LSD research, which include both established and recently developed in vitro methods, with general utility or specifically addressing lysosomal features. We also review animal models of LSDs together with cutting-edge technology that may reduce the need for animals in the study of these devastating diseases.
Collapse
Affiliation(s)
- Ángel Gaudioso
- Centro Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Teresa P Silva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | | |
Collapse
|
15
|
Can M, Sengül T, Demir SA, İnci OK, Basırlı H, Seyrantepe V. Analysis of Brain Lipids in the Early-Onset Tay–Sachs Disease Mouse Model With the Combined Deficiency of β-Hexosaminidase A and Neuraminidase 3. Front Mol Biosci 2022; 9:892248. [PMID: 36003081 PMCID: PMC9393265 DOI: 10.3389/fmolb.2022.892248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction: Tay–Sachs disease is an autosomal recessively inherited lysosomal storage disease that results from loss-of-function mutations in the HEXA gene coding β-hexosaminidase A. HEXA gene deficiency affects the central nervous system owing to GM2 ganglioside accumulation in lysosomes resulting in progressive neurodegeneration in patients. We recently generated a novel mice model with a combined deficiency of β-hexosaminidase A and neuraminidase 3 (Hexa−/−Neu3−/−) that mimics both the neuropathological and clinical abnormalities of early-onset Tay–Sachs disease. Here, we aimed to explore the secondary accumulation of lipids in the brain of Hexa−/−Neu3−/− mice.Materials and Methods: In the cortex and hippocampus of five-month-old WT, Hexa−/−, Neu3−/−, and Hexa−/−Neu3−/− mice, lipid levels belonging to glycerolipids, glycerophospholipids, and sterol lipids were evaluated using a shotgun lipidomics approach. The levels of myelin were also assessed by luxol fast blue staining and immunohistochemistry using antibodies against myelin basic protein. We further examined glycoconjugate and cholesterol levels by periodic acid–Schiff and filipin staining, respectively. Toluidine blue staining was also performed to display axonal degeneration.Results: Among glycerophospholipids, we demonstrated elevated levels of phosphatidylcholine-ether and lysophosphatidylcholine while decreased levels of phosphatidylcholine and phosphatidylserine in both cortex and hippocampus of Hexa−/−Neu3−/− mice. In the glycerolipid class, we showed an alleviated level of sphingomyelin in both cortex and hippocampus, but the higher levels of diacylglycerol and triacylglycerol were detected in only the hippocampus of Hexa−/−Neu3−/− mice. The lower level of sterol was also detected in the cortex of Hexa−/−Neu3−/− mice but not in the hippocampus.Histochemical studies showed a decrease in the myelin level and axonal degeneration indicating neuronal pathology in the brain of Hexa−/−Neu3−/− mice. Although glycoconjugate accumulation was evident both in the cortex and hippocampus, we did not detect any changes in the level of cholesterol.Conclusion: Our results indicate that alterations in lipid metabolism and neuropathology, such as demyelination and axonal degeneration, might be related to the dysfunctionality of lipid-related cellular pathways like autophagy. Understanding of brain-specific lipid alterations contributes to evaluating the effectiveness of treatments in Hexa−/−Neu3−/− mice in future studies.
Collapse
Affiliation(s)
- Melike Can
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Tugce Sengül
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | | | - Orhan K. İnci
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Hande Basırlı
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
| | - Volkan Seyrantepe
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Turkey
- Izmir Institute of Technology, IYTEDEHAM, Izmir, Turkey
- *Correspondence: Volkan Seyrantepe,
| |
Collapse
|
16
|
Haslund-Gourley BS, Aziz PV, Heithoff DM, Restagno D, Fried JC, Ilse MB, Bäumges H, Mahan MJ, Lübke T, Marth JD. Establishment of blood glycosidase activities and their excursions in sepsis. PNAS NEXUS 2022; 1:pgac113. [PMID: 35967980 PMCID: PMC9364217 DOI: 10.1093/pnasnexus/pgac113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023]
Abstract
Glycosidases are hydrolytic enzymes studied principally in the context of intracellular catabolism within the lysosome. Therefore, glycosidase activities are classically measured in experimentally acidified assay conditions reflecting their low pH optima. However, glycosidases are also present in the bloodstream where they may retain sufficient activity to participate in the regulation of glycoprotein half-lives, proteostasis, and disease pathogenesis. We have, herein, established at physiological pH 7.4 in blood plasma and sera the normal ranges of four major glycosidase activities essential for blood glycoprotein remodeling in healthy mice and humans. These activities included β-galactosidase, β-N-acetylglucosaminidase, α-mannosidase, and α-fucosidase. We have identified their origins to include the mammalian genes Glb1, HexB, Man2a1, and Fuca1. In experimental sepsis, excursions of glycosidase activities occurred with differences in host responses to discrete bacterial pathogens. Among similar excursions in human sepsis, the elevation of β-galactosidase activity was a prognostic indicator of increased likelihood of patient death.
Collapse
Affiliation(s)
- Benjamin S Haslund-Gourley
- Sanford-Burnham-Prebys Medical Discovery Institute, Infectious and Inflammatory Diseases Center, La Jolla, CA 92037, USA
| | - Peter V Aziz
- Sanford-Burnham-Prebys Medical Discovery Institute, Infectious and Inflammatory Diseases Center, La Jolla, CA 92037, USA
| | - Douglas M Heithoff
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, CA 93106, USA
| | - Damien Restagno
- Sanford-Burnham-Prebys Medical Discovery Institute, Infectious and Inflammatory Diseases Center, La Jolla, CA 92037, USA
| | - Jeffrey C Fried
- Department of Pulmonary and Critical Care Medicine, Cottage Hospital of Santa Barbara, Santa Barbara, CA 93105, USA
| | - Mai-Britt Ilse
- Department of Chemistry, Biochemistry, Bielefeld University, D-33615, Germany
| | - Hannah Bäumges
- Department of Chemistry, Biochemistry, Bielefeld University, D-33615, Germany
| | - Michael J Mahan
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, CA 93106, USA
| | - Torben Lübke
- Department of Chemistry, Biochemistry, Bielefeld University, D-33615, Germany
| | - Jamey D Marth
- Sanford-Burnham-Prebys Medical Discovery Institute, Infectious and Inflammatory Diseases Center, La Jolla, CA 92037, USA
| |
Collapse
|
17
|
Suetterlin KJ, Männikkö R, Matthews E, Greensmith L, Hanna MG, Bostock H, Tan SV. Excitability properties of mouse and human skeletal muscle fibres compared by muscle velocity recovery cycles. Neuromuscul Disord 2022; 32:347-357. [PMID: 35339342 PMCID: PMC7614892 DOI: 10.1016/j.nmd.2022.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 01/27/2022] [Accepted: 02/22/2022] [Indexed: 11/21/2022]
Abstract
Mouse models of skeletal muscle channelopathies are not phenocopies of human disease. In some cases (e.g. Myotonia Congenita) the phenotype is much more severe, whilst in others (e.g. Hypokalaemic periodic paralysis) rodent physiology is protective. This suggests a species' difference in muscle excitability properties. In humans these can be measured indirectly by the post-impulse changes in conduction velocity, using Muscle Velocity Recovery Cycles (MVRCs). We performed MVRCs in mice and compared their muscle excitability properties with humans. Mouse Tibialis Anterior MVRCs (n = 70) have only one phase of supernormality (increased conduction velocity), which is smaller in magnitude (p = 9 × 10-21), and shorter in duration (p = 3 × 10-24) than human (n = 26). This abbreviated supernormality is followed by a period of late subnormality (reduced velocity) in mice, which overlaps in time with the late supernormality seen in human MVRCs. The period of late subnormality suggests increased t-tubule Na+/K+-pump activity. The subnormal phase in mice was converted to supernormality by blocking ClC-1 chloride channels, suggesting relatively higher chloride conductance in skeletal muscle. Our findings help explain discrepancies in phenotype between mice and humans with skeletal muscle channelopathies and potentially other neuromuscular disorders. MVRCs are a valuable new tool to compare in vivo muscle membrane properties between species and will allow further dissection of the molecular mechanisms regulating muscle excitability.
Collapse
Affiliation(s)
- K J Suetterlin
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; AGE Research Group, NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - R Männikkö
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - E Matthews
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Atkinson Morley Neuromuscular Centre, Department of Neurology, St Georges University Hospitals NHS Foundation Trust, London, United Kingdom
| | - L Greensmith
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - M G Hanna
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - H Bostock
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom
| | - S V Tan
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; Department of Neurology and Clinical Neurophysiology, Guy's & St Thomas' NHS Foundation Trust and Institute of Psychiatry, Psychology & Neuroscience, Division of Neuroscience, King's College London, United Kingdom
| |
Collapse
|
18
|
Benatti HR, Gray-Edwards HL. Adeno-Associated Virus Delivery Limitations for Neurological Indications. Hum Gene Ther 2022; 33:1-7. [PMID: 35049369 DOI: 10.1089/hum.2022.29196.hrb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Hector Ribeiro Benatti
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA.,Department of Radiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
19
|
de Morais-Pinto L, da Veiga ML, Almeida da Anunciação AR. Central nervous system development of cats (Felis catus L. 1758). Res Vet Sci 2021; 141:81-94. [PMID: 34700148 DOI: 10.1016/j.rvsc.2021.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/07/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022]
Abstract
The morphological similarities of vertebrates' embryonic development are used as a criterion for choosing animal models that can be used in biomedical research. This study describes the embryonic and fetal development of the domestic cat's central nervous system from 15 days after conception until birth. In total, fifty-seven samples of embryos and fetuses were carefully dissected and analyzed microscopically. The closure of the neural tube was observed between 14-15th days of gestation. The differentiation of the primordial cerebral vesicles was observed from the 17th day of gestation. On the 19th day of gestation, the formation of the choroid plexus began, and on the 20th day of gestation, the brain and brainstem were well-identified macroscopically. On the 24th day of gestation, four layers of cells from the cerebral cortex were described, and on the 60th day, six layers of cells were present. The cerebellar cortex had the three classic cortical layers at this stage. The morphological aspects of embryonic and fetal development in cats were very similar to the stages of development of the human nervous system. As such, this study provided relevant information that highlights the domestic cat as an animal model option for preclinical research on infectious and non-infectious neurological diseases in humans.
