1
|
Lopez ME, Wendt D, Lawrence R, Gong K, Ong H, Yip B, Chen J, Mangini L, Handyside B, Giaramita A, Lamichhane A, Lo M, Agrawal V, Van Vleet J, Abolhesn A, Felix JB, Villalpando I, Bhat V, De Angelis R, Ru Y, Khan A, Fong S, Christianson T, Bullens S, Crawford BE, Bunting S, Aoyagi-Scharber M. Intracerebroventricular administration of a modified hexosaminidase ameliorates late-stage neurodegeneration in a GM2 mouse model. PLoS One 2025; 20:e0315005. [PMID: 39752451 PMCID: PMC11698352 DOI: 10.1371/journal.pone.0315005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 11/19/2024] [Indexed: 01/06/2025] Open
Abstract
The GM2 gangliosidoses, Tay-Sachs disease and Sandhoff disease, are devastating neurodegenerative disorders caused by β-hexosaminidase A (HexA) deficiency. In the Sandhoff disease mouse model, rescue potential was severely reduced when HexA was introduced after disease onset. Here, we assess the effect of recombinant HexA and HexD3, a newly engineered mimetic of HexA optimized for the treatment of Tay-Sachs disease and Sandhoff disease. Enzyme replacement therapy was administered by repeat intracerebroventricular injections in Sandhoff disease model mice with dosing beginning before and after signs of neurodegeneration. As previously observed, HexA effectively increased the lifespan of Sandhoff disease mice by 3.5-fold only when treatment was started before onset of neurodegeneration. In contrast, HexD3 halted motor decline and ameliorated late-stage disease severity even when dosing began late, after neurodegeneration onset. Additionally, HexD3 had advantages over HexA in enzyme stability, distribution potential, and homodimer activity. Overall, our data indicate that advanced therapeutics may widen the treatment window for neurodegenerative disorders.
Collapse
Affiliation(s)
- Manuel E. Lopez
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Daniel Wendt
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Roger Lawrence
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Kerui Gong
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Hoonsan Ong
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Bryan Yip
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Joseph Chen
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Linley Mangini
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Britta Handyside
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | | | - Melanie Lo
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Vishal Agrawal
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Jeremy Van Vleet
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Amanda Abolhesn
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Jessica B. Felix
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Vikas Bhat
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Yuanbin Ru
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Ayesha Khan
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | - Sylvia Fong
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Sherry Bullens
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | | - Stuart Bunting
- BioMarin Pharmaceutical Inc., Novato, CA, United States of America
| | | |
Collapse
|
2
|
Bensalem J, Hein LK, Hassiotis S, Trim PJ, Proud CG, Heilbronn LK, Sargeant TJ. Modifying Dietary Protein Impacts mTOR Signaling and Brain Deposition of Amyloid β in a Knock-In Mouse Model of Alzheimer Disease. J Nutr 2023; 153:1407-1419. [PMID: 36870538 DOI: 10.1016/j.tjnut.2023.02.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Alzheimer disease (AD) is a neurodegenerative condition defined by the build-up of amyloid plaques in the brain and intraneuronal tangles of the protein tau. Autophagy is a cellular cleaning process involved in the degradation of proteins, including proteins directly responsible for amyloid plaques, but its activity is compromised in AD. The mechanistic target of rapamycin complex (mTORC) 1 inhibits autophagy when activated by amino acids. OBJECTIVES We hypothesized that reducing amino acid intake by decreasing dietary protein could promote autophagy, which in turn could prevent amyloid plaque deposition in AD mice. METHODS Homozygote (2-mo-old) and heterozygote (4-mo-old) amyloid precursor protein NL-G-F mice, a model of brain amyloid deposition, were used in this study to test this hypothesis. Male and female mice were fed with isocaloric low-protein, control, or high-protein diets for 4 mo and killed for analysis. Locomotor performance was measured using the inverted screen test, and body composition was measured using EchoMRI. Samples were analyzed using western blotting, enzyme-linked immunosorbent assay, mass spectrometry, and immunohistochemical staining. RESULTS mTORC1 activity in the cerebral cortex was inversely covaried with protein consumption in both homozygote and heterozygote mice. Low-protein diet improved metabolic parameters and restored locomotor performance only in male homozygous mice. Dietary protein adjustment did not affect amyloid deposition in homozygous mice. However, in the heterozygous amyloid precursor protein NL-G-F mice, amyloid plaque was lower in male mice consuming the low protein compared with that in mice fed with the control diet. CONCLUSIONS This study showed that reducing protein intake reduces mTORC1 activity and may prevent amyloid accumulation, at least in male mice. Moreover, dietary protein is a tool that can be used to change mTORC1 activity and amyloid deposition in the mouse brain, and the murine brain's response to dietary protein is sex specific.
