1
|
Sabater-Arcis M, Moreno N, Sevilla T, Perez Alonso M, Bargiela A, Artero R. Msi2 enhances muscle dysfunction in a myotonic dystrophy type 1 mouse model. Biomed J 2024; 47:100667. [PMID: 37797921 PMCID: PMC11340596 DOI: 10.1016/j.bj.2023.100667] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/19/2023] [Accepted: 09/29/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Myotonic dystrophy type 1 (DM1) is a rare neuromuscular disease caused by a CTG repeat expansion in the 3' untranslated region of the DM1 protein kinase gene. Characteristic degenerative muscle symptoms include myotonia, atrophy, and weakness. We previously proposed an Musashi homolog 2 (MSI2)>miR-7>autophagy axis whereby MSI2 overexpression repressed miR-7 biogenesis that subsequently de-repressed muscle catabolism through excessive autophagy. Because the DM1 HSALR mouse model expressing expanded CUG repeats shows weak muscle-wasting phenotypes, we hypothesized that MSI2 overexpression was sufficient to promote muscle dysfunction in vivo. METHODS By means of recombinant AAV murine MSI2 was overexpressed in neonates HSALR mice skeletal muscle to induce DM1-like phenotypes. RESULTS Sustained overexpression of the murine MSI2 protein in HSALR neonates induced autophagic flux and expression of critical autophagy proteins, increased central nuclei and reduced myofibers area, and weakened muscle strength. Importantly, these changes were independent of MBNL1, MBNL2, and Celf1 protein levels, which remained unchanged upon Msi2 overexpression. CONCLUSIONS Globally, molecular, histological, and functional data from these experiments in the HSALR mouse model confirms the pathological role of MSI2 expression levels as an atrophy-associated component that impacts the characteristic muscle dysfunction symptoms in DM1 patients.
Collapse
Affiliation(s)
- Maria Sabater-Arcis
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Nerea Moreno
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Teresa Sevilla
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe, Valencia, Spain
- Centre for Biomedical Network Research on Rare Diseases, Valencia, Spain
- Department of Medicine, University of Valencia, Valencia, Spain
| | - Manuel Perez Alonso
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| | - Ariadna Bargiela
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe, Valencia, Spain
| | - Ruben Artero
- Human Translational Genomics Group, University Institute for Biotechnology and Biomedicine, Valencia, Spain
- INCLIVA Biomedical Research Institute, Valencia, Spain
| |
Collapse
|
2
|
Costa A, Cruz AC, Martins F, Rebelo S. Protein Phosphorylation Alterations in Myotonic Dystrophy Type 1: A Systematic Review. Int J Mol Sci 2023; 24:ijms24043091. [PMID: 36834509 PMCID: PMC9965115 DOI: 10.3390/ijms24043091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/21/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Among the most common muscular dystrophies in adults is Myotonic Dystrophy type 1 (DM1), an autosomal dominant disorder characterized by myotonia, muscle wasting and weakness, and multisystemic dysfunctions. This disorder is caused by an abnormal expansion of the CTG triplet at the DMPK gene that, when transcribed to expanded mRNA, can lead to RNA toxic gain of function, alternative splicing impairments, and dysfunction of different signaling pathways, many regulated by protein phosphorylation. In order to deeply characterize the protein phosphorylation alterations in DM1, a systematic review was conducted through PubMed and Web of Science databases. From a total of 962 articles screened, 41 were included for qualitative analysis, where we retrieved information about total and phosphorylated levels of protein kinases, protein phosphatases, and phosphoproteins in DM1 human samples and animal and cell models. Twenty-nine kinases, 3 phosphatases, and 17 phosphoproteins were reported altered in DM1. Signaling pathways that regulate cell functions such as glucose metabolism, cell cycle, myogenesis, and apoptosis were impaired, as seen by significant alterations to pathways such as AKT/mTOR, MEK/ERK, PKC/CUGBP1, AMPK, and others in DM1 samples. This explains the complexity of DM1 and its different manifestations and symptoms, such as increased insulin resistance and cancer risk. Further studies can be done to complement and explore in detail specific pathways and how their regulation is altered in DM1, to find what key phosphorylation alterations are responsible for these manifestations, and ultimately to find therapeutic targets for future treatments.