Collapse
Affiliation(s)
- Luciano de Morais-Pinto
- Laboratório de Design Anatômico/LabDA, Departamento de Morfologia, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil.
| | - Marcelo Leite da Veiga
- Laboratório de Morfofisiologia Experimental e Comparada/LABITEX, Departamento de Morfologia, Universidade Federal de Santa Maria, Rio Grande do Sul, Brazil
| | | |
Collapse
|
20
|
Osmon KJ, Thompson P, Woodley E, Karumuthil-Melethil S, Heindel C, Keimel JG, Kaemmerer WF, Gray SJ, Walia JS. Treatment of GM2 Gangliosidosis in Adult Sandhoff Mice using an Intravenous Self-Complementary Hexosaminidase Vector. Curr Gene Ther 2021; 22:262-276. [PMID: 34530708 DOI: 10.2174/1566523221666210916153051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/01/2021] [Accepted: 07/16/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND GM2 gangliosidosis is a neurodegenerative, lysosomal storage disease caused by the deficiency of β-hexosaminidase A enzyme (HexA), an α/β-subunit heterodimer. A novel variant of the human hexosaminidase α-subunit, coded by HEXM, has previously been shown to form a stable homodimer, HexM, that hydrolyzes GM2 gangliosides (GM2) in vivo. MATERIALS & METHODS The current study assessed the efficacy of intravenous (IV) delivery of a self-complementary adeno-associated virus serotype 9 (scAAV9) vector incorporating the HEXM transgene, scAAV9/HEXM, including the outcomes based on the dosages provided to the Sandhoff (SD) mice. Six-week-old SD mice were injected with either 2.5E+12 vector genomes (low dose, LD) or 1.0E+13 vg (high dose, HD). We hypothesized that when examining the dosage comparison for scAAV9/HEXM in adult SD mice, the HD group would have more beneficial outcomes than the LD cohort. Assessments included survival, behavioral outcomes, vector biodistribution, and enzyme activity within the central nervous system. RESULTS Toxicity was observed in the HD cohort, with 8 of 14 mice dying within one month of the injection. As compared to untreated SD mice, which have typical survival of 16 weeks, the LD cohort and the remaining HD mice had a significant survival benefit with an average/median survival of 40.6/34.5 and 55.9/56.7 weeks, respectively. Significant behavioral, biochemical and molecular benefits were also observed. The second aim of the study was to investigate the effects of IV mannitol infusions on the biodistribution of the LD scAAV9/HEXM vector and the survival of the SD mice. Increases in both the biodistribution of the vector as well as the survival benefit (average/median of 41.6/49.3 weeks) were observed. CONCLUSION These results demonstrate the potential benefit and critical limitations of the treatment of GM2 gangliosidosis using IV delivered AAV vectors.
Collapse
Affiliation(s)
- Karlaina Jl Osmon
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario. Canada
| | - Patrick Thompson
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario. Canada
| | - Evan Woodley
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario. Canada
| | | | - Cliff Heindel
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina. United States
| | - John G Keimel
- New Hope Research Foundation, North Oaks, Minnesota. United States
| | | | - Steven J Gray
- Gene Therapy Center, University of North Carolina, Chapel Hill, North Carolina. United States
| | - Jagdeep S Walia
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario. Canada
| |
Collapse
|
21
|
Efficient and precise generation of Tay-Sachs disease model in rabbit by prime editing system. Cell Discov 2021; 7:50. [PMID: 34230459 PMCID: PMC8260710 DOI: 10.1038/s41421-021-00276-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/22/2021] [Indexed: 11/08/2022] Open
|
22
|
Kot S, Karumuthil-Melethil S, Woodley E, Zaric V, Thompson P, Chen Z, Lykken E, Keimel JG, Kaemmerer WF, Gray SJ, Walia JS. Investigating Immune Responses to the scAAV9- HEXM Gene Therapy Treatment in Tay-Sachs Disease and Sandhoff Disease Mouse Models. Int J Mol Sci 2021; 22:ijms22136751. [PMID: 34201771 PMCID: PMC8268035 DOI: 10.3390/ijms22136751] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 12/15/2022] Open
Abstract
GM2 gangliosidosis disorders are a group of neurodegenerative diseases that result from a functional deficiency of the enzyme β-hexosaminidase A (HexA). HexA consists of an α- and β-subunit; a deficiency in either subunit results in Tay–Sachs Disease (TSD) or Sandhoff Disease (SD), respectively. Viral vector gene transfer is viewed as a potential method of treating these diseases. A recently constructed isoenzyme to HexA, called HexM, has the ability to effectively catabolize GM2 gangliosides in vivo. Previous gene transfer studies have revealed that the scAAV9-HEXM treatment can improve survival in the murine SD model. However, it is speculated that this treatment could elicit an immune response to the carrier capsid and “non-self”-expressed transgene. This study was designed to assess the immunocompetence of TSD and SD mice, and test the immune response to the scAAV9-HEXM gene transfer. HexM vector-treated mice developed a significant anti-HexM T cell response and antibody response. This study confirms that TSD and SD mouse models are immunocompetent, and that gene transfer expression can create an immune response in these mice. These mouse models could be utilized for investigating methods of mitigating immune responses to gene transfer-expressed “non-self” proteins, and potentially improve treatment efficacy.
Collapse
Affiliation(s)
- Shalini Kot
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (S.K.); (E.W.)
| | - Subha Karumuthil-Melethil
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.K.-M.); (V.Z.); (E.L.); (S.J.G.)
| | - Evan Woodley
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (S.K.); (E.W.)
| | - Violeta Zaric
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.K.-M.); (V.Z.); (E.L.); (S.J.G.)
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Patrick Thompson
- Medical Genetics, Department of Pediatrics, Queen’s University, Kingston, ON K7L 2V7, Canada; (P.T.); (Z.C.)
| | - Zhilin Chen
- Medical Genetics, Department of Pediatrics, Queen’s University, Kingston, ON K7L 2V7, Canada; (P.T.); (Z.C.)
| | - Erik Lykken
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.K.-M.); (V.Z.); (E.L.); (S.J.G.)
| | - John G. Keimel
- New Hope Research Foundation, North Oaks, MN 55127, USA; (J.G.K.); (W.F.K.)
| | | | - Steven J. Gray
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.K.-M.); (V.Z.); (E.L.); (S.J.G.)
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jagdeep S. Walia
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (S.K.); (E.W.)
- Medical Genetics, Department of Pediatrics, Queen’s University, Kingston, ON K7L 2V7, Canada; (P.T.); (Z.C.)
- Correspondence:
| |
Collapse
|
23
|
Bankoti K, Generotti C, Hwa T, Wang L, O'Malley BW, Li D. Advances and challenges in adeno-associated viral inner-ear gene therapy for sensorineural hearing loss. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:209-236. [PMID: 33850952 PMCID: PMC8010215 DOI: 10.1016/j.omtm.2021.03.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There is growing attention and effort focused on treating the root cause of sensorineural hearing loss rather than managing associated secondary characteristic features. With recent substantial advances in understanding sensorineural hearing-loss mechanisms, gene delivery has emerged as a promising strategy for the biological treatment of hearing loss associated with genetic dysfunction. There are several successful and promising proof-of-principle examples of transgene deliveries in animal models; however, there remains substantial further progress to be made in these avenues before realizing their clinical application in humans. Herein, we review different aspects of development, ongoing preclinical studies, and challenges to the clinical transition of transgene delivery of the inner ear toward the restoration of lost auditory and vestibular function.
Collapse
Affiliation(s)
- Kamakshi Bankoti
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles Generotti
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tiffany Hwa
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lili Wang
- Department of Medicine, Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bert W O'Malley
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daqing Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Cavender C, Mangini L, Van Vleet JL, Corado C, McCullagh E, Gray-Edwards HL, Martin DR, Crawford BE, Lawrence R. Natural history study of glycan accumulation in large animal models of GM2 gangliosidoses. PLoS One 2020; 15:e0243006. [PMID: 33259552 PMCID: PMC7707493 DOI: 10.1371/journal.pone.0243006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 11/12/2020] [Indexed: 11/19/2022] Open
Abstract
β-hexosaminidase is an enzyme responsible for the degradation of gangliosides, glycans, and other glycoconjugates containing β-linked hexosamines that enter the lysosome. GM2 gangliosidoses, such as Tay-Sachs and Sandhoff, are lysosomal storage disorders characterized by β-hexosaminidase deficiency and subsequent lysosomal accumulation of its substrate metabolites. These two diseases result in neurodegeneration and early mortality in children. A significant difference between these two disorders is the accumulation in Sandhoff disease of soluble oligosaccharide metabolites that derive from N- and O-linked glycans. In this paper we describe our results from a longitudinal biochemical study of a feline model of Sandhoff disease and an ovine model of Tay-Sachs disease to investigate the accumulation of GM2/GA2 gangliosides, a secondary biomarker for phospholipidosis, bis-(monoacylglycero)-phosphate, and soluble glycan metabolites in both tissue and fluid samples from both animal models. While both Sandhoff cats and Tay-Sachs sheep accumulated significant amounts of GM2 and GA2 gangliosides compared to age-matched unaffected controls, the Sandhoff cats having the more severe disease, accumulated larger amounts of gangliosides compared to Tay-Sachs sheep in their occipital lobes. For monitoring glycan metabolites, we developed a quantitative LC/MS assay for one of these free glycans in order to perform longitudinal analysis. The Sandhoff cats showed significant disease-related increases in this glycan in brain and in other matrices including urine which may provide a useful clinical tool for measuring disease severity and therapeutic efficacy. Finally, we observed age-dependent increasing accumulation for a number of analytes, especially in Sandhoff cats where glycosphingolipid, phospholipid, and glycan levels showed incremental increases at later time points without signs of peaking. This large animal natural history study for Sandhoff and Tay-Sachs is the first of its kind, providing insight into disease progression at the biochemical level. This report may help in the development and testing of new therapies to treat these disorders.