Collapse
Affiliation(s)
- Julien Bensalem
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Leanne K Hein
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Sofia Hassiotis
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Paul J Trim
- Proteomics, Metabolomics and MS-Imaging Core Facility, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Christopher G Proud
- Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Leonie K Heilbronn
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; Nutrition, Metabolism & Gut Health, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Timothy J Sargeant
- Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
3
|
The GM2 gangliosidoses: Unlocking the mysteries of pathogenesis and treatment. Neurosci Lett 2021; 764:136195. [PMID: 34450229 PMCID: PMC8572160 DOI: 10.1016/j.neulet.2021.136195] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/28/2022]
|
4
|
Maguire AS, Martin DR. White Matter Pathology as a Barrier to Gangliosidosis Gene Therapy. Front Cell Neurosci 2021; 15:682106. [PMID: 34456684 PMCID: PMC8397537 DOI: 10.3389/fncel.2021.682106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/23/2021] [Indexed: 11/25/2022] Open
Abstract
The gangliosidoses are a family of neurodegenerative lysosomal storage diseases that have recently seen promising advances in gene therapy. White matter deficits are well established components of gangliosidosis pathology that are now receiving more attention because they are partially refractory to correction by gene therapy. After a brief synopsis of normal myelinogenesis, this review outlines current viewpoints on the origin of white matter deficits in the gangliosidoses and potential obstacles to treating them effectively by gene therapy. Dysmyelinogenesis (failure of myelin sheaths to form properly) is proposed as the predominant contributor to white matter pathology, but precise mechanistic details are not well understood. The involvement of neuronal storage deficits may extend beyond secondary demyelination (destruction of myelin due to axonal loss) and contribute to dysmyelinogenesis. Preclinical studies in animal models of the gangliosidoses have substantially improved lifespan and quality of life, leading to the initiation of several clinical trials. However, improvement of white matter pathology has lagged behind other metrics and few evidence-based explanations have been proposed to date. Research groups in the field are encouraged to include myelin-specific investigations in future gene therapy work to address this gap in knowledge.
Collapse
Affiliation(s)
- Anne S. Maguire
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL, United States
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| | - Douglas R. Martin
- Scott-Ritchey Research Center, Auburn University College of Veterinary Medicine, Auburn, AL, United States
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, United States
| |
Collapse
|
5
|
Allende ML, Zhu H, Kono M, Hoachlander-Hobby LE, Huso VL, Proia RL. Genetic defects in the sphingolipid degradation pathway and their effects on microglia in neurodegenerative disease. Cell Signal 2021; 78:109879. [PMID: 33296739 PMCID: PMC7775721 DOI: 10.1016/j.cellsig.2020.109879] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022]
Abstract
Sphingolipids, which function as plasma membrane lipids and signaling molecules, are highly enriched in neuronal and myelin membranes in the nervous system. They are degraded in lysosomes by a defined sequence of enzymatic steps. In the related group of disorders, the sphingolipidoses, mutations in the genes that encode the individual degradative enzymes cause lysosomal accumulation of sphingolipids and often result in severe neurodegenerative disease. Here we review the information indicating that microglia, which actively clear sphingolipid-rich membranes in the brain during development and homeostasis, are directly affected by these mutations and promote neurodegeneration in the sphingolipidoses. We also identify parallels between the sphingolipidoses and more common forms of neurodegeneration, which both exhibit evidence of defective sphingolipid clearance in the nervous system.
