1
|
d’Orsi G, Liantonio A, Imbrici P, Gambacorta N, Dinoi G, Altomare CD, DEFEAT-LD Study Group, Carella M. Empagliflozin Repurposing for Lafora Disease: A Pilot Clinical Trial and Preclinical Investigation of Novel Therapeutic Targets. Methods Protoc 2025; 8:48. [PMID: 40407475 PMCID: PMC12101192 DOI: 10.3390/mps8030048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/24/2025] [Accepted: 05/04/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Lafora disease (LD) is an ultra-rare and fatal neurodegenerative disorder with limited therapeutic options. Current treatments primarily address symptoms, with modest efficacy in halting disease progression, thus highlighting the urgent need for novel therapeutic approaches. Gene therapy, antisense oligonucleotides, and recombinant enzymes have recently been, and still are, under investigation. Drug repurposing may offer a promising approach to identify new, possibly effective, therapies. METHODS This study aims to investigate the conditions for repurposing empagliflozin, an SGLT2 (sodium/glucose cotransporter-2) inhibitor, as a potential treatment for LD and to establish a clinical protocol. Clinical phase: This 12-month prospective observational study will assess the safety and clinical efficacy of empagliflozin in two patients with early to intermediate LD stage. The primary endpoints will include changes in the severity of epilepsy and cognitive function, while the secondary endpoints will assess motor function, global function, and autonomy. Multiple clinical and instrumental evaluations (including MRI and PET with 18F-fluorodeoxyglucose) will be performed before and during treatment. Safety monitoring will include regular clinical assessments and reports of adverse events. Preclinical phase: In silico studies (using both molecular docking calculations and reverse ligand-based screening) and in vitro cell-based assays will allow us to investigate the effects of empagliflozin (and other gliflozins) on some key targets likely implicated in LD pathogenesis, such as GLUT1, GLUT3, glycogen synthase (hGYS), and glycogen phosphorylase (GP), as suggested in the literature and digital platforms for in silico target fishing. RESULTS The expected outcome of this study is twofold, i.e., (i) assessing the safety and tolerability of empagliflozin in LD patients and (ii) gathering preliminary data on its potential efficacy in improving clinical and neurologic features. Additionally, the in silico and in vitro studies may provide new insights into the mechanisms through which empagliflozin may exert its therapeutic effects in LD. CONCLUSION The findings of this study are expected to provide evidence in support of the repurposing of empagliflozin for the treatment of LD.
Collapse
Affiliation(s)
- Giuseppe d’Orsi
- Neurology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Antonella Liantonio
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.L.); (P.I.); (N.G.); (G.D.); (C.D.A.)
| | - Paola Imbrici
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.L.); (P.I.); (N.G.); (G.D.); (C.D.A.)
| | - Nicola Gambacorta
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.L.); (P.I.); (N.G.); (G.D.); (C.D.A.)
| | - Giorgia Dinoi
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.L.); (P.I.); (N.G.); (G.D.); (C.D.A.)
| | - Cosimo Damiano Altomare
- Department of Pharmacy—Drug Sciences, University of Bari “Aldo Moro”, 70121 Bari, Italy; (A.L.); (P.I.); (N.G.); (G.D.); (C.D.A.)
| | | | - Massimo Carella
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Aslam Z, Zubaida B, Khan R, Badshah M, Naeem M. Identification of a pathogenic NHLRC1 variant in a consanguineous Pakistani family affected with severe and rapidly progressive Lafora disease. ACTA EPILEPTOLOGICA 2025; 7:7. [PMID: 40217338 PMCID: PMC11960279 DOI: 10.1186/s42494-024-00193-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/20/2024] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Autosomal recessively inherited progressive myoclonic epilepsy of Lafora, which is also known as Lafora disease, is a fatal neurodegenerative disorder. It affects individuals in late childhood or early adolescence and presents with symptoms of progressive mental and physical deterioration. This disease is caused by pathogenic genetic variants in either of two genes: EPM2A on chromosome 6q24, encoding laforin, or NHLRC1 (EPM2B) on chromosome 6p22, encoding malin. CASE PRESENTATION In this study, we report a clinical and molecular investigation of Lafora disease segregating in a consanguineous Pakistani family. The proband presented with symptoms of rapidly progressive myoclonic epilepsy, but laboratory studies were negative for Lafora bodies and ragged red fibres. Sanger DNA sequencing of the genomic DNA of the proband for EPM2A and NHLRC1 identified a previously reported pathogenic nonsense variant in NHLRC1 (NM_198586.3:c.793C > T), which encodes the E3 ubiquitin ligase called malin. The NHLRC1 variant was found in a homozygous state in the proband, predicting a premature stop codon at position 265 (NP_940988.2:p.Arg265Ter). If the mRNA escaped nonsense-mediated decay, it would result in a truncated protein lacking 130 amino acids, including three NHL (NCL-1, HT2A, LIN-41) repeats. CONCLUSIONS Our study emphasizes the role of molecular genetic testing in patients presenting with symptoms of progressive myoclonic epilepsy.