Collapse
Affiliation(s)
- Catlyn Cavender
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Linley Mangini
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Jeremy L. Van Vleet
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Carley Corado
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Emma McCullagh
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Douglas R. Martin
- Scott-Ritchey Research Center and Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States of America
| | - Brett E. Crawford
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Roger Lawrence
- Research, BioMarin Pharmaceutical Inc., Novato, CA, United States of America
- * E-mail:
| |
Collapse
|
25
|
Demir SA, Timur ZK, Ateş N, Martínez LA, Seyrantepe V. GM2 ganglioside accumulation causes neuroinflammation and behavioral alterations in a mouse model of early onset Tay-Sachs disease. J Neuroinflammation 2020; 17:277. [PMID: 32951593 PMCID: PMC7504627 DOI: 10.1186/s12974-020-01947-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 09/01/2020] [Indexed: 11/10/2022] Open
Abstract
Background Tay-Sachs disease (TSD), a type of GM2-gangliosidosis, is a progressive neurodegenerative lysosomal storage disorder caused by mutations in the α subunit of the lysosomal β-hexosaminidase enzyme. This disease is characterized by excessive accumulation of GM2 ganglioside, predominantly in the central nervous system. Although Tay-Sachs patients appear normal at birth, the progressive accumulation of undegraded GM2 gangliosides in neurons leads to death. Recently, an early onset Tay-Sachs disease mouse model, with genotype Hexa−/−Neu3−/−, was generated. Progressive accumulation of GM2 led to premature death of the double KO mice. Importantly, this double-deficient mouse model displays typical features of Tay-Sachs patients, such as cytoplasmic vacuolization of nerve cells, deterioration of Purkinje cells, neuronal death, deceleration in movement, ataxia, and tremors. GM2-gangliosidosis is characterized by acute neurodegeneration preceded by activated microglia expansion, macrophage, and astrocyte activation, along with the production of inflammatory mediators. However, the mechanism of disease progression in Hexa−/−Neu3−/− mice, relevant to neuroinflammation is poorly understood. Method In this study, we investigated the onset and progression of neuroinflammatory changes in the cortex, cerebellum, and retina of Hexa−/−Neu3−/− mice and control littermates by using a combination of molecular genetics and immunochemical procedures. Results We found elevated levels of pro-inflammatory cytokine and chemokine transcripts, such as Ccl2, Ccl3, Ccl4, and Cxcl10 and also extensive microglial and astrocyte activation and proliferation, accompanied by peripheral blood mononuclear cell infiltration in the vicinity of neurons and oligodendrocytes. Behavioral tests demonstrated a high level of anxiety, and age-dependent loss in both spatial learning and fear memory in Hexa−/−Neu3−/− mice compared with that in the controls. Conclusion Altogether, our data suggest that Hexa−/−Neu3−/− mice display a phenotype similar to Tay-Sachs patients suffering from chronic neuroinflammation triggered by GM2 accumulation. Furthermore, our work contributes to better understanding of the neuropathology in a mouse model of early onset Tay-Sachs disease.
Collapse
Affiliation(s)
- Seçil Akyıldız Demir
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Izmir, Turkey
| | - Zehra Kevser Timur
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Izmir, Turkey
| | - Nurselin Ateş
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Izmir, Turkey
| | - Luis Alarcón Martínez
- Institute of Neurological Science and Psychiatry, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| | - Volkan Seyrantepe
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430, Izmir, Turkey.
| |
Collapse
|
26
|
Ryckman AE, Brockhausen I, Walia JS. Metabolism of Glycosphingolipids and Their Role in the Pathophysiology of Lysosomal Storage Disorders. Int J Mol Sci 2020; 21:E6881. [PMID: 32961778 PMCID: PMC7555265 DOI: 10.3390/ijms21186881] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/04/2020] [Accepted: 09/12/2020] [Indexed: 12/11/2022] Open
Abstract
Glycosphingolipids (GSLs) are a specialized class of membrane lipids composed of a ceramide backbone and a carbohydrate-rich head group. GSLs populate lipid rafts of the cell membrane of eukaryotic cells, and serve important cellular functions including control of cell-cell signaling, signal transduction and cell recognition. Of the hundreds of unique GSL structures, anionic gangliosides are the most heavily implicated in the pathogenesis of lysosomal storage diseases (LSDs) such as Tay-Sachs and Sandhoff disease. Each LSD is characterized by the accumulation of GSLs in the lysosomes of neurons, which negatively interact with other intracellular molecules to culminate in cell death. In this review, we summarize the biosynthesis and degradation pathways of GSLs, discuss how aberrant GSL metabolism contributes to key features of LSD pathophysiology, draw parallels between LSDs and neurodegenerative proteinopathies such as Alzheimer's and Parkinson's disease and lastly, discuss possible therapies for patients.
Collapse
Affiliation(s)
| | - Inka Brockhausen
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 2V5, Canada;
| | - Jagdeep S. Walia
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 2V5, Canada;
| |
Collapse
|
27
|
Eikelberg D, Lehmbecker A, Brogden G, Tongtako W, Hahn K, Habierski A, Hennermann JB, Naim HY, Felmy F, Baumgärtner W, Gerhauser I. Axonopathy and Reduction of Membrane Resistance: Key Features in a New Murine Model of Human G M1-Gangliosidosis. J Clin Med 2020; 9:jcm9041004. [PMID: 32252429 PMCID: PMC7230899 DOI: 10.3390/jcm9041004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/29/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
GM1-gangliosidosis is caused by a reduced activity of β-galactosidase (Glb1), resulting in intralysosomal accumulations of GM1. The aim of this study was to reveal the pathogenic mechanisms of GM1-gangliosidosis in a new Glb1 knockout mouse model. Glb1−/− mice were analyzed clinically, histologically, immunohistochemically, electrophysiologically and biochemically. Morphological lesions in the central nervous system were already observed in two-month-old mice, whereas functional deficits, including ataxia and tremor, did not start before 3.5-months of age. This was most likely due to a reduced membrane resistance as a compensatory mechanism. Swollen neurons exhibited intralysosomal storage of lipids extending into axons and amyloid precursor protein positive spheroids. Additionally, axons showed a higher kinesin and lower dynein immunoreactivity compared to wildtype controls. Glb1−/− mice also demonstrated loss of phosphorylated neurofilament positive axons and a mild increase in non-phosphorylated neurofilament positive axons. Moreover, marked astrogliosis and microgliosis were found, but no demyelination. In addition to the main storage material GM1, GA1, sphingomyelin, phosphatidylcholine and phosphatidylserine were elevated in the brain. In summary, the current Glb1−/− mice exhibit a so far undescribed axonopathy and a reduced membrane resistance to compensate the functional effects of structural changes. They can be used for detailed examinations of axon–glial interactions and therapy trials of lysosomal storage diseases.
Collapse
Affiliation(s)
- Deborah Eikelberg
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| | - Annika Lehmbecker
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| | - Graham Brogden
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (G.B.); (H.Y.N.)
| | - Witchaya Tongtako
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
- c/o Faculty of Veterinary Science, Prince of Sonkla University, 5 Karnjanavanich Rd., Hat Yai, Songkhla 90110, Thailand
| | - Kerstin Hahn
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| | - Andre Habierski
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| | - Julia B. Hennermann
- Villa Metabolica, University of Mainz, Langenbeckstraße 2, D-55131 Mainz, Germany;
| | - Hassan Y. Naim
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (G.B.); (H.Y.N.)
| | - Felix Felmy
- Department for Physiology and Cell Biology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany;
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
- Correspondence:
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, D-30559 Hannover, Germany; (D.E.); (A.L.); (W.T.); (K.H.); (A.H.); (I.G.)
| |
Collapse
|
28
|
Moro CA, Hanna-Rose W. Animal Model Contributions to Congenital Metabolic Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:225-244. [PMID: 32304075 PMCID: PMC8404832 DOI: 10.1007/978-981-15-2389-2_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Genetic model systems allow researchers to probe and decipher aspects of human disease, and animal models of disease are frequently specifically engineered and have been identified serendipitously as well. Animal models are useful for probing the etiology and pathophysiology of disease and are critical for effective discovery and development of novel therapeutics for rare diseases. Here we review the impact of animal model organism research in three examples of congenital metabolic disorders to highlight distinct advantages of model system research. First, we discuss phenylketonuria research where a wide variety of research fields and models came together to make impressive progress and where a nearly ideal mouse model has been central to therapeutic advancements. Second, we review advancements in Lesch-Nyhan syndrome research to illustrate the role of models that do not perfectly recapitulate human disease as well as the need for multiple models of the same disease to fully investigate human disease aspects. Finally, we highlight research on the GM2 gangliosidoses Tay-Sachs and Sandhoff disease to illustrate the important role of both engineered traditional laboratory animal models and serendipitously identified atypical models in congenital metabolic disorder research. We close with perspectives for the future for animal model research in congenital metabolic disorders.
