1
|
Viana R, Rubio T, Campos-Rodríguez Á, Sanz P. Glial Alterations in the Glutamatergic and GABAergic Signalling Pathways in a Mouse Model of Lafora Disease, a Severe Form of Progressive Myoclonus Epilepsy. Neuropathol Appl Neurobiol 2025; 51:e70009. [PMID: 40035482 DOI: 10.1111/nan.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
AIMS Lafora disease (LD; OMIM#254780) is a rare form of progressive myoclonus epilepsy characterised by the accumulation of insoluble deposits of glycogen in the brain and peripheral tissues. In mouse models of LD, we have identified neuroinflammation as a secondary hallmark of the disease, characterised by increased levels of reactive astrocytes and activated microglia. Our previous work demonstrated that the TNF and IL-6 inflammatory signalling pathways are the primary drivers of this neuroinflammatory phenotype. In this work, we aimed to investigate whether TNF and IL-6 pathway activation contributes to alterations in the glutamatergic and GABAergic signalling pathways. METHODS We performed immunofluorescence and western blot analyses on the hippocampus of a mouse model of LD to evaluate potential changes in proteins associated with glutamatergic and GABAergic signalling pathways. RESULTS Our findings reveal dysregulation in the expression of subunits of excitatory glutamatergic receptors (phospho-GluN2B and GluK2), as well as an increase in the levels of the GABA transporter GAT1. In addition, we detected activated forms of the Src and Lyn protein kinases in the hippocampus. More importantly, these alterations predominantly occur in nonneuronal cells, such as reactive astrocytes and microglia, underscoring the critical involvement of glial cells in the pathophysiology of LD. CONCLUSIONS The observed upregulation of glutamatergic receptor subunits likely amplifies excitatory glutamatergic signalling, whereas the increased expression of GAT1 may reduce the inhibitory GABAergic tone. These changes contribute to the characteristic hyperexcitability of LD.
Collapse
Affiliation(s)
- Rosa Viana
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - Teresa Rubio
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Valencia, Spain
- Faculty of Health Science, Universidad Europea de Valencia, Valencia, Spain
| | - Ángela Campos-Rodríguez
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Valencia, Spain
| | - Pascual Sanz
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Valencia, Spain
| |
Collapse
|
2
|
Nitschke S, Montalbano AP, Whiting ME, Smith BH, Mukherjee-Roy N, Marchioni CR, Sullivan MA, Zhao X, Wang P, Mount H, Verma M, Minassian BA, Nitschke F. Glycogen synthase GYS1 overactivation contributes to glycogen insolubility and malto-oligoglucan-associated neurodegenerative disease. EMBO J 2025; 44:1379-1413. [PMID: 39806098 PMCID: PMC11876434 DOI: 10.1038/s44318-024-00339-3] [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: 05/01/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025] Open
Abstract
Polyglucosans are glycogen molecules with overlong chains, which are hyperphosphorylated in the neurodegenerative Lafora disease (LD). Brain polyglucosan bodies (PBs) cause fatal neurodegenerative diseases including Lafora disease and adult polyglucosan body disease (ABPD), for which treatments, biomarkers, and good understanding of their pathogenesis are currently missing. Mutations in the genes for the phosphatase laforin or the E3 ubiquitin ligase malin can cause LD. By depleting PTG, an activator of the glycogen chain-elongating enzyme glycogen synthase (GYS1), in laforin- and malin-deficient LD mice, we show that abnormal glycogen chain lengths and not hyperphosphorylation underlie polyglucosan formation, and that polyglucosan bodies induce neuroinflammation. We provide evidence indicating that a small pool of overactive GYS1 contributes to glycogen insolubility in LD and APBD. In contrast to previous findings, metabolomics experiments using in situ-fixed brains reveal only modest metabolic changes in laforin-deficient mice. These changes are not replicated in malin-deficient or APBD mice, and are not normalized in rescued LD mice. Finally, we identify a pool of metabolically volatile malto-oligoglucans as a polyglucosan body- and neuroinflammation-associated brain energy source, and promising candidate biomarkers for LD and APBD, including malto-oligoglucans and the neurodegeneration marker CHI3L1/YKL40.
Collapse
Affiliation(s)
- Silvia Nitschke
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Alina P Montalbano
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Megan E Whiting
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Brandon H Smith
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Neije Mukherjee-Roy
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Charlotte R Marchioni
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Biochemistry and Molecular Genetics Department, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Mitchell A Sullivan
- Glycation and Diabetes Complications, Mater Research Institute - The University of Queensland, Translational Research Institute, Brisbane, QLD, 4102, Australia
- School of Health, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia
| | - Xiaochu Zhao
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada
| | - Peixiang Wang
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, M5G 0A4, Canada
| | - Howard Mount
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Psychiatry and Physiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Mayank Verma
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Berge A Minassian
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Felix Nitschke
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
3
|
Gumusgoz E, Kasiri S, Youssef I, Verma M, Chopra R, Villarreal Acha D, Wu J, Marriam U, Alao E, Chen X, Guisso DR, Gray SJ, Shah BR, Minassian BA. Focused ultrasound widely broadens AAV-delivered Cas9 distribution and activity. Gene Ther 2025:10.1038/s41434-025-00517-w. [PMID: 39893321 DOI: 10.1038/s41434-025-00517-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 01/09/2025] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
Because children have little temporal exposure to environment and aging, most pediatric neurological diseases are inherent, i.e. genetic. Since postnatal neurons and astrocytes are mostly non-replicating, gene therapy and genome editing present enormous promise in child neurology. Unlike in other organs, which are highly permissive to adeno-associated viruses (AAV), the mature blood-brain barrier (BBB) greatly limits circulating AAV distribution to the brain. Intrathecal administration improves distribution but to no more than 20% of brain cells. Focused ultrasound (FUS) opens the BBB transiently and safely. In the present work we opened the hippocampal BBB and delivered a Cas9 gene via AAV9 intrathecally. This allowed brain first-pass, and subsequent vascular circulation and re-entry through the opened BBB. The mouse model used was of Lafora disease, a neuroinflammatory disease due to accumulations of misshapen overlong-branched glycogen. Cas9 was targeted to the gene of the glycogen branch-elongating enzyme glycogen synthase. We show that FUS dramatically (2000-fold) improved hippocampal Cas9 distribution and greatly reduced the pathogenic glycogen accumulations and hippocampal inflammation. FUS is in regular clinical use for other indications. Our work shows that it has the potential to vastly broaden gene delivery or editing along with clearance of corresponding pathologic basis of brain disease.
Collapse
Affiliation(s)
- Emrah Gumusgoz
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sahba Kasiri
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ibrahim Youssef
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, Egypt
- Focused Ultrasound Lab and Program, Department of Radiology, UTSW Medical Center, Dallas, TX, USA
- FUS Instruments, Inc, Addison, TX, USA
| | - Mayank Verma
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Rajiv Chopra
- Focused Ultrasound Lab and Program, Department of Radiology, UTSW Medical Center, Dallas, TX, USA
- FUS Instruments, Inc, Addison, TX, USA
- Advanced Imaging Research Center, UTSW Medical Center, Dallas, TX, USA
- Solenic Medical Inc., Addison, TX, USA
| | - Daniel Villarreal Acha
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jun Wu
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ummay Marriam
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Esther Alao
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xin Chen
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Dikran R Guisso
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Steven J Gray
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Bhavya R Shah
- Focused Ultrasound Lab and Program, Department of Radiology, UTSW Medical Center, Dallas, TX, USA.
- Advanced Neuroscience Imaging Research Lab, Department of Radiology, UTSW Medical Center, Dallas, TX, USA.
- Department of Neurology, UTSW Medical Center, Dallas, TX, USA.
| | - Berge A Minassian
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
4
|
Della Vecchia S, Imbrici P, Liantonio A, Naef V, Damiani D, Licitra R, Bernardi S, Marchese M, Santorelli FM. Dapagliflozin ameliorates Lafora disease phenotype in a zebrafish model. Biomed Pharmacother 2025; 183:117800. [PMID: 39753095 PMCID: PMC11794196 DOI: 10.1016/j.biopha.2024.117800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 02/08/2025] Open
Abstract
Lafora disease (LD) is an ultra-rare and still incurable neurodegenerative condition. Although several therapeutic strategies are being explored, including gene therapy, there are currently no treatments that can alleviate the course of the disease and slow its progression. Recently, gliflozins, a series of SGLT2 transporter inhibitors approved for use in type 2 diabetes mellitus, heart failure and chronic kidney disease, have been proposed as possible repositioning drugs for the treatment of LD. With this in mind, we tested dapagliflozin (50 µM), canagliflozin (2.5 µM) and empagliflozin (200 µM) in our epm2a-/- zebrafish model, investigating their effects on pathological behaviour. In the case of dapagliflozin, we also investigated the possible mechanisms of action. Overall, the gliflozins reduced or rescued neuronal hyperexcitability and locomotor impairment. Dapagliflozin also reduced spontaneous seizure-like events in epm2a-/- larvae. At the biochemical and molecular level, dapagliflozin was found to slightly reduce glycogen content, and suppress inflammation and oxidative stress. It also ameliorates autophagic homeostasis and improves lysosomal markers. In conclusion, our preclinical study showed that dapagliflozin was able to ameliorate part of the pathological phenotype of epm2a-/- zebrafish larvae and could potentially be a suitable drug for repurposing in LD. However, since our model does not present Lafora bodies (LBs), at this early disease stage at least, it would be important to use mouse models in order to ascertain whether it is able to prevent or reduce LB formation.
Collapse
Affiliation(s)
- Stefania Della Vecchia
- IRCCS Stella Maris Foundation, Calambrone, via dei Giacinti 2, Pisa 56128, Italy; Department of Neurosciences, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Viale Pieraccini, 6, Florence 50139, Italy.
| | - Paola Imbrici
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Antonella Liantonio
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Valentina Naef
- IRCCS Stella Maris Foundation, Calambrone, via dei Giacinti 2, Pisa 56128, Italy
| | - Devid Damiani
- IRCCS Stella Maris Foundation, Calambrone, via dei Giacinti 2, Pisa 56128, Italy
| | - Rosario Licitra
- Department of Veterinary Sciences, University of Pisa, Pisa 56124, Italy
| | - Sara Bernardi
- IRCCS Stella Maris Foundation, Calambrone, via dei Giacinti 2, Pisa 56128, Italy
| | - Maria Marchese
- IRCCS Stella Maris Foundation, Calambrone, via dei Giacinti 2, Pisa 56128, Italy
| | | |
Collapse
|
5
|
Zafra-Puerta L, Iglesias-Cabeza N, Burgos DF, Sciaccaluga M, González-Fernández J, Bellingacci L, Canonichesi J, Sánchez-Martín G, Costa C, Sánchez MP, Serratosa JM. Gene therapy for Lafora disease in the Epm2a -/- mouse model. Mol Ther 2024; 32:2130-2149. [PMID: 38796707 PMCID: PMC11286821 DOI: 10.1016/j.ymthe.2024.05.032] [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: 12/22/2023] [Revised: 04/23/2024] [Accepted: 05/23/2024] [Indexed: 05/28/2024] Open
Abstract
Lafora disease is a rare and fatal form of progressive myoclonic epilepsy typically occurring early in adolescence. The disease results from mutations in the EPM2A gene, encoding laforin, or the EPM2B gene, encoding malin. Laforin and malin work together in a complex to control glycogen synthesis and prevent the toxicity produced by misfolded proteins via the ubiquitin-proteasome system. Disruptions in either protein cause alterations in this complex, leading to the formation of Lafora bodies containing abnormal, insoluble, and hyperphosphorylated forms of glycogen. We used the Epm2a-/- knockout mouse model of Lafora disease to apply gene therapy by administering intracerebroventricular injections of a recombinant adeno-associated virus carrying the human EPM2A gene. We evaluated the effects of this treatment through neuropathological studies, behavioral tests, video-electroencephalography, electrophysiological recordings, and proteomic/phosphoproteomic analysis. Gene therapy ameliorated neurological and histopathological alterations, reduced epileptic activity and neuronal hyperexcitability, and decreased the formation of Lafora bodies. Moreover, differential quantitative proteomics and phosphoproteomics revealed beneficial changes in various molecular pathways altered in Lafora disease. Our results represent proof of principle for gene therapy with the coding region of the human EPM2A gene as a treatment for EPM2A-related Lafora disease.