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, 9000 Rockville Pike, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hongling Zhu
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, 9000 Rockville Pike, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mari Kono
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, 9000 Rockville Pike, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lila E Hoachlander-Hobby
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, 9000 Rockville Pike, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vienna L Huso
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, 9000 Rockville Pike, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard L Proia
- Genetics of Development and Disease Section, Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases, 9000 Rockville Pike, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Pewzner-Jung Y, Joseph T, Blumenreich S, Vardi A, Ferreira NS, Cho SM, Eilam R, Tsoory M, Biton IE, Brumfeld V, Haffner-Krausz R, Brenner O, Sharabi N, Addadi Y, Salame TM, Rotkopf R, Wigoda N, Yayon N, Merrill AH, Schiffmann R, Futerman AH. Brain pathology and cerebellar purkinje cell loss in a mouse model of chronic neuronopathic Gaucher disease. Prog Neurobiol 2020; 197:101939. [PMID: 33152398 DOI: 10.1016/j.pneurobio.2020.101939] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/03/2020] [Accepted: 10/25/2020] [Indexed: 12/12/2022]
Abstract
Gaucher disease (GD) is currently the focus of considerable attention due primarily to the association between the gene that causes GD (GBA) and Parkinson's disease. Mouse models exist for the systemic (type 1) and for the acute neuronopathic forms (type 2) of GD. Here we report the generation of a mouse that phenotypically models chronic neuronopathic type 3 GD. Gba-/-;Gbatg mice, which contain a Gba transgene regulated by doxycycline, accumulate moderate levels of the offending substrate in GD, glucosylceramide, and live for up to 10 months, i.e. significantly longer than mice which model type 2 GD. Gba-/-;Gbatg mice display behavioral abnormalities at ∼4 months, which deteriorate with age, along with significant neuropathology including loss of Purkinje neurons. Gene expression is altered in the brain and in isolated microglia, although the changes in gene expression are less extensive than in mice modeling type 2 disease. Finally, bone deformities are consistent with the Gba-/-;Gbatg mice being a genuine type 3 GD model. Together, the Gba-/-;Gbatg mice share pathological pathways with acute neuronopathic GD mice but also display differences that might help understand the distinct disease course and progression of type 2 and 3 patients.
Collapse
Affiliation(s)
- Yael Pewzner-Jung
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.
| | - Tammar Joseph
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shani Blumenreich
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Ayelet Vardi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | - Soo Min Cho
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Raya Eilam
- Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Tsoory
- Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Inbal E Biton
- Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Vlad Brumfeld
- Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | | | - Ori Brenner
- Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Nir Sharabi
- Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tomer-Meir Salame
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Rotkopf
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Wigoda
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Nadav Yayon
- Department of Biological Chemistry, The Life Sciences Institute, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alfred H Merrill
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
7
|
Leal AF, Benincore-Flórez E, Solano-Galarza D, Garzón Jaramillo RG, Echeverri-Peña OY, Suarez DA, Alméciga-Díaz CJ, Espejo-Mojica AJ. GM2 Gangliosidoses: Clinical Features, Pathophysiological Aspects, and Current Therapies. Int J Mol Sci 2020; 21:ijms21176213. [PMID: 32867370 PMCID: PMC7503724 DOI: 10.3390/ijms21176213] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022] Open
Abstract
GM2 gangliosidoses are a group of pathologies characterized by GM2 ganglioside accumulation into the lysosome due to mutations on the genes encoding for the β-hexosaminidases subunits or the GM2 activator protein. Three GM2 gangliosidoses have been described: Tay-Sachs disease, Sandhoff disease, and the AB variant. Central nervous system dysfunction is the main characteristic of GM2 gangliosidoses patients that include neurodevelopment alterations, neuroinflammation, and neuronal apoptosis. Currently, there is not approved therapy for GM2 gangliosidoses, but different therapeutic strategies have been studied including hematopoietic stem cell transplantation, enzyme replacement therapy, substrate reduction therapy, pharmacological chaperones, and gene therapy. The blood-brain barrier represents a challenge for the development of therapeutic agents for these disorders. In this sense, alternative routes of administration (e.g., intrathecal or intracerebroventricular) have been evaluated, as well as the design of fusion peptides that allow the protein transport from the brain capillaries to the central nervous system. In this review, we outline the current knowledge about clinical and physiopathological findings of GM2 gangliosidoses, as well as the ongoing proposals to overcome some limitations of the traditional alternatives by using novel strategies such as molecular Trojan horses or advanced tools of genome editing.