Collapse
Affiliation(s)
- Corinna A Moro
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA
| | - Wendy Hanna-Rose
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
29
|
Novel bicistronic lentiviral vectors correct β-Hexosaminidase deficiency in neural and hematopoietic stem cells and progeny: implications for in vivo and ex vivo gene therapy of GM2 gangliosidosis. Neurobiol Dis 2019; 134:104667. [PMID: 31682993 DOI: 10.1016/j.nbd.2019.104667] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/28/2019] [Accepted: 10/31/2019] [Indexed: 01/03/2023] Open
Abstract
The favorable outcome of in vivo and ex vivo gene therapy approaches in several Lysosomal Storage Diseases suggests that these treatment strategies might equally benefit GM2 gangliosidosis. Tay-Sachs and Sandhoff disease (the main forms of GM2 gangliosidosis) result from mutations in either the HEXA or HEXB genes encoding, respectively, the α- or β-subunits of the lysosomal β-Hexosaminidase enzyme. In physiological conditions, α- and β-subunits combine to generate β-Hexosaminidase A (HexA, αβ) and β-Hexosaminidase B (HexB, ββ). A major impairment to establishing in vivo or ex vivo gene therapy for GM2 gangliosidosis is the need to synthesize the α- and β-subunits at high levels and with the correct stoichiometric ratio, and to safely deliver the therapeutic products to all affected tissues/organs. Here, we report the generation and in vitro validation of novel bicistronic lentiviral vectors (LVs) encoding for both the murine and human codon optimized Hexa and Hexb genes. We show that these LVs drive the safe and coordinate expression of the α- and β-subunits, leading to supranormal levels of β-Hexosaminidase activity with prevalent formation of a functional HexA in SD murine neurons and glia, murine bone marrow-derived hematopoietic stem/progenitor cells (HSPCs), and human SD fibroblasts. The restoration/overexpression of β-Hexosaminidase leads to the reduction of intracellular GM2 ganglioside storage in transduced and in cross-corrected SD murine neural progeny, indicating that the transgenic enzyme is secreted and functional. Importantly, bicistronic LVs safely and efficiently transduce human neurons/glia and CD34+ HSPCs, which are target and effector cells, respectively, in prospective in vivo and ex vivo GT approaches. We anticipate that these bicistronic LVs may overcome the current requirement of two vectors co-delivering the α- or β-subunits genes. Careful assessment of the safety and therapeutic potential of these bicistronic LVs in the SD murine model will pave the way to the clinical development of LV-based gene therapy for GM2 gangliosidosis.
Collapse
|
30
|
Ogawa Y, Sasanuma Y, Shitara S, Koshizuka A, Okada R, Sakuraba H, Oishi K. Abnormal organization during neurodevelopment in a mouse model of Sandhoff disease. Neurosci Res 2019; 155:12-19. [PMID: 31340161 DOI: 10.1016/j.neures.2019.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 12/24/2022]
Abstract
Sandhoff disease (SD) is a genetic disorder caused by a mutation of HEXB, which is the β-subunit gene of β-hexosaminidase A and B (HexA and HexB) in humans. HEXB mutation reduces HexA and HexB enzymatic activities, and results in the massive accumulation of ganglioside GM2 in the nervous system. Severe phenotypes of SD show progressive neurodegeneration in human infants, and lysosomal dysfunction that may affect the early development of the nervous system. In a previous study, neural stem cells (NSCs) and induced pluripotent stem cells derived from SD model mice, which are Hexb-deficient (Hexb-/-), demonstrated impaired neuronal differentiation. This study investigated early neurodevelopment in vivo using Hexb-/- mice. The structure of adult cerebral cortices of Hexb-/- mice was normal. However, the expression of Sox2, an NSC-related gene, was reduced in the embryonic cerebral cortices of Hexb-/- mice. Moreover, a reduction of early neuronal migration and differentiation was observed in the embryonic cerebral cortices of Hexb-/- mice. In addition, we showed that the production of layer-specific neurons was delayed in somatosensory cerebral cortices of Hexb-/- mice. These findings suggest that the alterations observed in embryonic Hexb-/- mice may contribute to deficits in neurodevelopment of SD.
Collapse
Affiliation(s)
- Yasuhiro Ogawa
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan.
| | - Yayoi Sasanuma
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Shuhei Shitara
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Asuna Koshizuka
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Rieko Okada
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Hitoshi Sakuraba
- Department of Clinical Genetics, Meiji Pharmaceutical University, Tokyo, Japan
| | - Kazuhiko Oishi
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
31
|
Niemir N, Rouvière L, Besse A, Vanier MT, Dmytrus J, Marais T, Astord S, Puech JP, Panasyuk G, Cooper JD, Barkats M, Caillaud C. Intravenous administration of scAAV9-Hexb normalizes lifespan and prevents pathology in Sandhoff disease mice. Hum Mol Genet 2019; 27:954-968. [PMID: 29325092 DOI: 10.1093/hmg/ddy012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 12/30/2017] [Indexed: 12/12/2022] Open
Abstract
Sandhoff disease (SD) is a rare inherited disorder caused by a deficiency of β-hexosaminidase activity which is fatal because no effective treatment is available. A mouse model of Hexb deficiency reproduces the key pathognomonic features of SD patients with severe ubiquitous lysosomal dysfunction, GM2 accumulation, neuroinflammation and neurodegeneration, culminating in death at 4 months. Here, we show that a single intravenous neonatal administration of a self-complementary adeno-associated virus 9 vector (scAAV9) expressing the Hexb cDNA in SD mice is safe and sufficient to prevent disease development. Importantly, we demonstrate for the first time that this treatment results in a normal lifespan (over 700 days) and normalizes motor function assessed by a battery of behavioral tests, with scAAV9-treated SD mice being indistinguishable from wild-type littermates. Biochemical analyses in multiple tissues showed a significant increase in hexosaminidase A activity, which reached 10-15% of normal levels. AAV9 treatment was sufficient to prevent GM2 and GA2 storage almost completely in the cerebrum (less so in the cerebellum), as well as thalamic reactive gliosis and thalamocortical neuron loss in treated Hexb-/- mice. In summary, this study demonstrated a widespread protective effect throughout the entire CNS after a single intravenous administration of the scAAV9-Hexb vector to neonatal SD mice.
Collapse
Affiliation(s)
- Natalia Niemir
- INSERM U1151, Institut Necker Enfants Malades, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Laura Rouvière
- INSERM U1151, Institut Necker Enfants Malades, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Aurore Besse
- Centre of Research in Myology, Institut de Myologie, Sorbonne Universités, UPMC University Paris 06, Inserm UMRS974, GH Pitié Salpêtrière, Paris 75013, France
| | - Marie T Vanier
- INSERM U820, Université de Lyon, Faculté de Médecine Lyon-Est, Lyon 69372, France
| | - Jasmin Dmytrus
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RX, UK
| | - Thibaut Marais
- Centre of Research in Myology, Institut de Myologie, Sorbonne Universités, UPMC University Paris 06, Inserm UMRS974, GH Pitié Salpêtrière, Paris 75013, France
| | - Stéphanie Astord
- Centre of Research in Myology, Institut de Myologie, Sorbonne Universités, UPMC University Paris 06, Inserm UMRS974, GH Pitié Salpêtrière, Paris 75013, France
| | - Jean-Philippe Puech
- Service de Biochimie, Métabolomique et Protéomique, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris 75015, France
| | - Ganna Panasyuk
- INSERM U1151, Institut Necker Enfants Malades, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jonathan D Cooper
- Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology & Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London SE5 9RX, UK.,Department of Pediatrics, Los Angeles Biomedical Research Institute, David Geffen School of Medicine, UCLA, Torrance, CA 90502, USA
| | - Martine Barkats
- Centre of Research in Myology, Institut de Myologie, Sorbonne Universités, UPMC University Paris 06, Inserm UMRS974, GH Pitié Salpêtrière, Paris 75013, France
| | - Catherine Caillaud
- INSERM U1151, Institut Necker Enfants Malades, Paris 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Biochimie, Métabolomique et Protéomique, Hôpital Universitaire Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris 75015, France
| |
Collapse
|
32
|
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: 8.0] [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
|
33
|
Efficacy of a Bicistronic Vector for Correction of Sandhoff Disease in a Mouse Model. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 12:47-57. [PMID: 30534578 PMCID: PMC6279944 DOI: 10.1016/j.omtm.2018.10.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 10/23/2018] [Indexed: 12/01/2022]
Abstract
GM2 gangliosidoses are a family of severe neurodegenerative disorders resulting from a deficiency in the β-hexosaminidase A enzyme. These disorders include Tay-Sachs disease and Sandhoff disease, caused by mutations in the HEXA gene and HEXB gene, respectively. The HEXA and HEXB genes are required to produce the α and β subunits of the β-hexosaminidase A enzyme, respectively. Using a Sandhoff disease mouse model, we tested for the first time the potential of a comparatively lower dose (2.04 × 1013 vg/kg) of systemically delivered single-stranded adeno-associated virus 9 expressing both human HEXB and human HEXA cDNA under the control of a single promoter with a P2A-linked bicistronic vector design to correct the neurological phenotype. A bicistronic design allows maximal overexpression and secretion of the Hex A enzyme. Neonatal mice were injected with either this ssAAV9-HexB-P2A-HexA vector or a vehicle solution via the superficial temporal vein. An increase in survival of 56% compared with vehicle-injected controls and biochemical analysis of the brain tissue and serum revealed an increase in enzyme activity and a decrease in brain GM2 ganglioside buildup. This is a proof-of-concept study showing the “correction efficacy” of a bicistronic AAV9 vector delivered intravenously for GM2 gangliosidoses. Further studies with higher doses are warranted.