Collapse
Affiliation(s)
- Luis Zafra-Puerta
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, 28029 Madrid, Spain; Fondazione Malattie Rare Mauro Baschirotto BIRD Onlus, Longare (VI), Italy
| | - Nerea Iglesias-Cabeza
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Daniel F Burgos
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; PhD Program in Neuroscience, Universidad Autonoma de Madrid-Cajal Institute, 28029 Madrid, Spain
| | - Miriam Sciaccaluga
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; Fondazione Malattie Rare Mauro Baschirotto BIRD Onlus, Longare (VI), Italy
| | - Juan González-Fernández
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain; Departament of Microbiology and Parasitology, Faculty of Pharmacy, Complutense University of Madrid, University of Perugia, 06132 Perugia, Italy
| | - Laura Bellingacci
- Section of Physiology and Biochemistry, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Jacopo Canonichesi
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Gema Sánchez-Martín
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Cinzia Costa
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Marina P Sánchez
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain.
| | - José M Serratosa
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain.
| |
Collapse
|
6
|
Rubio T, Campos-Rodríguez Á, Sanz P. Beneficial Effect of Fingolimod in a Lafora Disease Mouse Model by Preventing Reactive Astrogliosis-Derived Neuroinflammation and Brain Infiltration of T-lymphocytes. Mol Neurobiol 2024; 61:3105-3120. [PMID: 37971656 PMCID: PMC11087365 DOI: 10.1007/s12035-023-03766-1] [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: 06/09/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
Lafora disease (LD; OMIM#254780) is a rare, devastating, and fatal form of progressive myoclonus epilepsy that affects young adolescents and has no treatment yet. One of the hallmarks of the disease is the accumulation of aberrant poorly branched forms of glycogen (polyglucosans, PGs) in the brain and peripheral tissues. The current hypothesis is that this accumulation is causative of the pathophysiology of the disease. Another hallmark of LD is the presence of neuroinflammation. We have recently reported the presence of reactive glia-derived neuroinflammation in LD mouse models and defined the main inflammatory pathways that operate in these mice, mainly TNF and IL-6 signaling pathways. In addition, we described the presence of infiltration of peripheral immune cells in the brain parenchyma, which could cooperate and aggravate the neuroinflammatory landscape of LD. In this work, we have checked the beneficial effect of two compounds with the capacity to ameliorate neuroinflammation and reduce leukocyte infiltration into the brain, namely fingolimod and dimethyl fumarate. Our results indicate a beneficial effect of fingolimod in reducing reactive astrogliosis-derived neuroinflammation and T-lymphocyte infiltration, which correlated with the improved behavioral performance of the treated Epm2b-/- mice. On the contrary, dimethyl fumarate, although it was able to reduce reactive astrogliosis, was less effective in preventing neuroinflammation and T-lymphocyte infiltration and in modifying behavioral tests.
Collapse
Affiliation(s)
- Teresa Rubio
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Jaime Roig 11, 46010, Valencia, Spain
| | - Ángela Campos-Rodríguez
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Jaime Roig 11, 46010, Valencia, Spain
| | - Pascual Sanz
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (CSIC), Jaime Roig 11, 46010, Valencia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010, Valencia, Spain.
| |
Collapse
|
7
|
Mitra S, Chen B, Shelton JM, Nitschke S, Wu J, Covington L, Dear M, Lynn T, Verma M, Nitschke F, Fuseya Y, Iwai K, Evers BM, Minassian BA. Myofiber-type-dependent 'boulder' or 'multitudinous pebble' formations across distinct amylopectinoses. Acta Neuropathol 2024; 147:46. [PMID: 38411740 DOI: 10.1007/s00401-024-02698-x] [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: 11/23/2023] [Revised: 01/13/2024] [Accepted: 01/24/2024] [Indexed: 02/28/2024]
Abstract
At least five enzymes including three E3 ubiquitin ligases are dedicated to glycogen's spherical structure. Absence of any reverts glycogen to a structure resembling amylopectin of the plant kingdom. This amylopectinosis (polyglucosan body formation) causes fatal neurological diseases including adult polyglucosan body disease (APBD) due to glycogen branching enzyme deficiency, Lafora disease (LD) due to deficiencies of the laforin glycogen phosphatase or the malin E3 ubiquitin ligase and type 1 polyglucosan body myopathy (PGBM1) due to RBCK1 E3 ubiquitin ligase deficiency. Little is known about these enzymes' functions in glycogen structuring. Toward understanding these functions, we undertake a comparative murine study of the amylopectinoses of APBD, LD and PGBM1. We discover that in skeletal muscle, polyglucosan bodies form as two main types, small and multitudinous ('pebbles') or giant and single ('boulders'), and that this is primarily determined by the myofiber types in which they form, 'pebbles' in glycolytic and 'boulders' in oxidative fibers. This pattern recapitulates what is known in the brain in LD, innumerable dust-like in astrocytes and single giant sized in neurons. We also show that oxidative myofibers are relatively protected against amylopectinosis, in part through highly increased glycogen branching enzyme expression. We present evidence of polyglucosan body size-dependent cell necrosis. We show that sex influences amylopectinosis in genotype, brain region and myofiber-type-specific fashion. RBCK1 is a component of the linear ubiquitin chain assembly complex (LUBAC), the only known cellular machinery for head-to-tail linear ubiquitination critical to numerous cellular pathways. We show that the amylopectinosis of RBCK1 deficiency is not due to loss of linear ubiquitination, and that another function of RBCK1 or LUBAC must exist and operate in the shaping of glycogen. This work opens multiple new avenues toward understanding the structural determinants of the mammalian carbohydrate reservoir critical to neurologic and neuromuscular function and disease.
Collapse
Affiliation(s)
- Sharmistha Mitra
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA.
| | - Baozhi Chen
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA
| | - John M Shelton
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9148, USA
| | - Silvia Nitschke
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA
| | - Jun Wu
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA
| | - Lindsay Covington
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9148, USA
| | - Mathew Dear
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA
| | - Tori Lynn
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA
| | - Mayank Verma
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA
| | - Felix Nitschke
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA
| | - Yasuhiro Fuseya
- Department of Molecular and Cellular Physiology, Kyoto University School of Medicine, Kyoto, 606-8501, Japan
| | - Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Kyoto University School of Medicine, Kyoto, 606-8501, Japan
| | - Bret M Evers
- Departments of Pathology and Ophthalmology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-9073, USA
| | - Berge A Minassian
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9063, USA.
| |
Collapse
|
8
|
Zimmern V, Minassian B. Progressive Myoclonus Epilepsy: A Scoping Review of Diagnostic, Phenotypic and Therapeutic Advances. Genes (Basel) 2024; 15:171. [PMID: 38397161 PMCID: PMC10888128 DOI: 10.3390/genes15020171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The progressive myoclonus epilepsies (PME) are a diverse group of disorders that feature both myoclonus and seizures that worsen gradually over a variable timeframe. While each of the disorders is individually rare, they collectively make up a non-trivial portion of the complex epilepsy and myoclonus cases that are seen in tertiary care centers. The last decade has seen substantial progress in our understanding of the pathophysiology, diagnosis, prognosis, and, in select disorders, therapies of these diseases. In this scoping review, we examine English language publications from the past decade that address diagnostic, phenotypic, and therapeutic advances in all PMEs. We then highlight the major lessons that have been learned and point out avenues for future investigation that seem promising.
Collapse
Affiliation(s)
- Vincent Zimmern
- Division of Child Neurology, University of Texas Southwestern, Dallas, TX 75390, USA;
| | | |
Collapse
|
9
|
Cordoba-Caballero J, Perkins JR, García-Criado F, Gallego D, Navarro-Sánchez A, Moreno-Estellés M, Garcés C, Bonet F, Romá-Mateo C, Toro R, Perez B, Sanz P, Kohl M, Rojano E, Seoane P, Ranea JAG. Exploring miRNA-target gene pair detection in disease with coRmiT. Brief Bioinform 2024; 25:bbae060. [PMID: 38436559 PMCID: PMC10939301 DOI: 10.1093/bib/bbae060] [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: 09/21/2023] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 03/05/2024] Open
Abstract
A wide range of approaches can be used to detect micro RNA (miRNA)-target gene pairs (mTPs) from expression data, differing in the ways the gene and miRNA expression profiles are calculated, combined and correlated. However, there is no clear consensus on which is the best approach across all datasets. Here, we have implemented multiple strategies and applied them to three distinct rare disease datasets that comprise smallRNA-Seq and RNA-Seq data obtained from the same samples, obtaining mTPs related to the disease pathology. All datasets were preprocessed using a standardized, freely available computational workflow, DEG_workflow. This workflow includes coRmiT, a method to compare multiple strategies for mTP detection. We used it to investigate the overlap of the detected mTPs with predicted and validated mTPs from 11 different databases. Results show that there is no clear best strategy for mTP detection applicable to all situations. We therefore propose the integration of the results of the different strategies by selecting the one with the highest odds ratio for each miRNA, as the optimal way to integrate the results. We applied this selection-integration method to the datasets and showed it to be robust to changes in the predicted and validated mTP databases. Our findings have important implications for miRNA analysis. coRmiT is implemented as part of the ExpHunterSuite Bioconductor package available from https://bioconductor.org/packages/ExpHunterSuite.