Collapse
Affiliation(s)
- Andrés Felipe Leal
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Eliana Benincore-Flórez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Daniela Solano-Galarza
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Rafael Guillermo Garzón Jaramillo
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Olga Yaneth Echeverri-Peña
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Diego A. Suarez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
- Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 110231, Colombia
| | - Carlos Javier Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
- Correspondence: (C.J.A.-D.); (A.J.E.-M.); Tel.: +57-1-3208320 (ext. 4140) (C.J.A.-D.); +57-1-3208320 (ext. 4099) (A.J.E.-M.)
| | - Angela Johana Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
- Correspondence: (C.J.A.-D.); (A.J.E.-M.); Tel.: +57-1-3208320 (ext. 4140) (C.J.A.-D.); +57-1-3208320 (ext. 4099) (A.J.E.-M.)
| |
Collapse
|
8
|
Kuil LE, López Martí A, Carreras Mascaro A, van den Bosch JC, van den Berg P, van der Linde HC, Schoonderwoerd K, Ruijter GJG, van Ham TJ. Hexb enzyme deficiency leads to lysosomal abnormalities in radial glia and microglia in zebrafish brain development. Glia 2019; 67:1705-1718. [PMID: 31140649 PMCID: PMC6772114 DOI: 10.1002/glia.23641] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/12/2019] [Accepted: 05/10/2019] [Indexed: 12/11/2022]
Abstract
Sphingolipidoses are severe, mostly infantile lysosomal storage disorders (LSDs) caused by defective glycosphingolipid degradation. Two of these sphingolipidoses, Tay Sachs and Sandhoff diseases, are caused by β-Hexosaminidase (HEXB) enzyme deficiency, resulting in ganglioside (GM2) accumulation and neuronal loss. The precise sequence of cellular events preceding, and leading to, neuropathology remains unclear, but likely involves inflammation and lysosomal accumulation of GM2 in multiple cell types. We aimed to determine the consequences of Hexb activity loss for different brain cell types using zebrafish. Hexb deficient zebrafish (hexb-/- ) showed lysosomal abnormalities already early in development both in radial glia, which are the neuronal and glial progenitors, and in microglia. Additionally, at 5 days postfertilization, hexb-/- zebrafish showed reduced locomotor activity. Although specific oligosaccharides accumulate in the adult brain, hexb-/- ) zebrafish are viable and apparently resistant to Hexb deficiency. In all, we identified cellular consequences of loss of Hexb enzyme activity during embryonic brain development, showing early effects on glia, which possibly underlie the behavioral aberrations. Hereby, we identified clues into the contribution of non-neuronal lysosomal abnormalities in LSDs affecting the brain and provide a tool to further study what underlies the relative resistance to Hexb deficiency in vivo.