Collapse
Affiliation(s)
- Zain Aslam
- Medical Genetics Research Laboratory, Department of Biotechnology, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Bibi Zubaida
- Medical Genetics Research Laboratory, Department of Biotechnology, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Ranjha Khan
- Medical Genetics Research Laboratory, Department of Biotechnology, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Mazhar Badshah
- Department of Neurology and Neurophysiology, Pakistan Institute of Medical Sciences, Islamabad, 04485, Pakistan
| | - Muhammad Naeem
- Medical Genetics Research Laboratory, Department of Biotechnology, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| |
Collapse
|
5
|
Muccioli L, Vignatelli L, Tappatà M, Mazzone S, Zenesini C, Armstrong D, Michelucci R, Bisulli F. VAL-1221 for the treatment of patients with Lafora disease: study protocol for a single-arm, open-label clinical trial. BMJ Open 2024; 14:e085062. [PMID: 39424378 PMCID: PMC11492954 DOI: 10.1136/bmjopen-2024-085062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024] Open
Abstract
INTRODUCTION Lafora disease (LD) is an ultrarare fatal progressive myoclonic epilepsy, causing drug-resistant epilepsy, myoclonus and psychomotor deterioration. LD is caused by mutations in EPM2A or NHLRC1, which lead to the accumulation of polyglucosans in the brain and neurodegeneration. There are no approved treatments for LD. VAL-1221 is a fusion protein comprising the Fab portion of a cell-penetrating antibody and recombinant human acid alpha glucosidase, and has demonstrated an ability to clear polyglucosans. We hypothesise that intravenous infusion of VAL-1221 might be able to degrade cerebral polyglucosans and stabilise or improve disease outcomes. The aim of this study is to assess the safety and preliminary efficacy of VAL-1221 in patients with LD. METHODS AND ANALYSIS The study is a phase 2, single-arm, open-label, baseline-controlled clinical trial which will be conducted in a single investigational study centre in Italy, namely the sponsor 'IRCCS Istituto delle Scienze Neurologiche di Bologna-Azienda USL di Bologna'. The study will enrol six genetically confirmed patients with mid- to late-stage LD. The global duration of the study for each participant will be 18 months, including screening period, open-label treatment (12 months) and follow-up period. VAL-1221 20 mg/kg will be administered as an intravenous infusion every week for 3 weeks, then every other week. Patients will undergo full clinical assessments at baseline, at an intermediate and at the end-of-treatment visit. The primary objective is to evaluate the safety. The exploratory efficacy endpoints will be related to epilepsy, neuropsychological and motor functions, global assessment and disease burden, in addition to biomarkers. Statistical analyses will be primarily descriptive. ETHICS AND DISSEMINATION The study protocol was approved by the local ethics committee (number 232-2023-FARM-AUSLBO-23020, 22 March 2023). The results of this study will be disseminated by the investigators through presentations at international scientific conferences and reported in peer-reviewed scientific journals. TRIAL REGISTRATION NUMBER European Union Clinical Trials Register (EudraCT 2023-000185-34).
Collapse
Affiliation(s)
- Lorenzo Muccioli
- IRCCS Istituto Delle Scienze Neurologiche di Bologna - Full member of the ERN EpiCARE, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Vignatelli
- IRCCS Istituto Delle Scienze Neurologiche di Bologna - Full member of the ERN EpiCARE, Bologna, Italy
| | - Maria Tappatà
- IRCCS Istituto Delle Scienze Neurologiche di Bologna - Full member of the ERN EpiCARE, Bologna, Italy
| | - Serena Mazzone
- IRCCS Istituto Delle Scienze Neurologiche di Bologna - Full member of the ERN EpiCARE, Bologna, Italy
| | - Corrado Zenesini
- IRCCS Istituto Delle Scienze Neurologiche di Bologna - Full member of the ERN EpiCARE, Bologna, Italy
| | | | - Roberto Michelucci
- IRCCS Istituto Delle Scienze Neurologiche di Bologna - Full member of the ERN EpiCARE, Bologna, Italy
| | - Francesca Bisulli
- IRCCS Istituto Delle Scienze Neurologiche di Bologna - Full member of the ERN EpiCARE, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
6
|
Viana R, Rubio T, Campos-Rodríguez Á, Sanz P. Glial alterations in the glutamatergic and GABAergic signaling pathways in a mouse model of Lafora disease, a severe form of progressive myoclonus epilepsy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612874. [PMID: 39314331 PMCID: PMC11419120 DOI: 10.1101/2024.09.13.612874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Lafora disease (LD; OMIM#254780) is a rare form of progressive myoclonus epilepsy characterized by the accumulation of insoluble deposits of aberrant glycogen (polyglucosans), named Lafora bodies (LBs), in the brain but also in peripheral tissues. It is assumed that the accumulation of LBs is related to the appearance of the characteristic pathological features of the disease. In mouse models of LD, we and others have reported an increase in the levels of reactive astrocytes and activated microglia, which triggers the expression of the different pro-inflammatory mediators. Recently, we have demonstrated that the TNF and IL-6 inflammatory signaling pathways are the main mediators of the neuroinflammatory phenotype associated with the disease. In this work, we present evidence that the activation of these pathways produces a dysregulation in the levels of different subunits of the excitatory ionotropic glutamatergic receptors (phopho-GluN2B, phospho-GluA2, GluK2) and also an increase in the levels of the GABA transporter GAT1 in the hippocampus of the Epm2b-/- mice. In addition, we present evidence of the presence of activated forms of the Src and Lyn protein kinases in this area. These effects may increase the excitatory glutamatergic signaling and decrease the inhibitory GABAergic tone, leading to hyper-excitability. More importantly, the enhanced production of these subunits occurs in non-neuronal cells such as activated microglia and reactive astrocytes, pointing out a key role of glia in the pathophysiology of LD.