Collapse
|
3
|
Kuntawala DH, Martins F, Vitorino R, Rebelo S. Automatic Text-Mining Approach to Identify Molecular Target Candidates Associated with Metabolic Processes for Myotonic Dystrophy Type 1. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2283. [PMID: 36767649 PMCID: PMC9915907 DOI: 10.3390/ijerph20032283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 06/18/2023]
Abstract
Myotonic dystrophy type 1 (DM1) is an autosomal dominant hereditary disease caused by abnormal expansion of unstable CTG repeats in the 3' untranslated region of the myotonic dystrophy protein kinase (DMPK) gene. This disease mainly affects skeletal muscle, resulting in myotonia, progressive distal muscle weakness, and atrophy, but also affects other tissues and systems, such as the heart and central nervous system. Despite some studies reporting therapeutic strategies for DM1, many issues remain unsolved, such as the contribution of metabolic and mitochondrial dysfunctions to DM1 pathogenesis. Therefore, it is crucial to identify molecular target candidates associated with metabolic processes for DM1. In this study, resorting to a bibliometric analysis, articles combining DM1, and metabolic/metabolism terms were identified and further analyzed using an unbiased strategy of automatic text mining with VOSviewer software. A list of candidate molecular targets for DM1 associated with metabolic/metabolism was generated and compared with genes previously associated with DM1 in the DisGeNET database. Furthermore, g:Profiler was used to perform a functional enrichment analysis using the Gene Ontology (GO) and REAC databases. Enriched signaling pathways were identified using integrated bioinformatics enrichment analyses. The results revealed that only 15 of the genes identified in the bibliometric analysis were previously associated with DM1 in the DisGeNET database. Of note, we identified 71 genes not previously associated with DM1, which are of particular interest and should be further explored. The functional enrichment analysis of these genes revealed that regulation of cellular metabolic and metabolic processes were the most associated biological processes. Additionally, a number of signaling pathways were found to be enriched, e.g., signaling by receptor tyrosine kinases, signaling by NRTK1 (TRKA), TRKA activation by NGF, PI3K-AKT activation, prolonged ERK activation events, and axon guidance. Overall, several valuable target candidates related to metabolic processes for DM1 were identified, such as NGF, NTRK1, RhoA, ROCK1, ROCK2, DAG, ACTA, ID1, ID2 MYOD, and MYOG. Therefore, our study strengthens the hypothesis that metabolic dysfunctions contribute to DM1 pathogenesis, and the exploitation of metabolic dysfunction targets is crucial for the development of future therapeutic interventions for DM1.
Collapse
|
4
|
Aoussim A, Légaré C, Roussel MP, Madore AM, Morissette MC, Laprise C, Duchesne E. Towards the Identification of Biomarkers for Muscle Function Improvement in Myotonic Dystrophy Type 1. J Neuromuscul Dis 2023; 10:1041-1053. [PMID: 37694373 PMCID: PMC10657677 DOI: 10.3233/jnd-221645] [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] [Accepted: 08/15/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy in adults. In DM1 patients, skeletal muscle is severely impaired, even atrophied and patients experience a progressive decrease in maximum strength. Strength training for these individuals can improve their muscle function and mass, however, the biological processes involved in these improvements remain unknown. OBJECTIVE This exploratory study aims at identifying the proteomic biomarkers and variables associated with the muscle proteome changes induced by training in DM1 individuals. METHODS An ion library was developed from liquid chromatography-tandem mass spectrometry proteomic analyses of Vastus Lateralis muscle biopsies collected in 11 individuals with DM1 pre-and post-training. RESULTS The proteomic analysis showed that the levels of 44 proteins were significantly modulated. A literature review (PubMed, UniProt, PANTHER, REACTOME) classified these proteins into biological sub-classes linked to training-induced response, including immunity, energy metabolism, apoptosis, insulin signaling, myogenesis and muscle contraction. Linear models identified key variables explaining the proteome modulation, including atrophy and hypertrophy factors. Finally, six proteins of interest involved in myogenesis, muscle contraction and insulin signaling were identified: calpain-3 (CAN3; Muscle development, positive regulation of satellite cell activation), 14-3-3 protein epsilon (1433E; Insulin/Insulin-like growth factor, PI3K/Akt signaling), myosin-binding protein H (MYBPH; Regulation of striated muscle contraction), four and a half LIM domains protein 3 (FHL3; Muscle organ development), filamin-C (FLNC; Muscle fiber development) and Cysteine and glycine-rich protein 3 (CSRP3). CONCLUSION These findings may lead to the identification for DM1 individuals of novel muscle biomarkers for clinical improvement induced by rehabilitation, which could eventually be used in combination with a targeted pharmaceutical approach to improving muscle function, but further studies are needed to confirm those results.