Collapse
Affiliation(s)
- Laura E. Kuil
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Anna López Martí
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Ana Carreras Mascaro
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Jeroen C. van den Bosch
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Paul van den Berg
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Herma C. van der Linde
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Kees Schoonderwoerd
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - George J. G. Ruijter
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| |
Collapse
|
9
|
Allende ML, Cook EK, Larman BC, Nugent A, Brady JM, Golebiowski D, Sena-Esteves M, Tifft CJ, Proia RL. Cerebral organoids derived from Sandhoff disease-induced pluripotent stem cells exhibit impaired neurodifferentiation. J Lipid Res 2018; 59:550-563. [PMID: 29358305 PMCID: PMC5832932 DOI: 10.1194/jlr.m081323] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/29/2017] [Indexed: 12/21/2022] Open
Abstract
Sandhoff disease, one of the GM2 gangliosidoses, is a lysosomal storage disorder characterized by the absence of β-hexosaminidase A and B activity and the concomitant lysosomal accumulation of its substrate, GM2 ganglioside. It features catastrophic neurodegeneration and death in early childhood. How the lysosomal accumulation of ganglioside might affect the early development of the nervous system is not understood. Recently, cerebral organoids derived from induced pluripotent stem (iPS) cells have illuminated early developmental events altered by disease processes. To develop an early neurodevelopmental model of Sandhoff disease, we first generated iPS cells from the fibroblasts of an infantile Sandhoff disease patient, then corrected one of the mutant HEXB alleles in those iPS cells using CRISPR/Cas9 genome-editing technology, thereby creating isogenic controls. Next, we used the parental Sandhoff disease iPS cells and isogenic HEXB-corrected iPS cell clones to generate cerebral organoids that modeled the first trimester of neurodevelopment. The Sandhoff disease organoids, but not the HEXB-corrected organoids, accumulated GM2 ganglioside and exhibited increased size and cellular proliferation compared with the HEXB-corrected organoids. Whole-transcriptome analysis demonstrated that development was impaired in the Sandhoff disease organoids, suggesting that alterations in neuronal differentiation may occur during early development in the GM2 gangliosidoses.
Collapse
Affiliation(s)
- Maria L Allende
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Emily K Cook
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Bridget C Larman
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Adrienne Nugent
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jacqueline M Brady
- National Institutes of Health Undiagnosed Diseases Program, National Institutes of Health Office of Rare Diseases Research and National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Diane Golebiowski
- Department of Neurology and Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605
| | - Miguel Sena-Esteves
- Department of Neurology and Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605
| | - Cynthia J Tifft
- National Institutes of Health Undiagnosed Diseases Program, National Institutes of Health Office of Rare Diseases Research and National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
10
|
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: 0.4] [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
|
11
|
Zigdon H, Meshcheriakova A, Futerman AH. From sheep to mice to cells: Tools for the study of the sphingolipidoses. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:1189-99. [DOI: 10.1016/j.bbalip.2014.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 12/12/2022]
|
12
|
Cachón-González MB, Wang SZ, Ziegler R, Cheng SH, Cox TM. Reversibility of neuropathology in Tay-Sachs-related diseases. Hum Mol Genet 2014; 23:730-48. [PMID: 24057669 PMCID: PMC3888261 DOI: 10.1093/hmg/ddt459] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 09/16/2013] [Indexed: 01/22/2023] Open
Abstract
The GM2 gangliosidoses are progressive neurodegenerative disorders due to defects in the lysosomal β-N-acetylhexosaminidase system. Accumulation of β-hexosaminidases A and B substrates is presumed to cause this fatal condition. An authentic mouse model of Sandhoff disease (SD) with pathological characteristics resembling those noted in infantile GM2 gangliosidosis has been described. We have shown that expression of β-hexosaminidase by intracranial delivery of recombinant adeno-associated viral vectors to young adult SD mice can prevent many features of the disease and extends lifespan. To investigate the nature of the neurological injury in GM2 gangliosidosis and the extent of its reversibility, we have examined the evolution of disease in the SD mouse; we have moreover explored the effects of gene transfer delivered at key times during the course of the illness. Here we report greatly increased survival only when the therapeutic genes are expressed either before the disease is apparent or during its early manifestations. However, irrespective of when treatment was administered, widespread and abundant expression of β-hexosaminidase with consequent clearance of glycoconjugates, α-synuclein and ubiquitinated proteins, and abrogation of inflammatory responses and neuronal loss was observed. We also show that defects in myelination occur in early life and cannot be easily resolved when treatment is given to the adult brain. These results indicate that there is a limited temporal opportunity in which function and survival can be improved-but regardless of resolution of the cardinal pathological features of GM2 gangliosidosis, a point is reached when functional deterioration and death cannot be prevented.