Collapse
|
7
|
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
|
8
|
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
|
9
|
Skurat AV, Segvich DM, Contreras CJ, Hu YC, Hurley TD, DePaoli-Roach AA, Roach PJ. Impaired malin expression and interaction with partner proteins in Lafora disease. J Biol Chem 2024; 300:107271. [PMID: 38588813 PMCID: PMC11063907 DOI: 10.1016/j.jbc.2024.107271] [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: 02/17/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
Lafora disease (LD) is an autosomal recessive myoclonus epilepsy with onset in the teenage years leading to death within a decade of onset. LD is characterized by the overaccumulation of hyperphosphorylated, poorly branched, insoluble, glycogen-like polymers called Lafora bodies. The disease is caused by mutations in either EPM2A, encoding laforin, a dual specificity phosphatase that dephosphorylates glycogen, or EMP2B, encoding malin, an E3-ubiquitin ligase. While glycogen is a widely accepted laforin substrate, substrates for malin have been difficult to identify partly due to the lack of malin antibodies able to detect malin in vivo. Here we describe a mouse model in which the malin gene is modified at the C-terminus to contain the c-myc tag sequence, making an expression of malin-myc readily detectable. Mass spectrometry analyses of immunoprecipitates using c-myc tag antibodies demonstrate that malin interacts with laforin and several glycogen-metabolizing enzymes. To investigate the role of laforin in these interactions we analyzed two additional mouse models: malin-myc/laforin knockout and malin-myc/LaforinCS, where laforin was either absent or the catalytic Cys was genomically mutated to Ser, respectively. The interaction of malin with partner proteins requires laforin but is not dependent on its catalytic activity or the presence of glycogen. Overall, the results demonstrate that laforin and malin form a complex in vivo, which stabilizes malin and enhances interaction with partner proteins to facilitate normal glycogen metabolism. They also provide insights into the development of LD and the rescue of the disease by the catalytically inactive phosphatase.
Collapse
Affiliation(s)
- Alexander V Skurat
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Lafora Epilepsy Cure Initiative, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Dyann M Segvich
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Lafora Epilepsy Cure Initiative, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Christopher J Contreras
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Lafora Epilepsy Cure Initiative, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Yueh-Chiang Hu
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Thomas D Hurley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Lafora Epilepsy Cure Initiative, University of Kentucky College of Medicine, Lexington, Kentucky, USA.
| | - Anna A DePaoli-Roach
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Lafora Epilepsy Cure Initiative, University of Kentucky College of Medicine, Lexington, Kentucky, USA.
| | - Peter J Roach
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA; Lafora Epilepsy Cure Initiative, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
10
|
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
|
11
|
Imbrici P, d'Orsi G, Carella M, Nicolotti O, De Luca A, Altomare CD, Liantonio A. Sodium-glucose cotransporter-2 inhibitors: A potential novel treatment for Lafora disease? Pharmacol Res 2024; 199:107012. [PMID: 38036198 DOI: 10.1016/j.phrs.2023.107012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 11/21/2023] [Indexed: 12/02/2023]
Affiliation(s)
- Paola Imbrici
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy.
| | - Giuseppe d'Orsi
- Neurology Unit, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Massimo Carella
- Division of Medical Genetics, Fondazione IRCCS-Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Orazio Nicolotti
- Department of Pharmacy - Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Annamaria De Luca
- 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.
| |
Collapse
|
12
|
Zafra-Puerta L, Burgos DF, Iglesias-Cabeza N, González-Fernández J, Sánchez-Martín G, Sánchez MP, Serratosa JM. Gene replacement therapy for Lafora disease in the Epm2a -/- mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571636. [PMID: 38168354 PMCID: PMC10760157 DOI: 10.1101/2023.12.14.571636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Lafora disease is a rare and fatal form of progressive myoclonic epilepsy typically occurring early in adolescence. Common symptoms include seizures, dementia, and a progressive neurological decline leading to death within 5-15 years from onset. The disease results from mutations transmitted with autosomal recessive inheritance in the EPM2A gene, encoding laforin, a dual-specificity phosphatase, or the EPM2B gene, encoding malin, an E3-ubiquitin ligase. Laforin has glucan phosphatase activity, is an adapter of enzymes involved in glycogen metabolism, is involved in endoplasmic reticulum-stress and protein clearance, and acts as a tumor suppressor protein. 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 can lead to alterations in this complex, leading to the formation of Lafora bodies that contain abnormal, insoluble, and hyperphosphorylated forms of glycogen called polyglucosans. We used the Epm2a -/- knock-out mouse model of Lafora disease to apply a gene replacement therapy by administering intracerebroventricular injections of a recombinant adeno-associated virus carrying the human EPM2A gene. We evaluated the effects of this treatment by means of neuropathological studies, behavioral tests, video-electroencephalography recording, and proteomic/phosphoproteomic analysis. Gene therapy with recombinant adeno-associated virus containing the EPM2A gene ameliorated neurological and histopathological alterations, reduced epileptic activity and neuronal hyperexcitability, and decreased the formation of Lafora bodies. Differential quantitative proteomics and phosphoproteomics revealed beneficial changes in various molecular pathways altered in Lafora disease. Improvements were observed for up to nine months following a single intracerebroventricular injection. In conclusion, gene replacement therapy with human EPM2A gene in the Epm2a -/- knock-out mice shows promise as a potential treatment for Lafora disease.