Collapse
Affiliation(s)
- Jose Cordoba-Caballero
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur, 31, Málaga, 29010, Spain
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
| | - James R Perkins
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur, 31, Málaga, 29010, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), C/ Severo Ochoa, 35, Parque Tecnológico de Andalucía (PTA), Campanillas, Málaga, 29590, Spain
| | - Federico García-Criado
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur, 31, Málaga, 29010, Spain
| | - Diana Gallego
- CIBER de Enfermedades Raras (CIBERER), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, Madrid, 28029, Spain
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular-SO UAM-CSIC, Universidad Autónoma de Madrid, Campus de Cantoblanco, Madrid, Spain
- Instituto de Investigación Sanitaria IdiPaZ, Madrid, Spain
| | - Alicia Navarro-Sánchez
- CIBER de Enfermedades Raras (CIBERER), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, Madrid, 28029, Spain
- Departament de Fisiologia, Facultat de Medicina i Odontologia, Universitat de València, Av. Blasco Ibáñez 15, 46010, València, Spain
| | - Mireia Moreno-Estellés
- CIBER de Enfermedades Raras (CIBERER), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, Madrid, 28029, Spain
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Jaime Roig 11, 46010, Valencia, Spain
| | - Concepción Garcés
- CIBER de Enfermedades Raras (CIBERER), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, Madrid, 28029, Spain
- Departament de Fisiologia, Facultat de Medicina i Odontologia, Universitat de València, Av. Blasco Ibáñez 15, 46010, València, Spain
| | - Fernando Bonet
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
- Medicine Department, School of Medicine, University of Cádiz, Cádiz, Spain
| | - Carlos Romá-Mateo
- CIBER de Enfermedades Raras (CIBERER), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, Madrid, 28029, Spain
- Departament de Fisiologia, Facultat de Medicina i Odontologia, Universitat de València, Av. Blasco Ibáñez 15, 46010, València, Spain
- Incliva Biomedical Research Institute, 46010, València, Spain
| | - Rocio Toro
- Research Unit, Biomedical Research and Innovation Institute of Cádiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
- Medicine Department, School of Medicine, University of Cádiz, Cádiz, Spain
| | - Belén Perez
- CIBER de Enfermedades Raras (CIBERER), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, Madrid, 28029, Spain
- Centro de Diagnóstico de Enfermedades Moleculares, Centro de Biología Molecular-SO UAM-CSIC, Universidad Autónoma de Madrid, Campus de Cantoblanco, Madrid, Spain
- Instituto de Investigación Sanitaria IdiPaZ, Madrid, Spain
| | - Pascual Sanz
- CIBER de Enfermedades Raras (CIBERER), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, Madrid, 28029, Spain
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Jaime Roig 11, 46010, Valencia, Spain
| | - Matthias Kohl
- Faculty of Medical and Life Sciences, Furtwangen University, Germany
| | - Elena Rojano
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur, 31, Málaga, 29010, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), C/ Severo Ochoa, 35, Parque Tecnológico de Andalucía (PTA), Campanillas, Málaga, 29590, Spain
| | - Pedro Seoane
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur, 31, Málaga, 29010, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), C/ Severo Ochoa, 35, Parque Tecnológico de Andalucía (PTA), Campanillas, Málaga, 29590, Spain
- CIBER de Enfermedades Raras (CIBERER), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, Madrid, 28029, Spain
| | - Juan A G Ranea
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Bulevar Louis Pasteur, 31, Málaga, 29010, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA-Plataforma BIONAND), C/ Severo Ochoa, 35, Parque Tecnológico de Andalucía (PTA), Campanillas, Málaga, 29590, Spain
- CIBER de Enfermedades Raras (CIBERER), Avda. Monforte de Lemos, 3-5, Pabellón 11, Planta 0, Madrid, 28029, Spain
- Instituto Nacional de Bioinformática (INB/ELIXIR-ES), Instituto de Salud Carlos III (ISCIII), C/ Sinesio Delgado, 4, Madrid, 28029, Spain
| |
Collapse
|
10
|
Donohue KJ, Fitzsimmons B, Bruntz RC, Markussen KH, Young LEA, Clarke HA, Coburn PT, Griffith LE, Sanders W, Klier J, Burke SN, Maurer AP, Minassian BA, Sun RC, Kordasiewisz HB, Gentry MS. Gys1 Antisense Therapy Prevents Disease-Driving Aggregates and Epileptiform Discharges in a Lafora Disease Mouse Model. Neurotherapeutics 2023; 20:1808-1819. [PMID: 37700152 PMCID: PMC10684475 DOI: 10.1007/s13311-023-01434-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 09/14/2023] Open
Abstract
Patients with Lafora disease have a mutation in EPM2A or EPM2B, resulting in dysregulation of glycogen metabolism throughout the body and aberrant glycogen molecules that aggregate into Lafora bodies. Lafora bodies are particularly damaging in the brain, where the aggregation drives seizures with increasing severity and frequency, coupled with neurodegeneration. Previous work employed mouse genetic models to reduce glycogen synthesis by approximately 50%, and this strategy significantly reduced Lafora body formation and disease phenotypes. Therefore, an antisense oligonucleotide (ASO) was developed to reduce glycogen synthesis in the brain by targeting glycogen synthase 1 (Gys1). To test the distribution and efficacy of this drug, the Gys1-ASO was administered to Epm2b-/- mice via intracerebroventricular administration at 4, 7, and 10 months. The mice were then sacrificed at 13 months and their brains analyzed for Gys1 expression, glycogen aggregation, and neuronal excitability. The mice treated with Gys1-ASO exhibited decreased Gys1 protein levels, decreased glycogen aggregation, and reduced epileptiform discharges compared to untreated Epm2b-/- mice. This work provides proof of concept that a Gys1-ASO halts disease progression of EPM2B mutations of Lafora disease.
Collapse
Affiliation(s)
- Katherine J Donohue
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Bethany Fitzsimmons
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Carlsbad, CA, 92010, USA
| | - Ronald C Bruntz
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Kia H Markussen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Lyndsay E A Young
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Harrison A Clarke
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, 32610, USA
| | - Peyton T Coburn
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Laiken E Griffith
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - William Sanders
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Jack Klier
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Sara N Burke
- Department of Neuroscience and Center for Cognitive Aging and Memory, University of Florida, Gainesville, FL, 32610, USA
| | - Andrew P Maurer
- Department of Neuroscience and Center for Cognitive Aging and Memory, University of Florida, Gainesville, FL, 32610, USA
| | - Berge A Minassian
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ramon C Sun
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, 32610, USA
| | - Holly B Kordasiewisz
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Carlsbad, CA, 92010, USA
| | - Matthew S Gentry
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
11
|
Timechko EE, Yakimov AM, Paramonova AI, Usoltseva AA, Utyashev NP, Ivin NO, Utyasheva AA, Yakunina AV, Kalinin VA, Dmitrenko DV. Mass Spectrometry as a Quantitative Proteomic Analysis Tool for the Search for Temporal Lobe Epilepsy Biomarkers: A Systematic Review. Int J Mol Sci 2023; 24:11130. [PMID: 37446307 DOI: 10.3390/ijms241311130] [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: 05/30/2023] [Revised: 06/25/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common form of epilepsy in adults. Tissue reorganization at the site of the epileptogenic focus is accompanied by changes in the expression patterns of protein molecules. The study of mRNA and its corresponding proteins is crucial for understanding the pathogenesis of the disease. Protein expression profiles do not always directly correlate with the levels of their transcripts; therefore, it is protein profiling that is no less important for understanding the molecular mechanisms and biological processes of TLE. The study and annotation of proteins that are statistically significantly different in patients with TLE is an approach to search for biomarkers of this disease, various stages of its development, as well as a method for searching for specific targets for the development of a further therapeutic strategy. When writing a systematic review, the following aggregators of scientific journals were used: MDPI, PubMed, ScienceDirect, Springer, and Web of Science. Scientific articles were searched using the following keywords: "proteomic", "mass-spectrometry", "protein expression", "temporal lobe epilepsy", and "biomarkers". Publications from 2003 to the present have been analyzed. Studies of brain tissues, experimental models of epilepsy, as well as biological fluids, were analyzed. For each of the groups, aberrantly expressed proteins found in various studies were isolated. Most of the studies omitted important characteristics of the studied patients, such as: duration of illness, type and response to therapy, gender, etc. Proteins that overlap across different tissue types and different studies have been highlighted: DPYSL, SYT1, STMN1, APOE, NME1, and others. The most common biological processes for them were the positive regulation of neurofibrillary tangle assembly, the regulation of amyloid fibril formation, lipoprotein catabolic process, the positive regulation of vesicle fusion, the positive regulation of oxidative stress-induced intrinsic apoptotic signaling pathway, removal of superoxide radicals, axon extension, and the regulation of actin filament depolymerization. MS-based proteomic profiling for a relevant study must accept a number of limitations, the most important of which is the need to compare different types of neurological and, in particular, epileptic disorders. Such a criterion could increase the specificity of the search work and, in the future, lead to the discovery of biomarkers for a particular disease.
Collapse
Affiliation(s)
- Elena E Timechko
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Alexey M Yakimov
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Anastasia I Paramonova
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Anna A Usoltseva
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Nikita P Utyashev
- Federal State Budgetary Institution "National Medical and Surgical Center Named after N.I. Pirogov", 105203 Moscow, Russia
| | - Nikita O Ivin
- Federal State Budgetary Institution "National Medical and Surgical Center Named after N.I. Pirogov", 105203 Moscow, Russia
| | - Anna A Utyasheva
- Federal State Budgetary Institution "National Medical and Surgical Center Named after N.I. Pirogov", 105203 Moscow, Russia
| | - Albina V Yakunina
- Department of Neurology and Neurobiology of Postgraduate Education, Samara State Medical University, 443079 Samara, Russia
| | - Vladimir A Kalinin
- Department of Neurology and Neurobiology of Postgraduate Education, Samara State Medical University, 443079 Samara, Russia
| | - Diana V Dmitrenko
- Department of Medical Genetics and Clinical Neurophysiology of Postgraduate Education, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| |
Collapse
|
12
|
Burgos DF, Sciaccaluga M, Worby CA, Zafra-Puerta L, Iglesias-Cabeza N, Sánchez-Martín G, Prontera P, Costa C, Serratosa JM, Sánchez MP. Epm2a R240X knock-in mice present earlier cognitive decline and more epileptic activity than Epm2a -/- mice. Neurobiol Dis 2023; 181:106119. [PMID: 37059210 DOI: 10.1016/j.nbd.2023.106119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/16/2023] Open
Abstract
Lafora disease is a rare recessive form of progressive myoclonic epilepsy, usually diagnosed during adolescence. Patients present with myoclonus, neurological deterioration, and generalized tonic-clonic, myoclonic, or absence seizures. Symptoms worsen until death, usually within the first ten years of clinical onset. The primary histopathological hallmark is the formation of aberrant polyglucosan aggregates called Lafora bodies in the brain and other tissues. Lafora disease is caused by mutations in either the EPM2A gene, encoding laforin, or the EPM2B gene, coding for malin. The most frequent EPM2A mutation is R241X, which is also the most prevalent in Spain. The Epm2a-/- and Epm2b-/- mouse models of Lafora disease show neuropathological and behavioral abnormalities similar to those seen in patients, although with a milder phenotype. To obtain a more accurate animal model, we generated the Epm2aR240X knock-in mouse line with the R240X mutation in the Epm2a gene, using genetic engineering based on CRISPR-Cas9 technology. Epm2aR240X mice exhibit most of the alterations reported in patients, including the presence of LBs, neurodegeneration, neuroinflammation, interictal spikes, neuronal hyperexcitability, and cognitive decline, despite the absence of motor impairments. The Epm2aR240X knock-in mouse displays some symptoms that are more severe that those observed in the Epm2a-/- knock-out, including earlier and more pronounced memory loss, increased levels of neuroinflammation, more interictal spikes and increased neuronal hyperexcitability, symptoms that more precisely resemble those observed in patients. This new mouse model can therefore be specifically used to evaluate how new therapies affects these features with greater precision.
Collapse
Affiliation(s)
- Daniel F Burgos
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid 28029, Spain; Program in Neuroscience, Autonoma de Madrid University-Cajal Institute, Madrid 28029, Spain
| | - Miriam Sciaccaluga
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy; Fondazione Malattie Rare Mauro Baschirotto BIRD Onlus, Longare (VI), Italy
| | - Carolyn A Worby
- University of California at San Diego, 9500 Gilman Drive, La Jolla CA92093-0721, USA
| | - Luis Zafra-Puerta
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid 28029, Spain; Program in Neuroscience, Autonoma de Madrid University-Cajal Institute, Madrid 28029, Spain; Fondazione Malattie Rare Mauro Baschirotto BIRD Onlus, Longare (VI), Italy
| | - Nerea Iglesias-Cabeza
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid 28029, Spain
| | - Gema Sánchez-Martín
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid 28029, Spain
| | - Paolo Prontera
- Medical Genetics Unit, S. Maria della Misericordia Hospital, Perugia 06132, Italy
| | - Cinzia Costa
- Section of Neurology, S. Maria della Misericordia Hospital, Department of Medicine and Surgery, University of Perugia, Perugia 06132, Italy
| | - José M Serratosa
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid 28029, Spain
| | - Marina P Sánchez
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid 28029, Spain.
| |
Collapse
|
13
|
Moreno-Estellés M, Campos-Rodríguez Á, Rubio-Villena C, Kumarasinghe L, Garcia-Gimeno MA, Sanz P. Deciphering the Polyglucosan Accumulation Present in Lafora Disease Using an Astrocytic Cellular Model. Int J Mol Sci 2023; 24:ijms24076020. [PMID: 37046993 PMCID: PMC10094345 DOI: 10.3390/ijms24076020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Lafora disease (LD) is a neurological disorder characterized by progressive myoclonus epilepsy. The hallmark of the disease is the presence of insoluble forms of glycogen (polyglucosan bodies, or PGBs) in the brain. The accumulation of PGBs is causative of the pathophysiological features of LD. However, despite the efforts made by different groups, the question of why PGBs accumulate in the brain is still unanswered. We have recently demonstrated that, in vivo, astrocytes accumulate most of the PGBs present in the brain, and this could lead to astrocyte dysfunction. To develop a deeper understanding of the defects present in LD astrocytes that lead to LD pathophysiology, we obtained pure primary cultures of astrocytes from LD mice from the postnatal stage under conditions that accumulate PGBs, the hallmark of LD. These cells serve as novel in vitro models for studying PGBs accumulation and related LD dysfunctions. In this sense, the metabolomics of LD astrocytes indicate that they accumulate metabolic intermediates of the upper part of the glycolytic pathway, probably as a consequence of enhanced glucose uptake. In addition, we also demonstrate the feasibility of using the model in the identification of different compounds that may reduce the accumulation of polyglucosan inclusions.