Collapse
|
34
|
Oliveira R, Hermo L, Pshezhetsky AV, Morales CR. Presence of aberrant epididymal tubules revealing undifferentiated epithelial cells and absence of spermatozoa in a combined neuraminidase-3 and -4 deficient adult mouse model. PLoS One 2018; 13:e0206173. [PMID: 30359429 PMCID: PMC6201937 DOI: 10.1371/journal.pone.0206173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/08/2018] [Indexed: 11/28/2022] Open
Abstract
Mammalian neuraminidases are responsible for the removal of sialic acids from glycoproteins and glycolipids and function in a variety of biological phenomena such as lysosomal catabolism and control of cell differentiation and growth. Disruption of Neu3 and Neu4 genes has led to the generation of a mouse model revealing severe neurological disorders. In this study a morphological analysis was performed on the epididymis of 3 month-old neu3-/-neu4-/- mice as compared with wild type animals. In neu3-/-neu4-/- mice the majority of tubules of the main epididymal duct were large and lined by differentiated epithelial cells, but revealing lysosomal abnormalities in principal and basally located cells. Of particular note was the presence of aberrant epididymal tubules (ATs) juxtaposed next to the main tubules. ATs were small and of different shapes. Layers of myoid cells encased ATs, which they shared with those of the main tubules, but no interstitial space existed between the two. While some ATs were a dense mass of cells, others revealed a distinct lumen devoid of spermatozoa. The latter revealed an undifferentiated epithelium consisting of cuboidal cells and basal cells, with junctional complexes evident at the luminal front. The absence of spermatozoa from the lumen of the ATs suggests that they were not in contact with the main duct, as also implied by the undifferentiated appearance of the epithelium suggesting lack of lumicrine factors. Despite the presence of ATs, the main duct contained ample spermatozoa, as the neu3-/-neu4-/- mice were fertile. Taken together the data suggest that absence of Neu3 and Neu4 leads to defects in cell adhesion and differentiation of epithelial cells resulting in aberrant tubular offshoots that fail to remain connected with the main duct. Hence Neu3 and Neu 4 play an essential role in the guidance of epithelial cells during early embryonic formation.
Collapse
Affiliation(s)
- Regiana Oliveira
- Department of Anatomy and Cell Biology, McGill University–Montreal, Canada
| | - Louis Hermo
- Department of Anatomy and Cell Biology, McGill University–Montreal, Canada
| | - Alexey V. Pshezhetsky
- Division of Medical Genetics, Centre Hospitalière Universitaire Sainte-Justine, University of Montréal—Montreal, Canada
| | - Carlos R. Morales
- Department of Anatomy and Cell Biology, McGill University–Montreal, Canada
- * E-mail:
| |
Collapse
|
35
|
Sandhoff R, Sandhoff K. Emerging concepts of ganglioside metabolism. FEBS Lett 2018; 592:3835-3864. [PMID: 29802621 DOI: 10.1002/1873-3468.13114] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 11/12/2022]
Abstract
Gangliosides (GGs) are sialic acid-containing glycosphingolipids (GSLs) and major membrane components enriched on cellular surfaces. Biosynthesis of mammalian GGs starts at the cytosolic leaflet of endoplasmic reticulum (ER) membranes with the formation of their hydrophobic ceramide anchors. After intracellular ceramide transfer to Golgi and trans-Golgi network (TGN) membranes, anabolism of GGs, as well as of other GSLs, is catalyzed by membrane-spanning glycosyltransferases (GTs) along the secretory pathway. Combined activity of only a few promiscuous GTs allows for the formation of cell-type-specific glycolipid patterns. Following an exocytotic vesicle flow to the cellular plasma membranes, GGs can be modified by metabolic reactions at or near the cellular surface. For degradation, GGs are endocytosed to reach late endosomes and lysosomes. Whereas membrane-spanning enzymes of the secretory pathway catalyze GSL and GG formation, a cooperation of soluble glycosidases, lipases and lipid-binding cofactors, namely the sphingolipid activator proteins (SAPs), act as the main players of GG and GSL catabolism at intralysosomal luminal vesicles (ILVs).
Collapse
Affiliation(s)
- Roger Sandhoff
- Lipid Pathobiochemistry Group (G131), German Cancer Research Center, Heidelberg, Germany
| | | |
Collapse
|
36
|
Cachón-González MB, Zaccariotto E, Cox TM. Genetics and Therapies for GM2 Gangliosidosis. Curr Gene Ther 2018; 18:68-89. [PMID: 29618308 PMCID: PMC6040173 DOI: 10.2174/1566523218666180404162622] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/10/2018] [Accepted: 01/27/2018] [Indexed: 12/30/2022]
Abstract
Tay-Sachs disease, caused by impaired β-N-acetylhexosaminidase activity, was the first GM2 gangliosidosis to be studied and one of the most severe and earliest lysosomal diseases to be described. The condition, associated with the pathological build-up of GM2 ganglioside, has acquired almost iconic status and serves as a paradigm in the study of lysosomal storage diseases. Inherited as a classical autosomal recessive disorder, this global disease of the nervous system induces developmental arrest with regression of attained milestones; neurodegeneration progresses rapidly to cause premature death in young children. There is no effective treatment beyond palliative care, and while the genetic basis of GM2 gangliosidosis is well established, the molecular and cellular events, from diseasecausing mutations and glycosphingolipid storage to disease manifestations, remain to be fully delineated. Several therapeutic approaches have been attempted in patients, including enzymatic augmentation, bone marrow transplantation, enzyme enhancement, and substrate reduction therapy. Hitherto, none of these stratagems has materially altered the course of the disease. Authentic animal models of GM2 gangliodidosis have facilitated in-depth evaluation of innovative applications such as gene transfer, which in contrast to other interventions, shows great promise. This review outlines current knowledge pertaining the pathobiology as well as potential innovative treatments for the GM2 gangliosidoses.
Collapse
Affiliation(s)
| | - Eva Zaccariotto
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
37
|
Kirkegaard T, Gray J, Priestman DA, Wallom KL, Atkins J, Olsen OD, Klein A, Drndarski S, Petersen NHT, Ingemann L, Smith DA, Morris L, Bornæs C, Jørgensen SH, Williams I, Hinsby A, Arenz C, Begley D, Jäättelä M, Platt FM. Heat shock protein-based therapy as a potential candidate for treating the sphingolipidoses. Sci Transl Med 2017; 8:355ra118. [PMID: 27605553 DOI: 10.1126/scitranslmed.aad9823] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 08/18/2016] [Indexed: 12/17/2022]
Abstract
Lysosomal storage diseases (LSDs) often manifest with severe systemic and central nervous system (CNS) symptoms. The existing treatment options are limited and have no or only modest efficacy against neurological manifestations of disease. We demonstrate that recombinant human heat shock protein 70 (HSP70) improves the binding of several sphingolipid-degrading enzymes to their essential cofactor bis(monoacyl)glycerophosphate in vitro. HSP70 treatment reversed lysosomal pathology in primary fibroblasts from 14 patients with eight different LSDs. HSP70 penetrated effectively into murine tissues including the CNS and inhibited glycosphingolipid accumulation in murine models of Fabry disease (Gla(-/-)), Sandhoff disease (Hexb(-/-)), and Niemann-Pick disease type C (Npc1(-/-)) and attenuated a wide spectrum of disease-associated neurological symptoms in Hexb(-/-) and Npc1(-/-) mice. Oral administration of arimoclomol, a small-molecule coinducer of HSPs that is currently in clinical trials for Niemann-Pick disease type C (NPC), recapitulated the effects of recombinant human HSP70, suggesting that heat shock protein-based therapies merit clinical evaluation for treating LSDs.
Collapse
Affiliation(s)
| | - James Gray
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | | | | | - Jennifer Atkins
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | - Ole Dines Olsen
- Orphazyme ApS, Copenhagen, Denmark. Cell Death and Metabolism Unit, Center for Autophagy, Recycling, and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Alexander Klein
- Institut für Chemie der Humboldt-Universität zu Berlin, Berlin, Germany
| | | | | | | | - David A Smith
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | - Lauren Morris
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | | | | | - Ian Williams
- Department of Pharmacology, University of Oxford, Oxford, U.K
| | | | - Christoph Arenz
- Institut für Chemie der Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Begley
- Institute of Pharmaceutical Science, King's College London, London, U.K
| | - Marja Jäättelä
- Cell Death and Metabolism Unit, Center for Autophagy, Recycling, and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, U.K
| |
Collapse
|
38
|
Functional evaluation of yuzu ( Citrus junos ) extracts containing limonoids and polyamine for life extension. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.09.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
39
|
Seyrantepe V, Demir SA, Timur ZK, Von Gerichten J, Marsching C, Erdemli E, Oztas E, Takahashi K, Yamaguchi K, Ates N, Dönmez Demir B, Dalkara T, Erich K, Hopf C, Sandhoff R, Miyagi T. Murine Sialidase Neu3 facilitates GM2 degradation and bypass in mouse model of Tay-Sachs disease. Exp Neurol 2017; 299:26-41. [PMID: 28974375 DOI: 10.1016/j.expneurol.2017.09.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 09/14/2017] [Accepted: 09/28/2017] [Indexed: 12/29/2022]
Abstract
Tay-Sachs disease is a severe lysosomal storage disorder caused by mutations in Hexa, the gene that encodes for the α subunit of lysosomal β-hexosaminidase A (HEXA), which converts GM2 to GM3 ganglioside. Unexpectedly, Hexa-/- mice have a normal lifespan and show no obvious neurological impairment until at least one year of age. These mice catabolize stored GM2 ganglioside using sialidase(s) to remove sialic acid and form the glycolipid GA2, which is further processed by β-hexosaminidase B. Therefore, the presence of the sialidase (s) allows the consequences of the Hexa defect to be bypassed. To determine if the sialidase NEU3 contributes to GM2 ganglioside degradation, we generated a mouse model with combined deficiencies of HEXA and NEU3. The Hexa-/-Neu3-/- mice were healthy at birth, but died at 1.5 to 4.5months of age. Thin-layer chromatography and mass spectrometric analysis of the brains of Hexa-/-Neu3-/- mice revealed the abnormal accumulation of GM2 ganglioside. Histological and immunohistochemical analysis demonstrated cytoplasmic vacuolation in the neurons. Electron microscopic examination of the brain, kidneys and testes revealed pleomorphic inclusions of many small vesicles and complex lamellar structures. The Hexa-/-Neu3-/- mice exhibited progressive neurodegeneration with neuronal loss, Purkinje cell depletion, and astrogliosis. Slow movement, ataxia, and tremors were the prominent neurological abnormalities observed in these mice. Furthermore, radiographs revealed abnormalities in the skeletal bones of the Hexa-/-Neu3-/- mice. Thus, the Hexa-/-Neu3-/- mice mimic the neuropathological and clinical abnormalities of the classical early-onset Tay-Sachs patients, and provide a suitable model for the future pre-clinical testing of potential treatments for this condition.