Collapse
|
13
|
Ferrari Aggradi CR, Rimoldi M, Romagnoli G, Velardo D, Meneri M, Iacobucci D, Ripolone M, Napoli L, Ciscato P, Moggio M, Comi GP, Ronchi D, Corti S, Abati E. Lafora Disease: A Case Report and Evolving Treatment Advancements. Brain Sci 2023; 13:1679. [PMID: 38137127 PMCID: PMC10742041 DOI: 10.3390/brainsci13121679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/20/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Lafora disease is a rare genetic disorder characterized by a disruption in glycogen metabolism. It manifests as progressive myoclonus epilepsy and cognitive decline during adolescence. Pathognomonic is the presence of abnormal glycogen aggregates that, over time, produce large inclusions (Lafora bodies) in various tissues. This study aims to describe the clinical and histopathological aspects of a novel Lafora disease patient, and to provide an update on the therapeutical advancements for this disorder. A 20-year-old Libyan boy presented with generalized tonic-clonic seizures, sporadic muscular jerks, eyelid spasms, and mental impairment. Electroencephalography showed multiple discharges across both brain hemispheres. Brain magnetic resonance imaging was unremarkable. Muscle biopsy showed increased lipid content and a very mild increase of intermyofibrillar glycogen, without the polyglucosan accumulation typically observed in Lafora bodies. Despite undergoing three lines of antiepileptic treatment, the patient's condition showed minimal to no improvement. We identified the homozygous variant c.137G>A, p.(Cys46Tyr), in the EPM2B/NHLRC1 gene, confirming the diagnosis of Lafora disease. To our knowledge, the presence of lipid aggregates without Lafora bodies is atypical. Lafora disease should be considered during the differential diagnosis of progressive, myoclonic, and refractory epilepsies in both children and young adults, especially when accompanied by cognitive decline. Although there are no effective therapies yet, the development of promising new strategies prompts the need for an early and accurate diagnosis.
Collapse
Affiliation(s)
- Carola Rita Ferrari Aggradi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (C.R.F.A.); (G.R.); (M.M.); (G.P.C.); (D.R.)
| | - Martina Rimoldi
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.R.); (D.V.); (M.R.); (P.C.); (M.M.)
- Medical Genetics Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Gloria Romagnoli
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (C.R.F.A.); (G.R.); (M.M.); (G.P.C.); (D.R.)
| | - Daniele Velardo
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.R.); (D.V.); (M.R.); (P.C.); (M.M.)
| | - Megi Meneri
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (C.R.F.A.); (G.R.); (M.M.); (G.P.C.); (D.R.)
- Stroke Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Davide Iacobucci
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Michela Ripolone
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.R.); (D.V.); (M.R.); (P.C.); (M.M.)
| | - Laura Napoli
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.R.); (D.V.); (M.R.); (P.C.); (M.M.)
| | - Patrizia Ciscato
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.R.); (D.V.); (M.R.); (P.C.); (M.M.)
| | - Maurizio Moggio
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.R.); (D.V.); (M.R.); (P.C.); (M.M.)
| | - Giacomo Pietro Comi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (C.R.F.A.); (G.R.); (M.M.); (G.P.C.); (D.R.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Dario Ronchi
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (C.R.F.A.); (G.R.); (M.M.); (G.P.C.); (D.R.)
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefania Corti
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (C.R.F.A.); (G.R.); (M.M.); (G.P.C.); (D.R.)
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.R.); (D.V.); (M.R.); (P.C.); (M.M.)
| | - Elena Abati
- Dino Ferrari Centre, Department of Pathophysiology and Transplantation (DEPT), University of Milan, 20122 Milan, Italy; (C.R.F.A.); (G.R.); (M.M.); (G.P.C.); (D.R.)
| |
Collapse
|
14
|
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
|
15
|
Cameron JM, Ellis CA, Berkovic SF, for the ILAE Genetics Commission. ILAE Genetics Literacy series: Progressive myoclonus epilepsies. Epileptic Disord 2023; 25:670-680. [PMID: 37616028 PMCID: PMC10947580 DOI: 10.1002/epd2.20152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/21/2023] [Accepted: 08/19/2023] [Indexed: 08/25/2023]
Abstract
Progressive Myoclonus Epilepsy (PME) is a rare epilepsy syndrome characterized by the development of progressively worsening myoclonus, ataxia, and seizures. A molecular diagnosis can now be established in approximately 80% of individuals with PME. Almost fifty genetic causes of PME have now been established, although some remain extremely rare. Herein, we provide a review of clinical phenotypes and genotypes of the more commonly encountered PMEs. Using an illustrative case example, we describe appropriate clinical investigation and therapeutic strategies to guide the management of this often relentlessly progressive and devastating epilepsy syndrome. This manuscript in the Genetic Literacy series maps to Learning Objective 1.2 of the ILAE Curriculum for Epileptology (Epileptic Disord. 2019;21:129).