Collapse
Affiliation(s)
- Mireia Moreno-Estellés
- Instituto de Biomedicina de Valencia (IBV-CSIC), 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-ISCIII, 28029 Madrid, Spain
| | - Ángela Campos-Rodríguez
- Instituto de Biomedicina de Valencia (IBV-CSIC), 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-ISCIII, 28029 Madrid, Spain
| | - Carla Rubio-Villena
- Institute for Integrative Systems Biology (I2SysBio), Consejo Superior de Investigaciones Científicas (CSIC)-Universitat de València (UV), Parc Científic, Cat. Agustín Escardino 9, 46980 Paterna, Spain
| | - Lorena Kumarasinghe
- Instituto de Biomedicina de Valencia (IBV-CSIC), 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-ISCIII, 28029 Madrid, Spain
| | - Maria Adelaida Garcia-Gimeno
- Department of Biotechnology, Escuela Técnica Superior de Ingeniería Agronómica y del Medio Natural (ETSIAMN), Universitat Politécnica de València, 46022 Valencia, Spain
| | - Pascual Sanz
- Instituto de Biomedicina de Valencia (IBV-CSIC), 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-ISCIII, 28029 Madrid, Spain
- Consejo Superior de Investigaciones Científicas, Jaime Roig 11, 46010 Valencia, Spain
| |
Collapse
|
14
|
Duran J. Role of Astrocytes in the Pathophysiology of Lafora Disease and Other Glycogen Storage Disorders. Cells 2023; 12:cells12050722. [PMID: 36899857 PMCID: PMC10000527 DOI: 10.3390/cells12050722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/05/2023] [Accepted: 02/22/2023] [Indexed: 03/12/2023] Open
Abstract
Lafora disease is a rare disorder caused by loss of function mutations in either the EPM2A or NHLRC1 gene. The initial symptoms of this condition are most commonly epileptic seizures, but the disease progresses rapidly with dementia, neuropsychiatric symptoms, and cognitive deterioration and has a fatal outcome within 5-10 years after onset. The hallmark of the disease is the accumulation of poorly branched glycogen in the form of aggregates known as Lafora bodies in the brain and other tissues. Several reports have demonstrated that the accumulation of this abnormal glycogen underlies all the pathologic traits of the disease. For decades, Lafora bodies were thought to accumulate exclusively in neurons. However, it was recently identified that most of these glycogen aggregates are present in astrocytes. Importantly, astrocytic Lafora bodies have been shown to contribute to pathology in Lafora disease. These results identify a primary role of astrocytes in the pathophysiology of Lafora disease and have important implications for other conditions in which glycogen abnormally accumulates in astrocytes, such as Adult Polyglucosan Body disease and the buildup of Corpora amylacea in aged brains.
Collapse
Affiliation(s)
- Jordi Duran
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), 08017 Barcelona, Spain;
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| |
Collapse
|
15
|
Kumarasinghe L, Garcia-Gimeno MA, Ramirez J, Mayor U, Zugaza JL, Sanz P. P-Rex1 is a novel substrate of the E3 ubiquitin ligase Malin associated with Lafora disease. Neurobiol Dis 2023; 177:105998. [PMID: 36638890 PMCID: PMC10682699 DOI: 10.1016/j.nbd.2023.105998] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
Laforin and Malin are two proteins that are encoded by the genes EPM2A and EPM2B, respectively. Laforin is a glucan phosphatase and Malin is an E3-ubiquitin ligase, and these two proteins function as a complex. Mutations occurring at the level of one of the two genes lead to the accumulation of an aberrant form of glycogen meant to cluster in polyglucosans that go under the name of Lafora bodies. Individuals affected by the appearance of these polyglucosans, especially at the cerebral level, experience progressive neurodegeneration and several episodes of epilepsy leading to the manifestation of a fatal form of a rare disease called Lafora disease (LD), for which, to date, no treatment is available. Despite the different dysfunctions described for this disease, many molecular aspects still demand elucidation. An effective way to unknot some of the nodes that prevent the achievement of better knowledge of LD is to focus on the substrates that are ubiquitinated by the E3-ubiquitin ligase Malin. Some substrates have already been provided by previous studies based on protein-protein interaction techniques and have been associated with some alterations that mark the disease. In this work, we have used an unbiased alternative approach based on the activity of Malin as an E3-ubiquitin ligase. We report the discovery of novel bonafide substrates of Malin and have characterized one of them more deeply, namely PIP3-dependent Rac exchanger 1 (P-Rex1). The analysis conducted upon this substrate sets the genesis of the delineation of a molecular pathway that leads to altered glucose uptake, which could be one of the origin of the accumulation of the polyglucosans present in the disease.
Collapse
Affiliation(s)
- L Kumarasinghe
- Instituto de Biomedicina de Valencia, IBV-CSIC, 46010, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-ISCIII, 28029 Madrid, Spain
| | - M A Garcia-Gimeno
- Department of Biotechnology, Escuela Técnica Superior de Ingeniería Agronómica y del Medio Natural (ETSIAMN), Universitat Politécnica de València, 46022, Valencia, Spain
| | - J Ramirez
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, UPV/EHU, Leioa, Bizkaia, Spain
| | - U Mayor
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, UPV/EHU, Leioa, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Plaza Euskadi, 48009 Bilbao, Spain
| | - J L Zugaza
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi, 48009 Bilbao, Spain; Achucarro Basque Center for Neuroscience, Scientific Park UPV/EHU, 48940 Leioa, Bizkaia, Spain; Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, UPV/EHU, 48940 Leioa, Bizkaia, Spain
| | - P Sanz
- Instituto de Biomedicina de Valencia, IBV-CSIC, 46010, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)-ISCIII, 28029 Madrid, Spain.
| |
Collapse
|
16
|
Romá-Mateo C, Lorente-Pozo S, Márquez-Thibaut L, Moreno-Estellés M, Garcés C, González D, Lahuerta M, Aguado C, García-Giménez JL, Sanz P, Pallardó FV. Age-Related microRNA Overexpression in Lafora Disease Male Mice Provides Links between Neuroinflammation and Oxidative Stress. Int J Mol Sci 2023; 24:ijms24021089. [PMID: 36674605 PMCID: PMC9865572 DOI: 10.3390/ijms24021089] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Lafora disease is a rare, fatal form of progressive myoclonus epilepsy characterized by continuous neurodegeneration with epileptic seizures, characterized by the intracellular accumulation of aberrant polyglucosan granules called Lafora bodies. Several works have provided numerous evidence of molecular and cellular alterations in neural tissue from experimental mouse models deficient in either laforin or malin, two proteins related to the disease. Oxidative stress, alterations in proteostasis, and deregulation of inflammatory signals are some of the molecular alterations underlying this condition in both KO animal models. Lafora bodies appear early in the animal's life, but many of the aforementioned molecular aberrant processes and the consequent neurological symptoms ensue only as animals age. Here, using small RNA-seq and quantitative PCR on brain extracts from laforin and malin KO male mice of different ages, we show that two different microRNA species, miR-155 and miR-146a, are overexpressed in an age-dependent manner. We also observed altered expression of putative target genes for each of the microRNAs studied in brain extracts. These results open the path for a detailed dissection of the molecular consequences of laforin and malin deficiency in brain tissue, as well as the potential role of miR-155 and miR-146a as specific biomarkers of disease progression in LD.
Collapse
Affiliation(s)
- Carlos Romá-Mateo
- Department of Physiology, Facultat de Medicina i Odontologia, Universitat de València, 46010 Valencia, Spain
- Fundación Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)—ISCIII, 46010 Valencia, Spain
- Correspondence: (C.R.-M.); (P.S.); Tel.: +34-963983170 (C.R.-M.); +34-963391760 (P.S.)
| | - Sheila Lorente-Pozo
- Neonatal Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Lucía Márquez-Thibaut
- Institut d’Investigació Biomèdica de Girona Dr. Josep Trueta (IDIBGI), Parc Hospitalari Martí i Julià de Salt, 17190 Girona, Spain
| | - Mireia Moreno-Estellés
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)—ISCIII, 46010 Valencia, Spain
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, 46010 Valencia, Spain
| | - Concepción Garcés
- Department of Physiology, Facultat de Medicina i Odontologia, Universitat de València, 46010 Valencia, Spain
| | - Daymé González
- EpiDisease S.L. (Spin-off From the CIBER-ISCIII), Parc Científic de la Universitat de València, 46980 Paterna, Spain
- Novartis Institutes for BioMedical Research (NIBR), Novartis Campus, CH-4056 Basel, Switzerland
| | - Marcos Lahuerta
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, 46010 Valencia, Spain
| | - Carmen Aguado
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)—ISCIII, 46010 Valencia, Spain
| | - José Luis García-Giménez
- Department of Physiology, Facultat de Medicina i Odontologia, Universitat de València, 46010 Valencia, Spain
- Fundación Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)—ISCIII, 46010 Valencia, Spain
| | - Pascual Sanz
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)—ISCIII, 46010 Valencia, Spain
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, 46010 Valencia, Spain
- Correspondence: (C.R.-M.); (P.S.); Tel.: +34-963983170 (C.R.-M.); +34-963391760 (P.S.)
| | - Federico V. Pallardó
- Department of Physiology, Facultat de Medicina i Odontologia, Universitat de València, 46010 Valencia, Spain
- Fundación Instituto de Investigación Sanitaria INCLIVA, 46010 Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)—ISCIII, 46010 Valencia, Spain
| |
Collapse
|
17
|
Rubio T, Viana R, Moreno-Estellés M, Campos-Rodríguez Á, Sanz P. TNF and IL6/Jak2 signaling pathways are the main contributors of the glia-derived neuroinflammation present in Lafora disease, a fatal form of progressive myoclonus epilepsy. Neurobiol Dis 2023; 176:105964. [PMID: 36526090 PMCID: PMC10682476 DOI: 10.1016/j.nbd.2022.105964] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Lafora disease (LD; OMIM#254780) is a rare form of progressive myoclonus epilepsy (prevalence <1:1,000,000) characterized by the accumulation of insoluble deposits of aberrant glycogen (polyglucosans), named Lafora bodies, in the brain but also in peripheral tissues. LD is the most severe form of the group of progressive myoclonus epilepsies, since patients present a rapid deterioration and dementia with amplification of seizures, leading to death after a decade from the onset of the first symptoms. We have recently described that reactive glia-derived neuroinflammation should be considered a novel hallmark of LD since we observed a florid upregulation of differentially expressed genes in both LD mouse lines, which were mainly related to mediators of inflammatory response. In this work, we define an upregulation of the expression of mediators of the TNF and IL6/JAK2 signaling pathways in LD. In addition, we describe the activation of the non-canonical form of the inflammasome. Furthermore, we describe the infiltration of peripheral immune cells in the brain parenchyma, which could aggravate glia-derived neuroinflammation. Finally, we describe CXCL10 and S100b as blood biomarkers of the disease, which will allow the study of the progression of the disease using serum blood samples. We consider that the identification of these initial inflammatory changes in LD will be very important to implement possible anti-inflammatory therapeutic strategies to prevent the development of the disease.