Collapse
Affiliation(s)
- Volkan Seyrantepe
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey.
| | - Secil Akyildiz Demir
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey
| | - Zehra Kevser Timur
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey
| | - Johanna Von Gerichten
- Lipid Pathobiochemistry Group (G131) German Cancer Research Center, INF 280, 69120 Heidelberg, Germany
| | - Christian Marsching
- Lipid Pathobiochemistry Group (G131) German Cancer Research Center, INF 280, 69120 Heidelberg, Germany; Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), Mannheim, Germany; Institute of Medical Technology (IMT) of Heidelberg University and Mannheim University of Applied Sciences, Paul-Wittsach-Str.10, 68163 Mannheim, Germany
| | - Esra Erdemli
- Departments of Histology and Embryology, Ankara University, Medical School, 06100, Sihhiye, Ankara, Turkey
| | - Emin Oztas
- Departments of Histology and Embryology, GATA Medical School, 06100 Ankara, Turkey
| | - Kohta Takahashi
- Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Japan
| | | | - Nurselin Ates
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey
| | - Buket Dönmez Demir
- Institutes of Neurological Science and Psychiatry, University of Hacettepe, 06100 Ankara, Turkey
| | - Turgay Dalkara
- Institutes of Neurological Science and Psychiatry, University of Hacettepe, 06100 Ankara, Turkey
| | - Katrin Erich
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), Mannheim, Germany; Institute of Medical Technology (IMT) of Heidelberg University and Mannheim University of Applied Sciences, Paul-Wittsach-Str.10, 68163 Mannheim, Germany
| | - Carsten Hopf
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), Mannheim, Germany; Institute of Medical Technology (IMT) of Heidelberg University and Mannheim University of Applied Sciences, Paul-Wittsach-Str.10, 68163 Mannheim, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry Group (G131) German Cancer Research Center, INF 280, 69120 Heidelberg, Germany
| | - Taeko Miyagi
- Miyagi Cancer Center Research Institute, Natori, Japan
| |
Collapse
|
40
|
Gray-Edwards HL, Randle AN, Maitland SA, Benatti HR, Hubbard SM, Canning PF, Vogel MB, Brunson BL, Hwang M, Ellis LE, Bradbury AM, Gentry AS, Taylor AR, Wooldridge AA, Wilhite DR, Winter RL, Whitlock BK, Johnson JA, Holland M, Salibi N, Beyers RJ, Sartin JL, Denney TS, Cox NR, Sena-Esteves M, Martin DR. Adeno-Associated Virus Gene Therapy in a Sheep Model of Tay-Sachs Disease. Hum Gene Ther 2017; 29:312-326. [PMID: 28922945 DOI: 10.1089/hum.2017.163] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Tay-Sachs disease (TSD) is a fatal neurodegenerative disorder caused by a deficiency of the enzyme hexosaminidase A (HexA). TSD also occurs in sheep, the only experimental model of TSD that has clinical signs of disease. The natural history of sheep TSD was characterized using serial neurological evaluations, 7 Tesla magnetic resonance imaging, echocardiograms, electrodiagnostics, and cerebrospinal fluid biomarkers. Intracranial gene therapy was also tested using AAVrh8 monocistronic vectors encoding the α-subunit of Hex (TSD α) or a mixture of two vectors encoding both the α and β subunits separately (TSD α + β) injected at high (1.3 × 1013 vector genomes) or low (4.2 × 1012 vector genomes) dose. Delay of symptom onset and/or reduction of acquired symptoms were noted in all adeno-associated virus-treated sheep. Postmortem evaluation showed superior HexA and vector genome distribution in the brain of TSD α + β sheep compared to TSD α sheep, but spinal cord distribution was low in all groups. Isozyme analysis showed superior HexA formation after treatment with both vectors (TSD α + β), and ganglioside clearance was most widespread in the TSD α + β high-dose sheep. Microglial activation and proliferation in TSD sheep-most prominent in the cerebrum-were attenuated after gene therapy. This report demonstrates therapeutic efficacy for TSD in the sheep brain, which is on the same order of magnitude as a child's brain.
Collapse
Affiliation(s)
- Heather L Gray-Edwards
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Ashley N Randle
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Stacy A Maitland
- 2 Department of Neurology and Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Hector R Benatti
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Spencer M Hubbard
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Peter F Canning
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Matthew B Vogel
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Brandon L Brunson
- 3 Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Misako Hwang
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Lauren E Ellis
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Allison M Bradbury
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama.,3 Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Atoska S Gentry
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Amanda R Taylor
- 4 Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Anne A Wooldridge
- 4 Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Dewey R Wilhite
- 3 Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Randolph L Winter
- 4 Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Brian K Whitlock
- 5 Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee , Knoxville, Tennessee
| | - Jacob A Johnson
- 4 Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Merilee Holland
- 4 Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Nouha Salibi
- 6 MR R&D Siemens Healthcare, Malvern, Pennsylvania
| | - Ronald J Beyers
- 7 Department of Electrical and Computer Engineering, Auburn University, Auburn, Alabama
| | - James L Sartin
- 3 Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Thomas S Denney
- 7 Department of Electrical and Computer Engineering, Auburn University, Auburn, Alabama
| | - Nancy R Cox
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama.,8 Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Miguel Sena-Esteves
- 2 Department of Neurology and Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Douglas R Martin
- 1 Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, Alabama.,3 Department of Anatomy, Physiology, and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| |
Collapse
|
41
|
Solomon M, Muro S. Lysosomal enzyme replacement therapies: Historical development, clinical outcomes, and future perspectives. Adv Drug Deliv Rev 2017; 118:109-134. [PMID: 28502768 PMCID: PMC5828774 DOI: 10.1016/j.addr.2017.05.004] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/26/2017] [Accepted: 05/08/2017] [Indexed: 01/06/2023]
Abstract
Lysosomes and lysosomal enzymes play a central role in numerous cellular processes, including cellular nutrition, recycling, signaling, defense, and cell death. Genetic deficiencies of lysosomal components, most commonly enzymes, are known as "lysosomal storage disorders" or "lysosomal diseases" (LDs) and lead to lysosomal dysfunction. LDs broadly affect peripheral organs and the central nervous system (CNS), debilitating patients and frequently causing fatality. Among other approaches, enzyme replacement therapy (ERT) has advanced to the clinic and represents a beneficial strategy for 8 out of the 50-60 known LDs. However, despite its value, current ERT suffers from several shortcomings, including various side effects, development of "resistance", and suboptimal delivery throughout the body, particularly to the CNS, lowering the therapeutic outcome and precluding the use of this strategy for a majority of LDs. This review offers an overview of the biomedical causes of LDs, their socio-medical relevance, treatment modalities and caveats, experimental alternatives, and future treatment perspectives.
Collapse
Affiliation(s)
- Melani Solomon
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University Maryland, College Park, MD 20742, USA.
| |
Collapse
|
42
|
Osmon KJL, Woodley E, Thompson P, Ong K, Karumuthil-Melethil S, Keimel JG, Mark BL, Mahuran D, Gray SJ, Walia JS. Systemic Gene Transfer of a Hexosaminidase Variant Using an scAAV9.47 Vector Corrects GM2 Gangliosidosis in Sandhoff Mice. Hum Gene Ther 2017; 27:497-508. [PMID: 27199088 DOI: 10.1089/hum.2016.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
GM2 gangliosidosis is a group of neurodegenerative diseases caused by β-hexosaminidase A (HexA) enzyme deficiency. There is currently no cure. HexA is composed of two similar, nonidentical subunits, α and β, which must interact with the GM2 activator protein (GM2AP), a substrate-specific cofactor, to hydrolyze GM2 ganglioside. Mutations in either subunit or the activator can result in the accumulation of GM2 ganglioside within neurons throughout the central nervous system. The resulting neuronal cell death induces the primary symptoms of the disease: motor impairment, seizures, and sensory impairments. This study assesses the long-term effects of gene transfer in a Sandhoff (β-subunit knockout) mouse model. The study utilized a modified human β-hexosaminidase α-subunit (μ-subunit) that contains critical sequences from the β-subunit that enables formation of a stable homodimer (HexM) and interaction with GM2AP to hydrolyze GM2 ganglioside. We investigated a self-complementary adeno-associated viral (scAAV) vector expressing HexM, through intravenous injections of the neonatal mice. We monitored one cohort for 8 weeks and another cohort long-term for survival benefit, behavioral, biochemical, and molecular analyses. Untreated Sandhoff disease (SD) control mice reached a humane endpoint at approximately 15 weeks, whereas treated mice had a median survival age of 40 weeks, an approximate 2.5-fold survival advantage. On behavioral tests, the treated mice outperformed their knockout age-matched controls and perform similarly to the heterozygous controls. Through the enzymatic and GM2 ganglioside analyses, we observed a significant decrease in the GM2 ganglioside level, even though the enzyme levels were not significantly increased. Molecular analyses revealed a global distribution of the vector between brain and spinal cord regions. In conclusion, the neonatal delivery of a novel viral vector expressing the human HexM enzyme is effective in ameliorating the SD mouse phenotype for long-term. Our data could have implications not only for treatment of SD but also for Tay-Sachs disease (α-subunit deficiency) and similar brain disorders.