Collapse
Affiliation(s)
- Jillian M. Cameron
- Epilepsy Research Centre, Department of MedicineUniversity of MelbourneAustin HealthMelbourneVictoriaAustralia
| | - Colin A. Ellis
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Samuel F. Berkovic
- Epilepsy Research Centre, Department of MedicineUniversity of MelbourneAustin HealthMelbourneVictoriaAustralia
| | | |
Collapse
|
16
|
Pondrelli F, Minardi R, Muccioli L, Zenesini C, Vignatelli L, Licchetta L, Mostacci B, Tinuper P, Vander Kooi CW, Gentry MS, Bisulli F. Prognostic value of pathogenic variants in Lafora Disease: systematic review and meta-analysis of patient-level data. Orphanet J Rare Dis 2023; 18:263. [PMID: 37658439 PMCID: PMC10474773 DOI: 10.1186/s13023-023-02880-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/24/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Lafora disease (LD) is a fatal form of progressive myoclonic epilepsy caused by biallelic pathogenic variants in EPM2A or NHLRC1. With a few exceptions, the influence of genetic factors on disease progression has yet to be confirmed. We present a systematic review and meta-analysis of the known pathogenic variants to identify genotype-phenotype correlations. METHODS We collected all reported cases with genetically-confirmed LD containing data on disease history. Pathogenic variants were classified into missense (MS) and protein-truncating (PT). Three genotype classes were defined according to the combination of the variants: MS/MS, MS/PT, and PT/PT. Time-to-event analysis was performed to evaluate survival and loss of autonomy. RESULTS 250 cases described in 70 articles were included. The mutated gene was NHLRC1 in 56% and EPM2A in 44% of cases. 114 pathogenic variants (67 EPM2A; 47 NHLRC1) were identified. The NHLRC1 genotype PT/PT was associated with shorter survival [HR 2.88; 95% CI 1.23-6.78] and a trend of higher probability of loss of autonomy [HR 2.03, 95% CI 0.75-5.56] at the multivariable Cox regression analysis. The population carrying the homozygous p.Asp146Asn variant of NHLRC1 genotype was confirmed to have a more favourable prognosis in terms of disease duration. CONCLUSIONS This study demonstrates the existence of prognostic genetic factors in LD, namely the genotype defined according to the functional impact of the pathogenic variants. Although the reasons why NHLRC1 genotype PT/PT is associated with a poorer prognosis have yet to be fully elucidated, it may be speculated that malin plays a pivotal role in LD pathogenesis.
Collapse
Affiliation(s)
- Federica Pondrelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Raffaella Minardi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Full Member of the European Reference Network for Rare and Complex Epilepsies (EpiCARE), Bologna, Italy
| | - Lorenzo Muccioli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.
| | - Corrado Zenesini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Full Member of the European Reference Network for Rare and Complex Epilepsies (EpiCARE), Bologna, Italy
| | - Luca Vignatelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Full Member of the European Reference Network for Rare and Complex Epilepsies (EpiCARE), Bologna, Italy
| | - Laura Licchetta
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Full Member of the European Reference Network for Rare and Complex Epilepsies (EpiCARE), Bologna, Italy
| | - Barbara Mostacci
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Full Member of the European Reference Network for Rare and Complex Epilepsies (EpiCARE), Bologna, Italy
| | - Paolo Tinuper
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Craig W Vander Kooi
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, USA
| | - Matthew S Gentry
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, USA
| | - Francesca Bisulli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Full Member of the European Reference Network for Rare and Complex Epilepsies (EpiCARE), Bologna, Italy
| |
Collapse
|
17
|
d'Orsi G, Farolfi A, Muccioli L, Palumbo O, Palumbo P, Modoni S, Allegri V, Garibotto V, Di Claudio MT, Di Muro E, Benvenuto M, Bisulli F, Carella M. Association of CSF and PET markers of neurodegeneration with electroclinical progression in Lafora disease. Front Neurol 2023; 14:1202971. [PMID: 37448753 PMCID: PMC10337130 DOI: 10.3389/fneur.2023.1202971] [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: 04/09/2023] [Accepted: 06/05/2023] [Indexed: 07/15/2023] Open
Abstract
Purpose To evaluate the electro-clinical features in association with laboratory and instrumental correlates of neurodegeneration to detect the progression of Lafora disease (LD). Methods We investigated the electro-clinical longitudinal data and CSF Aβ42, p-tau181 and t-tauAg, amyloid, and 18F-FDG PET of five unrelated LD families. Results Three progressive electro-clinical stages were identified. The early phase was characterized by rare, generalized tonic-clonic and focal visual seizures, followed by the occurrence of myoclonus after a period ranging from 2 to 12 months. The intermediate stage, usually occurring 2 years after the onset of epilepsy, is characterized by a worsening of epilepsy and myoclonus associated with progressive dementia and cerebellar signs. Finally, the late stage, evolving after a mean period of 7 ± 1.41 years from the onset of the disease, was characterized by gait ataxia resulting in bedriddenness, severe dementia, daily/pluri-daily myoclonus, drug-resistant epilepsy, clusters of seizures or status epilepticus, and medical complications. Amyloid (CSF Aβ42, amyloid PET) and neurodegenerative (CSF p-tau181 and t-tauAg, FDG-PET) biomarkers indicate a pattern of cognitive impairment of the non-Alzheimer's disease type. A total of 80% of the LD patients showed more severe hypometabolism in the second FDG-PET scan compared to the first scan performed in a lower phase; the lateral temporal lobe and the thalamus hypometabolism were associated with the presence of intermediate or late phase. Conclusions Three electroclinical and 18F-FDG PET evolutive stages are useful biomarkers for the progression of LD and could help to evaluate the efficacy of new disease-modifying treatments. The combination of traditional CSF biomarkers improves the diagnostic accuracy of cognitive decline in LD patients, indicating a cognitive impairment of the non-Alzheimer's disease type.