Collapse
Affiliation(s)
- Teresa Rubio
- Instituto de Biomedicina de Valencia, CSIC, Jaime Roig 11, 46010 Valencia, Spain
| | - Rosa Viana
- Instituto de Biomedicina de Valencia, CSIC, Jaime Roig 11, 46010 Valencia, Spain
| | - Mireia Moreno-Estellés
- Instituto de Biomedicina de Valencia, CSIC, Jaime Roig 11, 46010 Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | | | - Pascual Sanz
- Instituto de Biomedicina de Valencia, CSIC, Jaime Roig 11, 46010 Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain..
| |
Collapse
|
18
|
Vezzani A, Ravizza T, Bedner P, Aronica E, Steinhäuser C, Boison D. Astrocytes in the initiation and progression of epilepsy. Nat Rev Neurol 2022; 18:707-722. [PMID: 36280704 PMCID: PMC10368155 DOI: 10.1038/s41582-022-00727-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 11/09/2022]
Abstract
Epilepsy affects ~65 million people worldwide. First-line treatment options include >20 antiseizure medications, but seizure control is not achieved in approximately one-third of patients. Antiseizure medications act primarily on neurons and can provide symptomatic control of seizures, but do not alter the onset and progression of epilepsy and can cause serious adverse effects. Therefore, medications with new cellular and molecular targets and mechanisms of action are needed. Accumulating evidence indicates that astrocytes are crucial to the pathophysiological mechanisms of epilepsy, raising the possibility that these cells could be novel therapeutic targets. In this Review, we discuss how dysregulation of key astrocyte functions - gliotransmission, cell metabolism and immune function - contribute to the development and progression of hyperexcitability in epilepsy. We consider strategies to mitigate astrocyte dysfunction in each of these areas, and provide an overview of how astrocyte activation states can be monitored in vivo not only to assess their contribution to disease but also to identify markers of disease processes and treatment effects. Improved understanding of the roles of astrocytes in epilepsy has the potential to lead to novel therapies to prevent the initiation and progression of epilepsy.
Collapse
Affiliation(s)
- Annamaria Vezzani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Teresa Ravizza
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
19
|
Della Vecchia S, Marchese M, Santorelli FM. Glial Contributions to Lafora Disease: A Systematic Review. Biomedicines 2022; 10:biomedicines10123103. [PMID: 36551859 PMCID: PMC9776290 DOI: 10.3390/biomedicines10123103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Lafora disease (LD) is a neurodegenerative condition characterized by the accumulation of polyglucosan bodies (PBs) throughout the brain. Alongside metabolic and molecular alterations, neuroinflammation has emerged as another key histopathological feature of LD. METHODS To investigate the role of astrocytes and microglia in LD, we performed a systematic review according to the PRISMA statement. PubMed, Scopus, and Web-of-Science database searches were performed independently by two reviewers. RESULTS Thirty-five studies analyzing the relationship of astrocytes and microglia with LD and/or the effects of anti-inflammatory treatments in LD animal models were identified and included in the review. Although LD has long been dominated by a neuronocentric view, a growing body of evidence suggests a role of glial cells in the disease, starting with the finding that these cells accumulate PBs. We discuss the potential meaning of glial PB accumulations, the likely factors activating glial cells, and the possible contribution of glial cells to LD neurodegeneration and epilepsy. CONCLUSIONS Given the evidence for the role of neuroinflammation in LD, future studies should consider glial cells as a potential therapeutic target for modifying/delaying LD progression; however, it should be kept in mind that these cells can potentially assume multiple reactive phenotypes, which could influence the therapeutic response.
Collapse
Affiliation(s)
- Stefania Della Vecchia
- Molecular Medicine and Neurogenetics, IRCCS Stella Maris Foundation, Calambrone, 56128 Pisa, Italy
- Correspondence: (S.D.V.); (F.M.S.)
| | - Maria Marchese
- Neurobiology, IRCCS Stella Maris Foundation, Calambrone, 56128 Pisa, Italy
| | - Filippo Maria Santorelli
- Molecular Medicine and Neurogenetics, IRCCS Stella Maris Foundation, Calambrone, 56128 Pisa, Italy
- Neurobiology, IRCCS Stella Maris Foundation, Calambrone, 56128 Pisa, Italy
- Correspondence: (S.D.V.); (F.M.S.)
| |
Collapse
|
20
|
Mollá B, Heredia M, Campos Á, Sanz P. Pharmacological Modulation of Glutamatergic and Neuroinflammatory Pathways in a Lafora Disease Mouse Model. Mol Neurobiol 2022; 59:6018-6032. [PMID: 35835895 PMCID: PMC9463199 DOI: 10.1007/s12035-022-02956-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022]
Abstract
Lafora disease (LD) is a fatal rare neurodegenerative disorder that affects young adolescents and has no treatment yet. The hallmark of LD is the presence of polyglucosan inclusions (PGs), called Lafora bodies (LBs), in the brain and peripheral tissues. LD is caused by mutations in either EPM2A or EPM2B genes, which, respectively, encode laforin, a glucan phosphatase, and malin, an E3-ubiquitin ligase, with identical clinical features. LD knockout mouse models (Epm2a - / - and Epm2b - / -) recapitulate PG body accumulation, as in the human pathology, and display alterations in glutamatergic transmission and neuroinflammatory pathways in the brain. In this work, we show the results of four pre-clinical trials based on the modulation of glutamatergic transmission (riluzole and memantine) and anti-neuroinflammatory interventions (resveratrol and minocycline) as therapeutical strategies in an Epm2b - / - mouse model. Drugs were administered in mice from 3 to 5 months of age, corresponding to early stage of the disease, and we evaluated the beneficial effect of the drugs by in vivo behavioral phenotyping and ex vivo histopathological brain analyses. The behavioral assessment was based on a battery of anxiety, cognitive, and neurodegenerative tests and the histopathological analyses included a panel of markers regarding PG accumulation, astrogliosis, and microgliosis. Overall, the outcome of ameliorating the excessive glutamatergic neurotransmission present in Epm2b - / - mice by memantine displayed therapeutic effectiveness at the behavioral levels. Modulation of neuroinflammation by resveratrol and minocycline also showed beneficial effects at the behavioral level. Therefore, our study suggests that both therapeutical strategies could be beneficial for the treatment of LD patients. A mouse model of Lafora disease (Epm2b-/-) was used to check the putative beneficial effect of different drugs aimed to ameliorate the alterations in glutamatergic transmission and/or neuroinflammation present in the model. Drugs in blue gave a more positive outcome than the rest.
Collapse
Affiliation(s)
- Belén Mollá
- Laboratory of Nutrient Signaling, Institute of Biomedicine of Valencia (CSIC), Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Miguel Heredia
- Laboratory of Nutrient Signaling, Institute of Biomedicine of Valencia (CSIC), Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
| | - Ángela Campos
- Laboratory of Nutrient Signaling, Institute of Biomedicine of Valencia (CSIC), Valencia, Spain
| | - Pascual Sanz
- Laboratory of Nutrient Signaling, Institute of Biomedicine of Valencia (CSIC), Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010 Valencia, Spain
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Jaime Roig 11, 46010 Valencia, Spain
| |
Collapse
|
21
|
Fabisiak T, Patel M. Crosstalk between neuroinflammation and oxidative stress in epilepsy. Front Cell Dev Biol 2022; 10:976953. [PMID: 36035987 PMCID: PMC9399352 DOI: 10.3389/fcell.2022.976953] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022] Open
Abstract
The roles of both neuroinflammation and oxidative stress in the pathophysiology of epilepsy have begun to receive considerable attention in recent years. However, these concepts are predominantly studied as separate entities despite the evidence that neuroinflammatory and redox-based signaling cascades have significant crosstalk. Oxidative post-translational modifications have been demonstrated to directly influence the function of key neuroinflammatory mediators. Neuroinflammation can further be controlled on the transcriptional level as the transcriptional regulators NF-KB and nrf2 are activated by reactive oxygen species. Further, neuroinflammation can induce the increased expression and activity of NADPH oxidase, leading to a highly oxidative environment. These factors additionally influence mitochondria function and the metabolic status of neurons and glia, which are already metabolically stressed in epilepsy. Given the implication of this relationship to disease pathology, this review explores the numerous mechanisms by which neuroinflammation and oxidative stress influence one another in the context of epilepsy. We further examine the efficacy of treatments targeting oxidative stress and redox regulation in animal and human epilepsies in the literature that warrant further investigation. Treatment approaches aimed at rectifying oxidative stress and aberrant redox signaling may enable control of neuroinflammation and improve patient outcomes.
Collapse
|
22
|
Varea O, Guinovart JJ, Duran J. Malin restoration as proof of concept for gene therapy for Lafora disease. Brain Commun 2022; 4:fcac168. [PMID: 35813879 PMCID: PMC9260307 DOI: 10.1093/braincomms/fcac168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/03/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Lafora disease is a fatal neurodegenerative childhood dementia caused by loss-of-function mutations in either the laforin or malin gene. The hallmark of the disease is the accumulation of abnormal glycogen aggregates known as Lafora bodies (LBs) in the brain and other tissues. These aggregates are responsible for the pathological features of the disease. As a monogenic disorder, Lafora disease is a good candidate for gene therapy-based approaches. However, most patients are diagnosed after the appearance of the first symptoms and thus when LBs are already present in the brain. In this context, it was not clear whether the restoration of a normal copy of the defective gene (either laforin or malin) would prove effective. Here we evaluated the effect of restoring malin in a malin-deficient mouse model of Lafora disease as a proof of concept for gene replacement therapy. To this end, we generated a malin-deficient mouse in which malin expression can be induced at a certain time. Our results reveal that malin restoration at an advanced stage of the disease arrests the accumulation of LBs in brain and muscle, induces the degradation of laforin and glycogen synthase bound to the aggregates, and ameliorates neuroinflammation. These results identify malin restoration as the first therapeutic strategy to show effectiveness when applied at advanced stages of Lafora disease.
Collapse
Affiliation(s)
- Olga Varea
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology , Barcelona 08028 , Spain
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology , Barcelona 08028 , Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) , Madrid 28029 , Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona , Barcelona 08028 , Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology , Barcelona 08028 , Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) , Madrid 28029 , Spain
- Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL) , Barcelona 08017 , Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology , Barcelona 08028 , Spain
| |
Collapse
|
23
|
Sinha P, Verma B, Ganesh S. Age-Dependent Reduction in the Expression Levels of Genes Involved in Progressive Myoclonus Epilepsy Correlates with Increased Neuroinflammation and Seizure Susceptibility in Mouse Models. Mol Neurobiol 2022; 59:5532-5548. [PMID: 35732865 DOI: 10.1007/s12035-022-02928-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/14/2022] [Indexed: 11/28/2022]
Abstract
Brain aging is characterized by a gradual decline in cellular homeostatic processes, thereby losing the ability to respond to physiological stress. At the anatomical level, the aged brain is characterized by degenerating neurons, proteinaceous plaques and tangles, intracellular deposition of glycogen, and elevated neuroinflammation. Intriguingly, such age-associated changes are also seen in neurodegenerative disorders suggesting that an accelerated aging process could be one of the contributory factors for the disease phenotype. Amongst these, the genetic forms of progressive myoclonus epilepsy (PME), resulting from loss-of-function mutations in genes, manifest symptoms that are common to age-associated disorders, and genes mutated in PME are involved in the cellular homeostatic processes. Intriguingly, the incidence and/or onset of epileptic seizures are known to increase with age, suggesting that physiological changes in the aged brain might contribute to increased susceptibility to seizures. We, therefore, hypothesized that the expression level of genes implicated in PME might decrease with age, thereby leading to a compromised neuronal response towards physiological stress and hence neuroinflammation in the aging brain. Using mice models, we demonstrate here that the expression level of PME genes shows an inverse correlation with age, neuroinflammation, and compromised heat shock response. We further show that the pharmacological suppression of neuroinflammation ameliorates seizure susceptibility in aged animals as well as in animal models for a PME. Taken together, our results indicate a functional role for the PME genes in normal brain aging and that neuroinflammation could be a major contributory player in susceptibility to seizures.