Collapse
Affiliation(s)
- Karlaina J L Osmon
- 1 Centre for Neuroscience Studies, Queen's University , Kingston, Ontario, Canada
| | - Evan Woodley
- 2 Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada
| | - Patrick Thompson
- 3 Medical Genetics/Departments of Pediatrics, Queen's University , Kingston, Ontario, Canada
| | - Katalina Ong
- 3 Medical Genetics/Departments of Pediatrics, Queen's University , Kingston, Ontario, Canada
| | | | - John G Keimel
- 5 New Hope Research Foundation , North Oaks, Minnesota
| | - Brian L Mark
- 6 Department of Microbiology, University of Manitoba , Winnipeg, Manitoba, Canada
| | - Don Mahuran
- 7 Genetics and Genome Biology, SickKids, Toronto, Ontario, Canada .,8 Department of Laboratory Medicine and Pathology, University of Toronto , Toronto, Ontario, Canada
| | - Steven J Gray
- 4 Gene Therapy Center, University of North Carolina , Chapel Hill, North Carolina.,9 Department of Ophthalmology, University of North Carolina , Chapel Hill, North Carolina
| | - Jagdeep S Walia
- 1 Centre for Neuroscience Studies, Queen's University , Kingston, Ontario, Canada .,2 Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario, Canada .,3 Medical Genetics/Departments of Pediatrics, Queen's University , Kingston, Ontario, Canada
| |
Collapse
|
43
|
Karumuthil-Melethil S, Nagabhushan Kalburgi S, Thompson P, Tropak M, Kaytor MD, Keimel JG, Mark BL, Mahuran D, Walia JS, Gray SJ. Novel Vector Design and Hexosaminidase Variant Enabling Self-Complementary Adeno-Associated Virus for the Treatment of Tay-Sachs Disease. Hum Gene Ther 2017; 27:509-21. [PMID: 27197548 DOI: 10.1089/hum.2016.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
GM2 gangliosidosis is a family of three genetic neurodegenerative disorders caused by the accumulation of GM2 ganglioside (GM2) in neuronal tissue. Two of these are due to the deficiency of the heterodimeric (α-β), "A" isoenzyme of lysosomal β-hexosaminidase (HexA). Mutations in the α-subunit (encoded by HEXA) lead to Tay-Sachs disease (TSD), whereas mutations in the β-subunit (encoded by HEXB) lead to Sandhoff disease (SD). The third form results from a deficiency of the GM2 activator protein (GM2AP), a substrate-specific cofactor for HexA. In their infantile, acute forms, these diseases rapidly progress with mental and psychomotor deterioration resulting in death by approximately 4 years of age. After gene transfer that overexpresses one of the deficient subunits, the amount of HexA heterodimer formed would empirically be limited by the availability of the other endogenous Hex subunit. The present study used a new variant of the human HexA α-subunit, μ, incorporating critical sequences from the β-subunit that produce a stable homodimer (HexM) and promote functional interactions with the GM2AP- GM2 complex. We report the design of a compact adeno-associated viral (AAV) genome using a synthetic promoter-intron combination to allow self-complementary (sc) packaging of the HEXM gene. Also, a previously published capsid mutant, AAV9.47, was used to deliver the gene to brain and spinal cord while having restricted biodistribution to the liver. The novel capsid and cassette design combination was characterized in vivo in TSD mice for its ability to efficiently transduce cells in the central nervous system when delivered intravenously in both adult and neonatal mice. This study demonstrates that the modified HexM is capable of degrading long-standing GM2 storage in mice, and it further demonstrates the potential of this novel scAAV vector design to facilitate widespread distribution of the HEXM gene or potentially other similar-sized genes to the nervous system.
Collapse
Affiliation(s)
| | | | - Patrick Thompson
- 2 Medical Genetics/Departments of Pediatrics, Queen's University , Kingston, Ontario, Canada
| | - Michael Tropak
- 3 Genetics and Genome Biology, SickKids, Toronto, Ontario, Canada
| | | | - John G Keimel
- 4 New Hope Research Foundation , North Oaks, Minnesota
| | - Brian L Mark
- 5 Department of Microbiology, University of Manitoba , Winnipeg, Manitoba, Canada
| | - Don Mahuran
- 3 Genetics and Genome Biology, SickKids, Toronto, Ontario, Canada .,6 Department of Laboratory Medicine and Pathology, University of Toronto, Toronto, Ontario, Canada
| | - Jagdeep S Walia
- 2 Medical Genetics/Departments of Pediatrics, Queen's University , Kingston, Ontario, Canada
| | - Steven J Gray
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,7 Department of Ophthalmology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| |
Collapse
|
44
|
Hooper AWM, Alamilla JF, Venier RE, Gillespie DC, Igdoura SA. Neuronal pentraxin 1 depletion delays neurodegeneration and extends life in Sandhoff disease mice. Hum Mol Genet 2017; 26:661-673. [PMID: 28007910 DOI: 10.1093/hmg/ddw422] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 12/09/2016] [Indexed: 01/15/2023] Open
Abstract
GM2 gangliosidoses are a group of lysosomal storage disorders which include Sandhoff disease and Tay-Sachs disease. Dysregulation of glutamate receptors has been recently postulated in the pathology of Sandhoff disease. Glutamate receptor association with neuronal pentraxins 1 and 2, and the neuronal pentraxin receptor facilitates receptor potentiation and synaptic shaping. In this study, we have observed an upregulation of a novel form of neuronal pentraxin 1 (NP1-38) in the brains of a mouse model of Sandhoff disease and Tay-Sachs disease. In order to determine the impact of NP1 on the pathophysiology of Sandhoff disease mouse models, we have generated an Np1-/-Hexb-/- double knockout mouse, and observed extended lifespan, improved righting reflex and enhanced body condition relative to Hexb-/- mice, with no effect on gliosis or apoptotic markers in the CNS. Sandhoff mouse brain slices reveals a reduction in AMPA receptor-mediated currents, and increased variability in total glutamate currents in the CA1 region of the hippocampus; Np1-/-Hexb-/- mice show a correction of this phenotype, suggesting NP1-38 may be interfering with glutamate receptor function. Indeed, some of the psychiatric aspects of Sandhoff and Tay-Sachs disease (particularly late onset) may be attributed to a dysfunctional hippocampal glutamatergic system. Our work highlights a potential role for synaptic proteins, such as NP1 and glutamate receptors in lysosomal storage diseases.
Collapse
Affiliation(s)
| | | | | | | | - Suleiman A Igdoura
- Department of Biology.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
45
|
Gray-Edwards HL, Regier DS, Shirley JL, Randle AN, Salibi N, Thomas SE, Latour YL, Johnston J, Golas G, Maguire AS, Taylor AR, Sorjonen DC, McCurdy VJ, Christopherson PW, Bradbury AM, Beyers RJ, Johnson AK, Brunson BL, Cox NR, Baker HJ, Denney TS, Sena-Esteves M, Tifft CJ, Martin DR. Novel Biomarkers of Human GM1 Gangliosidosis Reflect the Clinical Efficacy of Gene Therapy in a Feline Model. Mol Ther 2017; 25:892-903. [PMID: 28236574 PMCID: PMC5383552 DOI: 10.1016/j.ymthe.2017.01.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 01/06/2017] [Accepted: 01/14/2017] [Indexed: 11/26/2022] Open
Abstract
GM1 gangliosidosis is a fatal neurodegenerative disease that affects individuals of all ages. Favorable outcomes using adeno-associated viral (AAV) gene therapy in GM1 mice and cats have prompted consideration of human clinical trials, yet there remains a paucity of objective biomarkers to track disease status. We developed a panel of biomarkers using blood, urine, cerebrospinal fluid (CSF), electrodiagnostics, 7 T MRI, and magnetic resonance spectroscopy in GM1 cats-either untreated or AAV treated for more than 5 years-and compared them to markers in human GM1 patients where possible. Significant alterations were noted in CSF and blood of GM1 humans and cats, with partial or full normalization after gene therapy in cats. Gene therapy improved the rhythmic slowing of electroencephalograms (EEGs) in GM1 cats, a phenomenon present also in GM1 patients, but nonetheless the epileptiform activity persisted. After gene therapy, MR-based analyses revealed remarkable preservation of brain architecture and correction of brain metabolites associated with microgliosis, neuroaxonal loss, and demyelination. Therapeutic benefit of AAV gene therapy in GM1 cats, many of which maintain near-normal function >5 years post-treatment, supports the strong consideration of human clinical trials, for which the biomarkers described herein will be essential for outcome assessment.
Collapse
Affiliation(s)
- Heather L Gray-Edwards
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Debra S Regier
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jamie L Shirley
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Ashley N Randle
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Nouha Salibi
- MR R&D, Siemens Healthcare, Malvern, PA 19355, USA
| | - Sarah E Thomas
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yvonne L Latour
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jean Johnston
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gretchen Golas
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Annie S Maguire
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Amanda R Taylor
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Donald C Sorjonen
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Victoria J McCurdy
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Peter W Christopherson
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Allison M Bradbury
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Ronald J Beyers
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL 36849, USA
| | - Aime K Johnson
- Department of Clinical Sciences, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Brandon L Brunson
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Nancy R Cox
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Henry J Baker
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA
| | - Thomas S Denney
- Department of Electrical and Computer Engineering, Auburn University, Auburn, AL 36849, USA
| | - Miguel Sena-Esteves
- Department of Neurology and Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Cynthia J Tifft
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA.
| |
Collapse
|
46
|
FcRγ-dependent immune activation initiates astrogliosis during the asymptomatic phase of Sandhoff disease model mice. Sci Rep 2017; 7:40518. [PMID: 28084424 PMCID: PMC5234013 DOI: 10.1038/srep40518] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 12/07/2016] [Indexed: 12/25/2022] Open
Abstract
Sandhoff disease (SD) is caused by the loss of β-hexosaminidase (Hex) enzymatic activity in lysosomes resulting from Hexb mutations. In SD patients, the Hex substrate GM2 ganglioside accumulates abnormally in neuronal cells, resulting in neuronal loss, microglial activation, and astrogliosis. Hexb−/− mice, which manifest a phenotype similar to SD, serve as animal models for examining the pathophysiology of SD. Hexb−/− mice reach ~8 weeks without obvious neurological defects; however, trembling begins at 12 weeks and is accompanied by startle reactions and increased limb tone. These symptoms gradually become severe by 16–18 weeks. Immune reactions caused by autoantibodies have been recently associated with the pathology of SD. The inhibition of immune activation may represent a novel therapeutic target for SD. Herein, SD mice (Hexb−/−) were crossed to mice lacking an activating immune receptor (FcRγ−/−) to elucidate the potential relationship between immune responses activated through SD autoantibodies and astrogliosis. Microglial activation and astrogliosis were observed in cortices of Hexb−/− mice during the asymptomatic phase, and were inhibited in Hexb−/−FcRγ−/− mice. Moreover, early astrogliosis and impaired motor coordination in Hexb−/− mice could be ameliorated by immunosuppressants, such as FTY720. Our findings demonstrate the importance of early treatment and the therapeutic effectiveness of immunosuppression in SD.