Collapse
Affiliation(s)
- Giuseppe d'Orsi
- Neurology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Andrea Farolfi
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Lorenzo Muccioli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Orazio Palumbo
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Pietro Palumbo
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Sergio Modoni
- Nuclear Medicine Department, Policlinico Riuniti, Foggia, Italy
| | - Vincenzo Allegri
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Valentina Garibotto
- Diagnostic Department, University Hospitals of Geneva, CIBM Center of Biomedical Imaging and NIMTLab, University of Geneva, Geneva, Switzerland
| | - Maria Teresa Di Claudio
- Neurology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Ester Di Muro
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Mario Benvenuto
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Francesca Bisulli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Epilepsy Center (Full Member of the European Reference Network EpiCARE), Bologna, Italy
| | - Massimo Carella
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
Burgos DF, Machío-Castello M, Iglesias-Cabeza N, Giráldez BG, González-Fernández J, Sánchez-Martín G, Sánchez MP, Serratosa JM. Early Treatment with Metformin Improves Neurological Outcomes in Lafora Disease. Neurotherapeutics 2023; 20:230-244. [PMID: 36303102 PMCID: PMC10119355 DOI: 10.1007/s13311-022-01304-w] [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] [Accepted: 09/14/2022] [Indexed: 10/31/2022] Open
Abstract
Lafora disease is a fatal form of progressive myoclonic epilepsy caused by mutations in the EPM2A or NHLRC1/EPM2B genes that usually appears during adolescence. The Epm2a-/- and Epm2b-/- knock-out mouse models of the disease develop behavioral and neurological alterations similar to those observed in patients. The aim of this work is to analyze whether early treatment with metformin (from conception to adulthood) ameliorates the formation of Lafora bodies and improves the behavioral and neurological outcomes observed with late treatment (during 2 months at 10 months of age). We also evaluated the benefits of metformin in patients with Lafora disease. To assess neurological improvements due to metformin administration in the two mouse models, we evaluated the effects on pentylenetetrazol sensitivity, posturing, motor coordination and activity, and memory. We also analyzed the effects on Lafora bodies, neurodegeneration, and astrogliosis. Furthermore, we conducted a follow-up study of an initial cohort of 18 patients with Lafora disease, 8 treated with metformin and 10 untreated. Our results indicate that early metformin was more effective than late metformin in Lafora disease mouse models improving neurological alterations of both models such as neuronal hyperexcitability, motor and memory alterations, neurodegeneration, and astrogliosis and decreasing the formation of Lafora bodies. Moreover, patients receiving metformin had a slower progression of the disease. Overall, early treatment improves the outcome seen with late metformin treatment in the two knock-out mouse models of Lafora disease. Metformin-treated patients exhibited an ameliorated course of the disease with slower deterioration of their daily living activities.
Collapse
Affiliation(s)
- Daniel F Burgos
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma de Madrid University (IIS-FJD, UAM), Av. Reyes Católicos, 2, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
- PhD Program in Neuroscience, Autonoma de Madrid University-Cajal Institute, 28029, Madrid, Spain
| | - María Machío-Castello
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma de Madrid University (IIS-FJD, UAM), Av. Reyes Católicos, 2, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Nerea Iglesias-Cabeza
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma de Madrid University (IIS-FJD, UAM), Av. Reyes Católicos, 2, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Beatriz G Giráldez
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma de Madrid University (IIS-FJD, UAM), Av. Reyes Católicos, 2, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Juan González-Fernández
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma de Madrid University (IIS-FJD, UAM), Av. Reyes Católicos, 2, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
- Department of Parasitology, School of Pharmacy, Complutense de Madrid University, 28040, Madrid, Spain
| | - Gema Sánchez-Martín
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma de Madrid University (IIS-FJD, UAM), Av. Reyes Católicos, 2, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029, Madrid, Spain
| | - Marina P Sánchez
- Laboratory of Neurology, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Autónoma de Madrid University (IIS-FJD, UAM), Av. Reyes Católicos, 2, 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, Autónoma de Madrid University (IIS-FJD, UAM), Av. Reyes Católicos, 2, 28040, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029, Madrid, Spain.