Collapse
Affiliation(s)
- Priyanka Sinha
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Bhupender Verma
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India. .,Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016, India.
| |
Collapse
|
24
|
Trehalose Treatment in Zebrafish Model of Lafora Disease. Int J Mol Sci 2022; 23:ijms23126874. [PMID: 35743315 PMCID: PMC9224929 DOI: 10.3390/ijms23126874] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 01/18/2023] Open
Abstract
Mutations in the EPM2A gene encoding laforin cause Lafora disease (LD), a progressive myoclonic epilepsy characterized by drug-resistant seizures and progressive neurological impairment. To date, rodents are the only available models for studying LD; however, their use for drug screening is limited by regulatory restrictions and high breeding costs. To investigate the role of laforin loss of function in early neurodevelopment, and to screen for possible new compounds for treating the disorder, we developed a zebrafish model of LD. Our results showed the epm2a−/− zebrafish to be a faithful model of LD, exhibiting the main disease features, namely motor impairment and neuronal hyperexcitability with spontaneous seizures. The model also showed increased inflammatory response and apoptotic death, as well as an altered autophagy pathway that occurs early in development and likely contributes to the disease progression. Early administration of trehalose was found to be effective for rescuing motor impairment and neuronal hyperexcitability associated with seizures. Our study adds a new tool for investigating LD and might help to identify new treatment opportunities.
Collapse
|
25
|
Gentry MS, Markussen KH, Donohue KJ. Two Diseases-One Preclinical Treatment Targeting Glycogen Synthesis. Neurotherapeutics 2022; 19:977-981. [PMID: 35460010 PMCID: PMC9294113 DOI: 10.1007/s13311-022-01240-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2022] [Indexed: 01/30/2023] Open
Affiliation(s)
- Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA.
| | - Kia H Markussen
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Katherine J Donohue
- Department of Molecular and Cellular Biochemistry, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
26
|
Gumusgoz E, Kasiri S, Guisso DR, Wu J, Dear M, Verhalen B, Minassian BA. AAV-Mediated Artificial miRNA Reduces Pathogenic Polyglucosan Bodies and Neuroinflammation in Adult Polyglucosan Body and Lafora Disease Mouse Models. Neurotherapeutics 2022; 19:982-993. [PMID: 35347645 PMCID: PMC9294094 DOI: 10.1007/s13311-022-01218-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 11/28/2022] Open
Abstract
Adult polyglucosan body disease (APBD) and Lafora disease (LD) are autosomal recessive glycogen storage neurological disorders. APBD is caused by mutations in the glycogen branching enzyme (GBE1) gene and is characterized by progressive upper and lower motor neuron dysfunction and premature death. LD is a fatal progressive myoclonus epilepsy caused by loss of function mutations in the EPM2A or EPM2B gene. These clinically distinct neurogenetic diseases share a common pathology. This consists of time-dependent formation, precipitation, and accumulation of an abnormal form of glycogen (polyglucosan) into gradually enlarging inclusions, polyglucosan bodies (PBs) in ever-increasing numbers of neurons and astrocytes. The growth and spread of PBs are followed by astrogliosis, microgliosis, and neurodegeneration. The key defect in polyglucosans is that their glucan branches are longer than those of normal glycogen, which prevents them from remaining in solution. Since the lengths of glycogen branches are determined by the enzyme glycogen synthase, we hypothesized that downregulating this enzyme could prevent or hinder the generation of the pathogenic PBs. Here, we pursued an adeno-associated virus vector (AAV) mediated RNA-interference (RNAi) strategy. This approach resulted in approximately 15% reduction of glycogen synthase mRNA and an approximately 40% reduction of PBs across the brain in the APBD and both LD mouse models. This was accompanied by improvements in early neuroinflammatory markers of disease. This work represents proof of principle toward developing a single lifetime dose therapy for two fatal neurological diseases: APBD and LD. The approach is likely applicable to other severe and common diseases of glycogen storage.
Collapse
Affiliation(s)
- Emrah Gumusgoz
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Sahba Kasiri
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Dikran R Guisso
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Jun Wu
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Matthew Dear
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Brandy Verhalen
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
- Present affiliation: Corteva Agriscience, Johnston, IA, 50131, USA
| | - Berge A Minassian
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA.
| |
Collapse
|
27
|
Pellegrini P, Hervera A, Varea O, Brewer MK, López-Soldado I, Guitart A, Aguilera M, Prats N, del Río JA, Guinovart JJ, Duran J. Lack of p62 Impairs Glycogen Aggregation and Exacerbates Pathology in a Mouse Model of Myoclonic Epilepsy of Lafora. Mol Neurobiol 2021; 59:1214-1229. [PMID: 34962634 PMCID: PMC8857170 DOI: 10.1007/s12035-021-02682-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/04/2021] [Indexed: 01/06/2023]
Abstract
Lafora disease (LD) is a fatal childhood-onset dementia characterized by the extensive accumulation of glycogen aggregates—the so-called Lafora Bodies (LBs)—in several organs. The accumulation of LBs in the brain underlies the neurological phenotype of the disease. LBs are composed of abnormal glycogen and various associated proteins, including p62, an autophagy adaptor that participates in the aggregation and clearance of misfolded proteins. To study the role of p62 in the formation of LBs and its participation in the pathology of LD, we generated a mouse model of the disease (malinKO) lacking p62. Deletion of p62 prevented LB accumulation in skeletal muscle and cardiac tissue. In the brain, the absence of p62 altered LB morphology and increased susceptibility to epilepsy. These results demonstrate that p62 participates in the formation of LBs and suggest that the sequestration of abnormal glycogen into LBs is a protective mechanism through which it reduces the deleterious consequences of its accumulation in the brain.
Collapse
Affiliation(s)
- Pasquale Pellegrini
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Arnau Hervera
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain
| | - Olga Varea
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - M. Kathryn Brewer
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Iliana López-Soldado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
| | - Anna Guitart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Mònica Aguilera
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - Neus Prats
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| | - José Antonio del Río
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain
| | - Joan J. Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Department of Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institut Químic de Sarrià, University Ramon Llull, 08017 Barcelona, Spain
| |
Collapse
|
28
|
Gene expression analysis method integration and co-expression module detection applied to rare glucide metabolism disorders using ExpHunterSuite. Sci Rep 2021; 11:15062. [PMID: 34301987 PMCID: PMC8302605 DOI: 10.1038/s41598-021-94343-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/09/2021] [Indexed: 12/13/2022] Open
Abstract
High-throughput gene expression analysis is widely used. However, analysis is not straightforward. Multiple approaches should be applied and methods to combine their results implemented and investigated. We present methodology for the comprehensive analysis of expression data, including co-expression module detection and result integration via data-fusion, threshold based methods, and a Naïve Bayes classifier trained on simulated data. Application to rare-disease model datasets confirms existing knowledge related to immune cell infiltration and suggest novel hypotheses including the role of calcium channels. Application to simulated and spike-in experiments shows that combining multiple methods using consensus and classifiers leads to optimal results. ExpHunter Suite is implemented as an R/Bioconductor package available from https://bioconductor.org/packages/ExpHunterSuite. It can be applied to model and non-model organisms and can be run modularly in R; it can also be run from the command line, allowing scalability with large datasets. Code and reports for the studies are available from https://github.com/fmjabato/ExpHunterSuiteExamples.
Collapse
|
29
|
Mitra S, Gumusgoz E, Minassian BA. Lafora disease: Current biology and therapeutic approaches. Rev Neurol (Paris) 2021; 178:315-325. [PMID: 34301405 DOI: 10.1016/j.neurol.2021.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/21/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022]
Abstract
The ubiquitin system impacts most cellular processes and is altered in numerous neurodegenerative diseases. However, little is known about its role in neurodegenerative diseases due to disturbances of glycogen metabolism such as Lafora disease (LD). In LD, insufficiently branched and long-chained glycogen forms and precipitates into insoluble polyglucosan bodies (Lafora bodies), which drive neuroinflammation, neurodegeneration and epilepsy. LD is caused by mutations in the gene encoding the glycogen phosphatase laforin or the gene coding for the laforin interacting partner ubiquitin E3 ligase malin. The role of the malin-laforin complex in regulating glycogen structure remains with full of gaps. In this review we bring together the disparate body of data on these two proteins and propose a mechanistic hypothesis of the disease in which malin-laforin's role to monitor and prevent over-elongation of glycogen branch chains, which drive glycogen molecules to precipitate and accumulate into Lafora bodies. We also review proposed connections between Lafora bodies and the ensuing neuroinflammation, neurodegeneration and intractable epilepsy. Finally, we review the exciting activities in developing therapies for Lafora disease based on replacing the missing genes, slowing the enzyme - glycogen synthase - that over-elongates glycogen branches, and introducing enzymes that can digest Lafora bodies. Much more work is needed to fill the gaps in glycogen metabolism in which laforin and malin operate. However, knowledge appears already adequate to advance disease course altering therapies for this catastrophic fatal disease.
Collapse
Affiliation(s)
- S Mitra
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - E Gumusgoz
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - B A Minassian
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
30
|
Canine Lafora Disease: An Unstable Repeat Expansion Disorder. Life (Basel) 2021; 11:life11070689. [PMID: 34357061 PMCID: PMC8304204 DOI: 10.3390/life11070689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 11/17/2022] Open
Abstract
Canine Lafora disease is a recessively inherited, rapidly progressing neurodegenerative disease caused by the accumulation of abnormally constructed insoluble glycogen Lafora bodies in the brain and other tissues due to the loss of NHL repeat containing E3 ubiquitin protein ligase 1 (NHLRC1). Dogs have a dodecamer repeat sequence within the NHLRC1 gene, which is prone to unstable (dynamic) expansion and loss of function. Progressive signs of Lafora disease include hypnic jerks, reflex and spontaneous myoclonus, seizures, vision loss, ataxia and decreased cognitive function. We studied five dogs (one Chihuahua, two French Bulldogs, one Griffon Bruxellois, one mixed breed) with clinical signs associated with canine Lafora disease. Identification of polyglucosan bodies (Lafora bodies) in myocytes supported diagnosis in the French Bulldogs; muscle areas close to the myotendinous junction and the myofascial union segment had the highest yield of inclusions. Postmortem examination of one of the French Bulldogs revealed brain Lafora bodies. Genetic testing for the known canine NHLRC1 mutation confirmed the presence of a homozygous mutation associated with canine Lafora disease. Our results show that Lafora disease extends beyond previous known breeds to the French Bulldog, Griffon Bruxellois and even mixed-breed dogs, emphasizing the likely species-wide nature of this genetic problem. It also establishes these breeds as animal models for the devastating human disease. Genetic testing should be used when designing breeding strategies to determine the frequency of the NHLRC1 mutation in affected breeds. Lafora diseases should be suspected in any older dog presenting with myoclonus, hypnic jerks or photoconvulsions.
Collapse
|
31
|
Mollá B, Heredia M, Sanz P. Modulators of Neuroinflammation Have a Beneficial Effect in a Lafora Disease Mouse Model. Mol Neurobiol 2021; 58:2508-2522. [PMID: 33447969 PMCID: PMC8167455 DOI: 10.1007/s12035-021-02285-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/07/2021] [Indexed: 12/22/2022]
Abstract
Lafora disease (LD; OMIM#274780) is a fatal rare neurodegenerative disorder characterized by generalized epileptic seizures and the presence of polyglucosan inclusions (PGs), called Lafora bodies (LBs), typically in the brain. LD is caused by mutations in two genes EPM2A or EPM2B, which encode respectively laforin, a glucan phosphatase, and malin, an E3-ubiquitin ligase. Much remains unknown about the molecular bases of LD and, unfortunately, appropriate treatment is still missing; therefore patients die within 10 years from the onset of the disease. Recently, we have identified neuroinflammation as one of the initial determinants in LD. In this work, we have investigated anti-inflammatory treatments as potential therapies in LD. With this aim, we have performed a preclinical study in an Epm2b-/- mouse model with propranolol, a β-adrenergic antagonist, and epigallocatechin gallate (EGCG), an antioxidant from green tea extract, both of which displaying additional anti-inflammatory properties. In vivo motor and cognitive behavioral tests and ex vivo histopathological brain analyses were used as parameters to assess the therapeutic potential of propranolol and EGCG. After 2 months of treatment, we observed an improvement not only in attention defects but also in neuronal disorganization, astrogliosis, and microgliosis present in the hippocampus of Epm2b-/- mice. In general, propranolol intervention was more effective than EGCG in preventing the appearance of astrocyte and microglia reactivity. In summary, our results confirm the potential therapeutic effectiveness of the modulators of inflammation as novel treatments in Lafora disease.