Collapse
Affiliation(s)
| | - Susan Z. Wang
- Department of Medicine, University of Cambridge, Cambridge, UK and
| | | | | | - Timothy M. Cox
- Department of Medicine, University of Cambridge, Cambridge, UK and
| |
Collapse
|
13
|
Matsuzaki Y, Oue M, Hirai H. Generation of a neurodegenerative disease mouse model using lentiviral vectors carrying an enhanced synapsin I promoter. J Neurosci Methods 2013; 223:133-43. [PMID: 24361760 DOI: 10.1016/j.jneumeth.2013.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 11/26/2013] [Accepted: 12/05/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND Certain inherited progressive neurodegenerative disorders, such as spinocerebellar ataxia (SCA), affect neurons in large areas of the central nervous system (CNS). The selective expression of disease-causing and therapeutic genes in susceptible regions and cell types is critical for the generation of animal models and development of gene therapies for these diseases. Previous studies have demonstrated the advantages of the short synapsin I (SynI) promoter (0.5 kb) as a neuron-specific promoter for robust transgene expression. However, the short SynI promoter has also shown some promoter activity in glia and a lack of transgene expression in significant areas of the CNS. New methods: To improve the SynI promoter, we used a SynI promoter that is twice as long (1.0 kb) as the short SynI promoter and incorporated a minimal CMV (minCMV) sequence. RESULTS We observed that the 1.0 kb rat SynI promoter with minCMV [rSynI(1.0)-minCMV] exhibited robust promoter strength, excellent neuronal specificity and wide-ranging transgene expression throughout the CNS. Comparison with existing methods: Compared with the two previously reported short (0.5 kb) promoters, the new promoter was superior with respect to neuronal specificity and more efficiently transduced neurons. Moreover, transgenic mice expressing the mutant protein ATXN1[Q98], which causes SCA type 1 (SCA1), under the control of the rSynI(1.0)-minCMV promoter showed robust transgene expression specifically in neurons throughout the CNS and exhibited progressive ataxia. CONCLUSION rSynI(1.0)-minCMV drives robust and neuron-specific transgene expression throughout the CNS and is therefore useful for viral vector-mediated neuron-specific gene delivery and generation of neuron-specific transgenic animals.
Collapse
Affiliation(s)
- Yasunori Matsuzaki
- Department of Neurophysiology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Miho Oue
- Department of Neurophysiology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hirokazu Hirai
- Department of Neurophysiology, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
14
|
Abstract
Gangliosides are the main glycolipids of neuronal plasma membranes. Their surface patterns are generated by coordinated processes, involving biosynthetic pathways of the secretory compartments, catabolic steps of the endolysosomal system, and intracellular trafficking. Inherited defects in ganglioside biosynthesis causing fatal neurodegenerative diseases have been described so far almost exclusively in mouse models, whereas inherited defects in ganglioside catabolism causing various clinical forms of GM1- and GM2-gangliosidoses have long been known. For digestion, gangliosides are endocytosed and reach intra-endosomal vesicles. At the level of late endosomes, they are depleted of membrane-stabilizing lipids like cholesterol and enriched with bis(monoacylglycero)phosphate (BMP). Lysosomal catabolism is catalyzed at acidic pH values by cationic sphingolipid activator proteins (SAPs), presenting lipids to their respective hydrolases, electrostatically attracted to the negatively charged surface of the luminal BMP-rich vesicles. Various inherited defects of ganglioside hydrolases, e.g., of β-galactosidase and β-hexosaminidases, and of GM2-activator protein, cause infantile (with tetraparesis, dementia, blindness) and different protracted clinical forms of GM1- and GM2-gangliosidoses. Mutations yielding proteins with small residual catabolic activities in the lysosome give rise to juvenile and adult clinical forms with a wide range of clinical symptomatology. Apart from patients' differences in their genetic background, clinical heterogeneity may be caused by rather diverse substrate specificities and functions of lysosomal hydrolases, multifunctional properties of SAPs, and the strong regulation of ganglioside catabolism by membrane lipids. Currently, there is no treatment available for neuronal ganglioside storage diseases. Therapeutic approaches in mouse models and patients with juvenile forms of gangliosidoses are discussed.
Collapse
|