| |
Collapse
|
25
|
Guo M, Chen S, Lao J, Liang J, Chen H, Tong J, Huang Y, Jia D, Li Q. 3BDO Alleviates Seizures and Improves Cognitive Function by Regulating Autophagy in Pentylenetetrazol (PTZ)-Kindled Epileptic Mice Model. Neurochem Res 2022; 47:3777-3791. [PMID: 36243819 DOI: 10.1007/s11064-022-03778-8] [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: 08/08/2022] [Revised: 09/24/2022] [Accepted: 09/29/2022] [Indexed: 12/13/2022]
Abstract
3-benzyl-5-((2-nitrophenoxy) methyl)-dihydrofuran-2(3 H)-one (3BDO) is a mTOR agonist that inhibits autophagy. The main purpose of this study is to investigate the effects of 3BDO on seizure and cognitive function by autophagy regulation in pentylenetetrazol (PTZ)-kindled epileptic mice model. The PTZ-kindled epileptic mice model was used in study. The behavioral changes and electroencephalogram (EEG) of the mice in each group were observed. The cognitive functions were tested by Morris water maze test. The loss of hippocampal neurons was detected by hematoxylin-eosin (HE) staining and immunofluorescence analysis. Immunohistochemistry, western blot and q-PCR were employed to detect the expression of autophagy-related proteins and mTOR in the hippocampus and cortex. Less seizures, increased hippocampal neurons and reduced astrocytes of hippocampus were observed in the 3BDO-treated epileptic mice than in the PTZ-kindled epileptic mice. Morris water maze test results showed that 3BDO significantly improved the cognitive function of the PTZ-kindled epileptic mice. Western blot analyses and q-PCR revealed that 3BDO inhibited the expression of LC3, Beclin-1, Atg5, Atg7 and p-ULK1/ULK1, but increased that of p-mTOR/mTOR, p-P70S6K/P70S6K in the hippocampus and temporal lobe cortex of epileptic mice. Immunohistochemistry and immunofluorescence also showed 3BDO inhibited the LC3 expression and increased the mTOR expression in the hippocampus of epileptic mice. In addition, the autophagy activator EN6 reversed the decrease in the 3BDO-induced autophagy and aggravated the seizures and cognitive dysfunction in the epileptic mice. 3BDO regulates autophagy by activating the mTOR signaling pathway in PTZ-kindled epileptic mice model, thereby alleviating hippocampus neuronal loss and astrocytes proliferation, reducing seizures and effectively improving cognitive function. Therefore, 3BDO may have potential value in the treatment of epilepsy.
Collapse
Affiliation(s)
- Meiwen Guo
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Shuang Chen
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jitong Lao
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiantang Liang
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Hao Chen
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Jingyi Tong
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | | | - Dandan Jia
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China.
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.
| | - Qifu Li
- Department of Neurology, the First Affiliated Hospital of Hainan Medical University, Haikou, China.
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.
| |
Collapse
|
26
|
d'Orsi G, Di Claudio MT, Palumbo O, Carella M. Electro-clinical features and management of the late stage of Lafora disease. Front Neurol 2022; 13:969297. [PMID: 36277909 PMCID: PMC9580008 DOI: 10.3389/fneur.2022.969297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose The aim of this study was to elucidate the electro-clinical features and management of the late stage of Lafora disease (LD). Methods We investigated the electro-clinical data and medical complications of three LD patients with mutations in EPM2A and two in NHLRC1 genes during the LD late stage. Results The late stage emerged after a mean period of 7 ± 1.41 years from the onset of the disease. All patients developed gait ataxia becoming bedbound with severe dementia. Pluri-monthly and drug-resistant myoclonic seizures, and myoclonic absence and tonic–clonic seizures were associated with daily/pluri-daily myoclonus, while the EEG/polygraphic findings showed diffusely slow activity with epileptiform abnormalities, often correlated with myoclonic jerks. Seizure emergencies with motor cluster/status epilepticus and medical complications dominated the clinical picture. In particular, video-EEG/polygraphic recordings disclosed status epilepticus with prominent motor symptoms of different subtypes refractory to IV new anti-seizure medications and responsive in 75% of cases to IV phenytoin. The main complications were dysphagia, aspiration pneumonia, acute respiratory failure, sepsis, immobility, and spasticity with bedsores. A coordinated and multidisciplinary management of the three patients with EPM2A mutations has demonstrated a reduction in seizure emergencies, medical complications and days of hospitalization, and a prolongation of the years of disease compared to the two patients with NHLRC1 mutations. Conclusion Status epilepticus with prominent motor symptoms of different subtypes, often responsive to IV phenytoin, and multiple medical complications characterize the LD late stage. An effective management requires a multidisciplinary medical and nursing team, coordinated by an epileptologist with the aim of reducing seizure emergencies and medical complications.
Collapse
Affiliation(s)
- Giuseppe d'Orsi
- Neurology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
- Department of Neurological Sciences, Policlinico Riuniti, University Neurology Unit, Foggia, Italy
- *Correspondence: Giuseppe d'Orsi
| | - Maria Teresa Di Claudio
- Neurology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
- Department of Neurological Sciences, Policlinico Riuniti, University Neurology Unit, Foggia, Italy
| | - Orazio Palumbo
- Division of Medical Genetics, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Massimo Carella
- Division of Medical Genetics, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| |
Collapse
|
27
|