Collapse
Affiliation(s)
- Belén Mollá
- Laboratory of Nutrient Signaling, Institute of Biomedicine of Valencia (CSIC), Consejo Superior de Investigaciones Científicas, Jaime Roig 11, 46010, Valencia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010, Valencia, Spain.
| | - Miguel Heredia
- Laboratory of Nutrient Signaling, Institute of Biomedicine of Valencia (CSIC), Consejo Superior de Investigaciones Científicas, Jaime Roig 11, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010, Valencia, Spain
| | - Pascual Sanz
- Laboratory of Nutrient Signaling, Institute of Biomedicine of Valencia (CSIC), Consejo Superior de Investigaciones Científicas, Jaime Roig 11, 46010, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 46010, Valencia, Spain
| |
Collapse
|
32
|
Markussen KH, Macedo JKA, Machío M, Dolce A, Goldberg YP, Vander Kooi CW, Gentry MS. The 6th International Lafora Epilepsy Workshop: Advances in the search for a cure. Epilepsy Behav 2021; 119:107975. [PMID: 33946009 PMCID: PMC8154720 DOI: 10.1016/j.yebeh.2021.107975] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 10/21/2022]
Abstract
Lafora disease (LD) is a fatal childhood dementia with severe epilepsy and also a glycogen storage disease that is caused by recessive mutations in either the EPM2A or EPM2B genes. Aberrant, cytoplasmic carbohydrate aggregates called Lafora bodies (LBs) are both a hallmark and driver of the disease. The 6th International Lafora Epilepsy Workshop was held online due to the pandemic. Nearly 300 clinicians, academic and industry scientists, trainees, NIH representatives, and LD friends and family members participated in the event. Speakers covered aspects of LD including progress towards the clinic, the importance of establishing clinical progression, translational progress with repurposed drugs and additional pre-clinical therapies, and novel discoveries that define foundational LD mechanisms.
Collapse
Affiliation(s)
- Kia H. Markussen
- Department of Molecular and Cellular Biochemistry, Epilepsy and Brain Metabolism Alliance, and Epilepsy Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA
| | - Jessica K. A. Macedo
- Department of Molecular and Cellular Biochemistry, Epilepsy and Brain Metabolism Alliance, and Epilepsy Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - María Machío
- Fundación Jimenez Diaz Hospital, UAM, 28045 Madrid, Spain
| | - Alison Dolce
- Division of Neurology, Department of Pediatrics, University of Texas-Southwestern, Dallas, Texas 75390, USA
| | - Y. Paul Goldberg
- Department of Clinical Development, Ionis Pharmaceuticals, Carlsbad, CA, 92008 USA
| | - Craig W. Vander Kooi
- Department of Molecular and Cellular Biochemistry, Epilepsy and Brain Metabolism Alliance, and Epilepsy Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA,Lafora Epilepsy Cure Initiative (LECI), USA
| | - Matthew S. Gentry
- Department of Molecular and Cellular Biochemistry, Epilepsy and Brain Metabolism Alliance, and Epilepsy Research Center, University of Kentucky College of Medicine, Lexington, Kentucky 40536, USA,Lafora Epilepsy Cure Initiative (LECI), USA
| |
Collapse
|
33
|
Beneficial Effects of Metformin on the Central Nervous System, with a Focus on Epilepsy and Lafora Disease. Int J Mol Sci 2021; 22:ijms22105351. [PMID: 34069559 PMCID: PMC8160983 DOI: 10.3390/ijms22105351] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Metformin is a drug in the family of biguanide compounds that is widely used in the treatment of type 2 diabetes (T2D). Interestingly, the therapeutic potential of metformin expands its prescribed use as an anti-diabetic drug. In this sense, it has been described that metformin administration has beneficial effects on different neurological conditions. In this work, we review the beneficial effects of this drug as a neuroprotective agent in different neurological diseases, with a special focus on epileptic disorders and Lafora disease, a particular type of progressive myoclonus epilepsy. In addition, we review the different proposed mechanisms of action of metformin to understand its function at the neurological level.
Collapse
|
34
|
Ahonen S, Nitschke S, Grossman TR, Kordasiewicz H, Wang P, Zhao X, Guisso DR, Kasiri S, Nitschke F, Minassian BA. Gys1 antisense therapy rescues neuropathological bases of murine Lafora disease. Brain 2021; 144:2985-2993. [PMID: 33993268 DOI: 10.1093/brain/awab194] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/01/2021] [Accepted: 04/25/2021] [Indexed: 11/14/2022] Open
Abstract
Lafora disease is a fatal progressive myoclonus epilepsy. At root, it is due to constant acquisition of branches that are too long in a subgroup of glycogen molecules, leading them to precipitate and accumulate into Lafora bodies, which drive a neuroinflammatory response and neurodegeneration. As a potential therapy, we aimed to downregulate glycogen synthase, the enzyme responsible for glycogen branch elongation, in the disease's mouse models. We synthesized an antisense oligonucleotide (Gys1-ASO) that targets the mRNA of the brain-expressed glycogen synthase 1 gene (Gys1). We administered Gys1-ASO by intracerebroventricular injection and analyzed the pathological hallmarks of Lafora disease, namely glycogen accumulation, Lafora body formation, and neuroinflammation. Gys1-ASO prevented Lafora body formation in young mice that had not yet formed them. In older mice that already exhibited Lafora bodies, Gys1-ASO inhibited further accumulation, markedly preventing large Lafora bodies characteristic of advanced disease. Inhibition of Lafora body formation was associated with prevention of astrogliosis and strong trends towards correction of dysregulated expression of disease immune and neuroinflammatory markers. Lafora disease manifests gradually in previously healthy teenagers. Our work provides proof of principle that an antisense oligonucleotide targeting the GYS1 mRNA could prevent, and halt progression of, this catastrophic epilepsy.
Collapse
Affiliation(s)
- Saija Ahonen
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Silvia Nitschke
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada.,Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tamar R Grossman
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Carlsbad, California, USA
| | - Holly Kordasiewicz
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Carlsbad, California, USA
| | - Peixiang Wang
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Xiaochu Zhao
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Dikran R Guisso
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sahba Kasiri
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Felix Nitschke
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Berge A Minassian
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada.,Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
35
|
TRIM32 and Malin in Neurological and Neuromuscular Rare Diseases. Cells 2021; 10:cells10040820. [PMID: 33917450 PMCID: PMC8067510 DOI: 10.3390/cells10040820] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/01/2021] [Accepted: 04/04/2021] [Indexed: 12/27/2022] Open
Abstract
Tripartite motif (TRIM) proteins are RING E3 ubiquitin ligases defined by a shared domain structure. Several of them are implicated in rare genetic diseases, and mutations in TRIM32 and TRIM-like malin are associated with Limb-Girdle Muscular Dystrophy R8 and Lafora disease, respectively. These two proteins are evolutionary related, share a common ancestor, and both display NHL repeats at their C-terminus. Here, we revmniew the function of these two related E3 ubiquitin ligases discussing their intrinsic and possible common pathophysiological pathways.
Collapse
|
36
|
Duran J, Hervera A, Markussen KH, Varea O, López-Soldado I, Sun RC, Del Río JA, Gentry MS, Guinovart JJ. Astrocytic glycogen accumulation drives the pathophysiology of neurodegeneration in Lafora disease. Brain 2021; 144:2349-2360. [PMID: 33822008 DOI: 10.1093/brain/awab110] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 01/05/2021] [Accepted: 01/14/2021] [Indexed: 11/13/2022] Open
Abstract
The hallmark of Lafora disease, a fatal neurodegenerative disorder, is the accumulation of intracellular glycogen aggregates, called Lafora bodies. Until recently, it was widely believed that brain Lafora bodies were present exclusively in neurons and thus that Lafora disease pathology derived from their accumulation in this cell population. However, recent evidence indicates that Lafora bodies are also present in astrocytes. To define the role of astrocytic Lafora bodies in Lafora disease pathology, we deleted glycogen synthase specifically from astrocytes in a mouse model of the disease (malinKO). Strikingly, blocking glycogen synthesis in astrocytes-thus impeding Lafora bodies accumulation in this cell type-prevented the increase in neurodegeneration markers, autophagy impairment, and metabolic changes characteristic of the malinKO model. Conversely, mice that overaccumulate glycogen in astrocytes showed an increase in these markers. These results unveil the deleterious consequences of the deregulation of glycogen metabolism in astrocytes and change the perspective that Lafora disease is caused solely by alterations in neurons.
Collapse
Affiliation(s)
- Jordi Duran
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, 28029, Spain
| | - Arnau Hervera
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, 28031, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, 08028, Spain.,Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain
| | - Kia H Markussen
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Olga Varea
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Iliana López-Soldado
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Ramon C Sun
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Jose Antonio Del Río
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, 28031, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, 08028, Spain.,Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain
| | - Matthew S Gentry
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Joan J Guinovart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, 28029, Spain.,Department of Biochemistry and Molecular Biomedicine, Universitat de Barcelona, Barcelona, 08028, Spain
| |
Collapse
|
37
|
Gumusgoz E, Guisso DR, Kasiri S, Wu J, Dear M, Verhalen B, Nitschke S, Mitra S, Nitschke F, Minassian BA. Targeting Gys1 with AAV-SaCas9 Decreases Pathogenic Polyglucosan Bodies and Neuroinflammation in Adult Polyglucosan Body and Lafora Disease Mouse Models. Neurotherapeutics 2021; 18:1414-1425. [PMID: 33830476 PMCID: PMC8423949 DOI: 10.1007/s13311-021-01040-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
Many adult and most childhood neurological diseases have a genetic basis. CRISPR/Cas9 biotechnology holds great promise in neurological therapy, pending the clearance of major delivery, efficiency, and specificity hurdles. We applied CRISPR/Cas9 genome editing in its simplest modality, namely inducing gene sequence disruption, to one adult and one pediatric disease. Adult polyglucosan body disease is a neurodegenerative disease resembling amyotrophic lateral sclerosis. Lafora disease is a severe late childhood onset progressive myoclonus epilepsy. The pathogenic insult in both is formation in the brain of glycogen with overlong branches, which precipitates and accumulates into polyglucosan bodies that drive neuroinflammation and neurodegeneration. We packaged Staphylococcus aureus Cas9 and a guide RNA targeting the glycogen synthase gene, Gys1, responsible for brain glycogen branch elongation in AAV9 virus, which we delivered by neonatal intracerebroventricular injection to one mouse model of adult polyglucosan body disease and two mouse models of Lafora disease. This resulted, in all three models, in editing of approximately 17% of Gys1 alleles and a similar extent of reduction of Gys1 mRNA across the brain. The latter led to approximately 50% reductions of GYS1 protein, abnormal glycogen accumulation, and polyglucosan bodies, as well as ameliorations of neuroinflammatory markers in all three models. Our work represents proof of principle for virally delivered CRISPR/Cas9 neurotherapeutics in an adult-onset (adult polyglucosan body) and a childhood-onset (Lafora) neurological diseases.
Collapse
Affiliation(s)
- Emrah Gumusgoz
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Dikran R Guisso
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sahba Kasiri
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jun Wu
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Matthew Dear
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Brandy Verhalen
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Present address: Corteva Agriscience, IA, 50131, Johnston, USA
| | - Silvia Nitschke
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sharmistha Mitra
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Felix Nitschke
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Berge A Minassian
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
38
|
Riva A, Striano P. Reversing Accumulation of Polyglucosan Bodies by Virally Delivered CRISPR/Cas9 Genome Editing. Neurotherapeutics 2021; 18:866-867. [PMID: 33847907 PMCID: PMC8423881 DOI: 10.1007/s13311-021-01054-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2021] [Indexed: 10/21/2022] Open
Affiliation(s)
- Antonella Riva
- IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Pasquale Striano
- IRCCS Istituto Giannina Gaslini, Genova, Italy.