Collapse
|
47
|
Hooper AWM, Igdoura SA. Bi-phasic gliosis drives neuropathology in a Sandhoff disease mouse model. J Neuroimmunol 2016; 299:19-27. [PMID: 27725117 DOI: 10.1016/j.jneuroim.2016.08.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 08/02/2016] [Accepted: 08/07/2016] [Indexed: 11/20/2022]
Abstract
Microgliosis and astrogliosis are known to be exacerbating factors in the progression of the lysosomal storage disorder Sandhoff disease. We have also found evidence for excitotoxicity via glutamate receptors in Sandhoff disease. To view the interaction of these cascades, we measured cerebellar expression of markers for gliosis, apoptosis, and excitatory synapses over the disease course in a Sandhoff disease mouse model. We observe a 2-stage model, with initial activation of microgliosis as early as 60days of age, followed by a later onset of astrogliosis, caspase-mediated apoptosis, and reduction in GluR1 at approximately 100days of age. These results implicate immune cells as first responders in Sandhoff disease.
Collapse
Affiliation(s)
| | - Suleiman A Igdoura
- Department of Biology, McMaster University, Hamilton, Ont. L8S 4K1, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ont. L8S 4L8, Canada.
| |
Collapse
|
48
|
Suzuki K, Yamaguchi A, Yamanaka S, Kanzaki S, Kawashima M, Togo T, Katsuse O, Koumitsu N, Aoki N, Iseki E, Kosaka K, Yamaguchi K, Hashimoto M, Aoki I, Hirayasu Y. Accumulated α-synuclein affects the progression of GM2 gangliosidoses. Exp Neurol 2016; 284:38-49. [PMID: 27453479 DOI: 10.1016/j.expneurol.2016.07.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 07/16/2016] [Accepted: 07/20/2016] [Indexed: 02/07/2023]
Abstract
The accumulation of α-synuclein (ASyn) has been observed in several lysosomal storage diseases (LSDs) but it remains unclear if ASyn accumulation contributes to LSD pathology. ASyn also accumulates in the neurons of Sandhoff disease (SD) patients and SD model mice (Hexb-/- ASyn+/+ mice). SD is a lysosomal storage disorder caused by the absence of a functional β-subunit on the β-hexosaminidase A and B enzymes, which leads to the accumulation of ganglioside in the central nervous system. Here, we explored the role of accumulated ASyn in the progression of Hexb-/- mice by creating a Hexb-/- ASyn-/- double-knockout mice. Our results show that Hexb-/- ASyn-/- mice demonstrated active microglia levels and less dopaminergic neuron loss, without altering the neuronal storage of ganglioside. The autophagy and ubiquitin proteasome pathways are defective in the neurons of Hexb-/- ASyn+/+ mice. In ultrastructural physiological studies, the mitochondria structures look degenerated and dysfunctional. As a result, expression of manganese superoxide dismutase 2 are reduced, and reactive oxygen species-mediated oxidative damage in the neurons of Hexb-/- ASyn+/+ mice. Interestingly, these dysfunctions improved in Hexb-/- ASyn-/- mice. But any clinical improvement were hardly observed in Hexb-/- ASyn-/- mice. Taken together, these findings suggest that ASyn accumulation plays an important role in the pathogenesis of neuropathy in SD and other LSDs, and is therefore a target for novel therapies.
Collapse
Affiliation(s)
- Kyoko Suzuki
- Department of Psychiatry, Yokohama City University School of Medicine, Japan
| | - Akira Yamaguchi
- Department of Pathology, Yokohama City University School of Medicine, Japan.
| | - Shoji Yamanaka
- Department of Pathology, Yokohama City University School of Medicine, Japan
| | - Seiichi Kanzaki
- Department of Pathology, Yokohama City University School of Medicine, Japan
| | - Masato Kawashima
- Department of Pathology, Yokohama City University School of Medicine, Japan
| | - Takashi Togo
- Department of Psychiatry, Yokohama City University School of Medicine, Japan
| | - Omi Katsuse
- Department of Psychiatry, Yokohama City University School of Medicine, Japan
| | - Noriko Koumitsu
- Department of Dermatology, Yokohama City University School of Medicine, Japan
| | - Naoya Aoki
- Department of Psychiatry, Yokohama City University School of Medicine, Japan
| | - Eizo Iseki
- Department of Psychiatry, Juntendo University School of Medicine, Japan
| | - Kenji Kosaka
- Department of Psychiatry, Yokohama City University School of Medicine, Japan
| | - Kayoko Yamaguchi
- Department of Pathology, Yokohama City University School of Medicine, Japan
| | | | - Ichiro Aoki
- Department of Pathology, Yokohama City University School of Medicine, Japan
| | - Yoshio Hirayasu
- Department of Psychiatry, Yokohama City University School of Medicine, Japan
| |
Collapse
|
49
|
Lawson CA, Martin DR. Animal models of GM2 gangliosidosis: utility and limitations. APPLICATION OF CLINICAL GENETICS 2016; 9:111-20. [PMID: 27499644 PMCID: PMC4959762 DOI: 10.2147/tacg.s85354] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
GM2 gangliosidosis, a subset of lysosomal storage disorders, is caused by a deficiency of the glycohydrolase, β-N-acetylhexosaminidase, and includes the closely related Tay–Sachs and Sandhoff diseases. The enzyme deficiency prevents the normal, stepwise degradation of ganglioside, which accumulates unchecked within the cellular lysosome, particularly in neurons. As a result, individuals with GM2 gangliosidosis experience progressive neurological diseases including motor deficits, progressive weakness and hypotonia, decreased responsiveness, vision deterioration, and seizures. Mice and cats are well-established animal models for Sandhoff disease, whereas Jacob sheep are the only known laboratory animal model of Tay–Sachs disease to exhibit clinical symptoms. Since the human diseases are relatively rare, animal models are indispensable tools for further study of pathogenesis and for development of potential treatments. Though no effective treatments for gangliosidoses currently exist, animal models have been used to test promising experimental therapies. Herein, the utility and limitations of gangliosidosis animal models and how they have contributed to the development of potential new treatments are described.
Collapse
Affiliation(s)
| | - Douglas R Martin
- Scott-Ritchey Research Center; Department of Anatomy, Physiology and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, USA
| |
Collapse
|
50
|
Alkelai A, Olender T, Haffner-Krausz R, Tsoory MM, Boyko V, Tatarskyy P, Gross-Isseroff R, Milgrom R, Shushan S, Blau I, Cohn E, Beeri R, Levy-Lahad E, Pras E, Lancet D. A role for TENM1 mutations in congenital general anosmia. Clin Genet 2016; 90:211-9. [PMID: 27040985 DOI: 10.1111/cge.12782] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 03/26/2016] [Accepted: 03/27/2016] [Indexed: 02/01/2023]
Abstract
Congenital general anosmia (CGA) is a neurological disorder entailing a complete innate inability to sense odors. While the mechanisms underlying vertebrate olfaction have been studied in detail, there are still gaps in our understanding of the molecular genetic basis of innate olfactory disorders. Applying whole-exome sequencing to a family multiply affected with CGA, we identified three members with a rare X-linked missense mutation in the TENM1 (teneurin 1) gene (ENST00000422452:c.C4829T). In Drosophila melanogaster, TENM1 functions in synaptic-partner-matching between axons of olfactory sensory neurons and target projection neurons and is involved in synapse organization in the olfactory system. We used CRISPR-Cas9 system to generate a Tenm1 disrupted mouse model. Tenm1(-/-) and point-mutated Tenm1(A) (/A) adult mice were shown to have an altered ability to locate a buried food pellet. Tenm1(A) (/A) mice also displayed an altered ability to sense aversive odors. Results of our study, that describes a new Tenm1 mouse, agree with the hypothesis that TENM1 has a role in olfaction. However, additional studies should be done in larger CGA cohorts, to provide statistical evidence that loss-of-function mutations in TENM1 can solely cause the disease in our and other CGA cases.
Collapse
Affiliation(s)
- A Alkelai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - T Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - R Haffner-Krausz
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - M M Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - V Boyko
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - P Tatarskyy
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - R Gross-Isseroff
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - R Milgrom
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - S Shushan
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel.,Department of Otolaryngology-Head and Neck Surgery, Edith Wolfson Medical Center, Holon, Israel
| | - I Blau
- Department of Otolaryngology, Meir Medical Center, Kfar Saba, Israel
| | - E Cohn
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - R Beeri
- Medical Genetics Institute, Shaare Zedek Medical Center, Jerusalem, Israel
| | - E Levy-Lahad
- Department of Otolaryngology, Meir Medical Center, Kfar Saba, Israel
| | - E Pras
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Ramat Gan, Israel.,The Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - D Lancet
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|