Lafora Disease and Alpha-Synucleinopathy in Two Adult Free-Ranging Moose (Alces alces) Presenting with Signs of Blindness and Circling. Animals (Basel) 2022; 12:ani12131633. [PMID: 35804532 PMCID: PMC9264765 DOI: 10.3390/ani12131633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Reports of behavioral signs, such as blindness and circling in free-ranging moose from different parts of the world, have spurred comprehensive pathological investigation to find the causes of the disease that have clinical relevance. In this case study, brains collected from two adult free-ranging moose (Alces alces) cows that were seemingly blind and found walking in circles were examined by light and electron microscopy with further ancillary testing. Here, we report for the first time Lafora disease and alpha-synucleinopathy in two wild free-ranging moose cows who presented with abnormal behavior and blindness, with similar neuronal polyglucosan body (PGB) accumulations identified in humans and other animals. Microscopic analysis of the hippocampus of brain revealed inclusion bodies resembling PGBs (Lafora disease) in the neurons with ultrastructural findings of aggregates of branching filaments, consistent with polyglucosan bodies. Furthermore, α-synuclein immunopositivity was noted in the hippocampus, with accumulations of small granules ultrastructurally distinct from PGBs and morphologically compatible with alpha-synucleinopathy (Lewy body). The apparent blindness found in these moose could be related to an injury associated with secondary bacterial invasion; however, an accumulation of neurotoxicants (PGBs and α-synucleins) in retinal ganglion cells could also be the cause. Lafora disease and alpha-synucleinopathy were considered in the differential diagnosis of the young adult moose who presented with signs of blindness and behavioral signs such as circling. Abstract Lafora disease is an autosomal recessive glycogen-storage disorder resulting from an accumulation of toxic polyglucosan bodies (PGBs) in the central nervous system, which causes behavioral and neurologic symptoms in humans and other animals. In this case study, brains collected from two young adult free-ranging moose (Alces alces) cows that were seemingly blind and found walking in circles were examined by light and electron microscopy. Microscopic analysis of the hippocampus of the brain revealed inclusion bodies resembling PGBs in the neuronal perikaryon, neuronal processes, and neuropil. These round inclusions measuring up to 30 microns in diameter were predominantly confined to the hippocampus region of the brain in both animals. The inclusions tested α-synuclein-negative by immunohistochemistry, α-synuclein-positive with PAS, GMS, and Bielschowsky’s staining; and diastase-resistant with central basophilic cores and faintly radiating peripheral lines. Ultrastructural examination of the affected areas of the hippocampus showed non-membrane-bound aggregates of asymmetrically branching filaments that bifurcated regularly, consistent with PGBs in both animals. Additionally, α-synuclein immunopositivity was noted in the different regions of the hippocampus with accumulations of small granules ultrastructurally distinct from PGBs and morphologically compatible with alpha-synucleinopathy (Lewy body). The apparent blindness found in these moose could be related to an injury associated with secondary bacterial invasion; however, an accumulation of neurotoxicants (PGBs and α-synuclein) in retinal ganglions cells could also be the cause. This is the first report demonstrating Lafora disease with concurrent alpha-synucleinopathy (Lewy body neuropathy) in a non-domesticated animal.
Collapse
|
28
|
Goodspeed K, Bailey RM, Prasad S, Sadhu C, Cardenas JA, Holmay M, Bilder DA, Minassian BA. Gene Therapy: Novel Approaches to Targeting Monogenic Epilepsies. Front Neurol 2022; 13:805007. [PMID: 35847198 PMCID: PMC9284605 DOI: 10.3389/fneur.2022.805007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 04/20/2022] [Indexed: 11/18/2022] Open
Abstract
Genetic epilepsies are a spectrum of disorders characterized by spontaneous and recurrent seizures that can arise from an array of inherited or de novo genetic variants and disrupt normal brain development or neuronal connectivity and function. Genetically determined epilepsies, many of which are due to monogenic pathogenic variants, can result in early mortality and may present in isolation or be accompanied by neurodevelopmental disability. Despite the availability of more than 20 antiseizure medications, many patients with epilepsy fail to achieve seizure control with current therapies. Patients with refractory epilepsy—particularly of childhood onset—experience increased risk for severe disability and premature death. Further, available medications inadequately address the comorbid developmental disability. The advent of next-generation gene sequencing has uncovered genetic etiologies and revolutionized diagnostic practices for many epilepsies. Advances in the field of gene therapy also present the opportunity to address the underlying mechanism of monogenic epilepsies, many of which have only recently been described due to advances in precision medicine and biology. To bring precision medicine and genetic therapies closer to clinical applications, experimental animal models are needed that replicate human disease and reflect the complexities of these disorders. Additionally, identifying and characterizing clinical phenotypes, natural disease course, and meaningful outcome measures from epileptic and neurodevelopmental perspectives are necessary to evaluate therapies in clinical studies. Here, we discuss the range of genetically determined epilepsies, the existing challenges to effective clinical management, and the potential role gene therapy may play in transforming treatment options available for these conditions.
Collapse
Affiliation(s)
- Kimberly Goodspeed
- Division of Child Neurology, Department of Pediatrics, University of Texas Southwestern, Dallas, TX, United States
| | - Rachel M. Bailey
- Division of Child Neurology, Department of Pediatrics, University of Texas Southwestern, Dallas, TX, United States
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern, Dallas, TX, United States
| | - Suyash Prasad
- Department of Research and Development, Taysha Gene Therapies, Dallas, TX, United States
| | - Chanchal Sadhu
- Department of Research and Development, Taysha Gene Therapies, Dallas, TX, United States
| | - Jessica A. Cardenas
- Department of Research and Development, Taysha Gene Therapies, Dallas, TX, United States
| | - Mary Holmay
- Department of Research and Development, Taysha Gene Therapies, Dallas, TX, United States
| | - Deborah A. Bilder
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Huntsman Mental Health Institute, University of Utah, Salt Lake City, UT, United States
| | - Berge A. Minassian
- Division of Child Neurology, Department of Pediatrics, University of Texas Southwestern, Dallas, TX, United States
- *Correspondence: Berge A. Minassian
| |
Collapse
|
29
|
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
|
30
|
Liang T, Wu J, Chen H, Qian J, Xu Z. Novel mutation of EPM2A causes progressive myoclonic epilepsy: a case report. Neurol Sci 2022; 43:3467-3471. [DOI: 10.1007/s10072-022-05986-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/25/2022] [Indexed: 11/29/2022]
|
31
|
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
|