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.
| |
Collapse
|
39
|
Perez-Jimenez E, Viana R, Muñoz-Ballester C, Vendrell-Tornero C, Moll-Diaz R, Garcia-Gimeno MA, Sanz P. Endocytosis of the glutamate transporter 1 is regulated by laforin and malin: Implications in Lafora disease. Glia 2020; 69:1170-1183. [PMID: 33368637 DOI: 10.1002/glia.23956] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/10/2020] [Accepted: 12/10/2020] [Indexed: 01/13/2023]
Abstract
Lafora disease (LD) is a fatal rare type of progressive myoclonus epilepsy that appears during early adolescence. The disease is caused by mutations in EPM2A or EPM2B genes, which encode laforin, a glucan phosphatase, and malin, an E3-ubiquitin ligase, respectively. Although the exact roles of laforin and malin are still not well understood, it is known that they work as a complex in which laforin recruits targets that will be ubiquitinated by malin. Recently, we suggested that the type of epilepsy that accompanies LD could be due to deficiencies in the function of the astrocytic glutamate transporter GLT-1. We described that astrocytes from LD mouse models presented decreased levels of GLT-1 at the plasma membrane, leading to increased levels of glutamate in the brain parenchyma. In this work, we present evidence indicating that in the absence of a functional laforin/malin complex (as in LD cellular models) there is an alteration in the ubiquitination of GLT-1, which could be the cause of the reduction in the levels of GLT-1 at the plasma membrane. On the contrary, overexpression of the laforin/malin complex promotes the retention of GLT-1 at the plasma membrane. This retention may be due to the direct ubiquitination of GLT-1 and/or to an opposite effect of this complex on the dynamics of the Nedd4.2-mediated endocytosis of the transporter. This work, therefore, presents new pieces of evidence on the regulation of GLT-1 by the laforin/malin complex, highlighting its value as a therapeutic target for the amelioration of the type of epilepsy that accompanies LD.
Collapse
Affiliation(s)
- Eva Perez-Jimenez
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | - Rosa Viana
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | - Carmen Muñoz-Ballester
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | - Carlos Vendrell-Tornero
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | - Raquel Moll-Diaz
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| | | | - Pascual Sanz
- Consejo Superior de Investigaciones Científicas, Instituto de Biomedicina de Valencia, Valencia, Spain.,CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Biomedicina de Valencia, Valencia, Spain
| |
Collapse
|
40
|
Nitschke S, Chown EE, Zhao X, Gabrielian S, Petković S, Guisso DR, Perri AM, Wang P, Ahonen S, Nitschke F, Minassian BA. An inducible glycogen synthase-1 knockout halts but does not reverse Lafora disease progression in mice. J Biol Chem 2020; 296:100150. [PMID: 33277363 PMCID: PMC7857511 DOI: 10.1074/jbc.ra120.015773] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/21/2022] Open
Abstract
Malstructured glycogen accumulates over time in Lafora disease (LD) and precipitates into Lafora bodies (LBs), leading to neurodegeneration and intractable fatal epilepsy. Constitutive reduction of glycogen synthase-1 (GYS1) activity prevents murine LD, but the effect of GYS1 reduction later in disease course is unknown. Our goal was to knock out Gys1 in laforin (Epm2a)-deficient LD mice after disease onset to determine whether LD can be halted in midcourse, or even reversed. We generated Epm2a-deficient LD mice with tamoxifen-inducible Cre-mediated Gys1 knockout. Tamoxifen was administered at 4 months and disease progression assessed at 12 months. We verified successful knockout at mRNA and protein levels using droplet digital PCR and Western blots. Glycogen determination and periodic acid-Schiff-diastase staining were used to analyze glycogen and LB accumulation. Immunohistochemistry using astrocytic (glial fibrillary acidic protein) and microglial (ionized calcium-binding adapter molecule 1) markers was performed to investigate neuroinflammation. In the disease-relevant organ, the brain, Gys1 mRNA levels were reduced by 85% and GYS1 protein depleted. Glycogen accumulation was halted at the 4-month level, while LB formation and neuroinflammation were significantly, though incompletely, prevented. Skeletal muscle analysis confirmed that Gys1 knockout inhibits glycogen and LB accumulation. However, tamoxifen-independent Cre recombination precluded determination of disease halting or reversal in this tissue. Our study shows that Gys1 knockdown is a powerful means to prevent LD progression, but this approach did not reduce brain glycogen or LBs to levels below those at the time of intervention. These data suggest that endogenous mechanisms to clear brain LBs are absent or, possibly, compromised in laforin-deficient murine LD.
Collapse
Affiliation(s)
- Silvia Nitschke
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Erin E Chown
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Xiaochu Zhao
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Shoghig Gabrielian
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Sara Petković
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Dikran R Guisso
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ami M Perri
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Peixiang Wang
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Saija Ahonen
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Felix Nitschke
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Berge A Minassian
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
41
|
Suppression of glycogen synthesis as a treatment for Lafora disease: Establishing the window of opportunity. Neurobiol Dis 2020; 147:105173. [PMID: 33171226 DOI: 10.1016/j.nbd.2020.105173] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/09/2020] [Accepted: 11/05/2020] [Indexed: 11/19/2022] Open
Abstract
Lafora disease (LD) is a fatal adolescence-onset neurodegenerative condition. The hallmark of LD is the accumulation of aberrant glycogen aggregates called Lafora bodies (LBs) in the brain and other tissues. Impeding glycogen synthesis from early embryonic stages by genetic suppression of glycogen synthase (MGS) in an animal model of LD prevents LB formation and ultimately the pathological manifestations of LD thereby indicating that LBs are responsible for the pathophysiology of the disease. However, it is not clear whether eliminating glycogen synthesis in an adult animal after LBs have already formed would halt or reverse the progression of LD. Herein we generated a mouse model of LD with inducible MGS suppression. We evaluated the effect of MGS suppression at different time points on LB accumulation as well as on the appearance of neuroinflammation, a pathologic trait of LD models. In the skeletal muscle, MGS suppression in adult LD mice blocked the formation of new LBs and reduced the number of glycogen aggregates. In the brain, early but not late MGS suppression halted the accumulation of LBs. However, the neuroinflammatory response was still present, as shown by the levels of reactive astrocytes, microglia and inflammatory cytokines. Our results confirm that MGS as a promising therapeutic target for LD and highlight the importance of an early diagnosis for effective treatment of the disease.
Collapse
|
42
|
Israelian L, Nitschke S, Wang P, Zhao X, Perri AM, Lee JPY, Verhalen B, Nitschke F, Minassian BA. Ppp1r3d deficiency preferentially inhibits neuronal and cardiac Lafora body formation in a mouse model of the fatal epilepsy Lafora disease. J Neurochem 2020; 157:1897-1910. [PMID: 32892347 DOI: 10.1111/jnc.15176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/27/2020] [Accepted: 08/31/2020] [Indexed: 12/20/2022]
Abstract
Mammalian glycogen chain lengths are subject to complex regulation, including by seven proteins (protein phosphatase-1 regulatory subunit 3, PPP1R3A through PPP1R3G) that target protein phosphatase-1 (PP1) to glycogen to activate the glycogen chain-elongating enzyme glycogen synthase and inactivate the chain-shortening glycogen phosphorylase. Lafora disease is a fatal neurodegenerative epilepsy caused by aggregates of long-chained, and as a result insoluble, glycogen, termed Lafora bodies (LBs). We previously eliminated PPP1R3C from a Lafora disease mouse model and studied the effect on LB formation. In the present work, we eliminate and study the effect of absent PPP1R3D. In the interim, brain cell type levels of all PPP1R3 genes have been published, and brain cell type localization of LBs clarified. Integrating these data we find that PPP1R3C is the major isoform in most tissues including brain. In the brain, PPP1R3C is expressed at 15-fold higher levels than PPP1R3D in astrocytes, the cell type where most LBs form. PPP1R3C deficiency eliminates ~90% of brain LBs. PPP1R3D is quantitatively a minor isoform, but possesses unique MAPK, CaMK2 and 14-3-3 binding domains and appears to have an important functional niche in murine neurons and cardiomyocytes. In neurons, it is expressed equally to PPP1R3C, and its deficiency eliminates ~50% of neuronal LBs. In heart, it is expressed at 25% of PPP1R3C where its deficiency eliminates ~90% of LBs. This work studies the role of a second (PPP1R3D) of seven PP1 subunits that regulate the structure of glycogen, toward better understanding of brain glycogen metabolism generally, and in Lafora disease.
Collapse
Affiliation(s)
- Lori Israelian
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Silvia Nitschke
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada.,Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Peixiang Wang
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Xiaochu Zhao
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Ami M Perri
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Jennifer P Y Lee
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Brandy Verhalen
- Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Felix Nitschke
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada.,Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Berge A Minassian
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada.,Division of Neurology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
43
|
Kim YS, Choi J, Yoon BE. Neuron-Glia Interactions in Neurodevelopmental Disorders. Cells 2020; 9:cells9102176. [PMID: 32992620 PMCID: PMC7601502 DOI: 10.3390/cells9102176] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Recent studies have revealed synaptic dysfunction to be a hallmark of various psychiatric diseases, and that glial cells participate in synapse formation, development, and plasticity. Glial cells contribute to neuroinflammation and synaptic homeostasis, the latter being essential for maintaining the physiological function of the central nervous system (CNS). In particular, glial cells undergo gliotransmission and regulate neuronal activity in tripartite synapses via ion channels (gap junction hemichannel, volume regulated anion channel, and bestrophin-1), receptors (for neurotransmitters and cytokines), or transporters (GLT-1, GLAST, and GATs) that are expressed on glial cell membranes. In this review, we propose that dysfunction in neuron-glia interactions may contribute to the pathogenesis of neurodevelopmental disorders. Understanding the mechanisms of neuron-glia interaction for synapse formation and maturation will contribute to the development of novel therapeutic targets of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yoo Sung Kim
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
| | - Juwon Choi
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
| | - Bo-Eun Yoon
- Department of Molecular Biology, Dankook University, Cheonan 31116, Korea; (Y.S.K.); (J.C.)
- Department of Nanobiomedical science, Dankook University, Cheonan 31116, Korea
- Correspondence: ; Tel.: +82-41-529-6085
| |
Collapse
|
44
|
Neuroinflammation and progressive myoclonus epilepsies: from basic science to therapeutic opportunities. Expert Rev Mol Med 2020; 22:e4. [PMID: 32938505 PMCID: PMC7520540 DOI: 10.1017/erm.2020.5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Progressive myoclonus epilepsies (PMEs) are a group of genetic neurological disorders characterised by the occurrence of epileptic seizures, myoclonus and progressive neurological deterioration including cerebellar involvement and dementia. The primary cause of PMEs is variable and alterations in the corresponding mutated genes determine the progression and severity of the disease. In most cases, they lead to the death of the patient after a period of prolonged disability. PMEs also share poor information on the pathophysiological bases and the lack of a specific treatment. Recent reports suggest that neuroinflammation is a common trait under all these conditions. Here, we review similarities and differences in neuroinflammatory response in several PMEs and discuss the window of opportunity of using anti-inflammatory drugs in the treatment of several of these conditions.
Collapse
|
45
|
Oxidative Stress, a Crossroad Between Rare Diseases and Neurodegeneration. Antioxidants (Basel) 2020; 9:antiox9040313. [PMID: 32326494 PMCID: PMC7222183 DOI: 10.3390/antiox9040313] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/06/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is an imbalance between production and accumulation of oxygen reactive species and/or reactive nitrogen species in cells and tissues, and the capacity of detoxifying these products, using enzymatic and non-enzymatic components, such as glutathione. Oxidative stress plays roles in several pathological processes in the nervous system, such as neurotoxicity, neuroinflammation, ischemic stroke, and neurodegeneration. The concepts of oxidative stress and rare diseases were formulated in the eighties, and since then, the link between them has not stopped growing. The present review aims to expand knowledge in the pathological processes associated with oxidative stress underlying some groups of rare diseases: Friedreich’s ataxia, diseases with neurodegeneration with brain iron accumulation, Charcot-Marie-Tooth as an example of rare neuromuscular disorders, inherited retinal dystrophies, progressive myoclonus epilepsies, and pediatric drug-resistant epilepsies. Despite the discrimination between cause and effect may not be easy on many occasions, all these conditions are Mendelian rare diseases that share oxidative stress as a common factor, and this may represent a potential target for therapies.
Collapse
|