Collapse
Affiliation(s)
- Amira Aoussim
- Département des sciences de la santé, Université du Québec à Chicoutimi, Québec, Canada
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires (GRIMN), Centre intégré universitaire de santé et de services sociaux du Saguenay– Lac-Saint-Jean, Hôpital de Jonquière, Québec, Canada
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi, Québec, Canada
| | - Cécilia Légaré
- Département des sciences de la santé, Université du Québec à Chicoutimi, Québec, Canada
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires (GRIMN), Centre intégré universitaire de santé et de services sociaux du Saguenay– Lac-Saint-Jean, Hôpital de Jonquière, Québec, Canada
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi, Québec, Canada
- RNA Institute, College of Arts and Sciences, University at Albany-SUNY, Albany, USA
| | - Marie-Pier Roussel
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires (GRIMN), Centre intégré universitaire de santé et de services sociaux du Saguenay– Lac-Saint-Jean, Hôpital de Jonquière, Québec, Canada
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi, Québec, Canada
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Québec, Canada
| | - Anne-Marie Madore
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi, Québec, Canada
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Québec, Canada
| | - Mathieu C. Morissette
- Department of Medicine, Université Laval, Québec, Canada
- Quebec Heart and Lung Institute – Université Laval, Québec, Canada
| | - Catherine Laprise
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi, Québec, Canada
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Québec, Canada
| | - Elise Duchesne
- Département des sciences de la santé, Université du Québec à Chicoutimi, Québec, Canada
- Groupe de recherche interdisciplinaire sur les maladies neuromusculaires (GRIMN), Centre intégré universitaire de santé et de services sociaux du Saguenay– Lac-Saint-Jean, Hôpital de Jonquière, Québec, Canada
- Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi, Québec, Canada
| |
Collapse
|
5
|
Franck S, Couvreu De Deckersberg E, Bubenik JL, Markouli C, Barbé L, Allemeersch J, Hilven P, Duqué G, Swanson MS, Gheldof A, Spits C, Sermon KD. Myotonic dystrophy type 1 embryonic stem cells show decreased myogenic potential, increased CpG methylation at the DMPK locus and RNA mis-splicing. Biol Open 2022; 11:273965. [PMID: 35019138 PMCID: PMC8764412 DOI: 10.1242/bio.058978] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle tissue is severely affected in myotonic dystrophy type 1 (DM1) patients, characterised by muscle weakness, myotonia and muscle immaturity in the most severe congenital form of the disease. Previously, it was not known at what stage during myogenesis the DM1 phenotype appears. In this study we differentiated healthy and DM1 human embryonic stem cells to myoblasts and myotubes and compared their differentiation potential using a comprehensive multi-omics approach. We found myogenesis in DM1 cells to be abnormal with altered myotube generation compared to healthy cells. We did not find differentially expressed genes between DM1 and non-DM1 cell lines within the same developmental stage. However, during differentiation we observed an aberrant inflammatory response and increased CpG methylation upstream of the CTG repeat at the myoblast level and RNA mis-splicing at the myotube stage. We show that early myogenesis modelled in hESC reiterates the early developmental manifestation of DM1. Summary: Early developmental abnormalities in myotonic dystrophy type 1 are reiterated in vitro in myotubes differentiated from human embryonic stem cells that carry the mutation.
Collapse
Affiliation(s)
- Silvie Franck
- Department Reproduction and Genetics, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | | | - Jodi L Bubenik
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Christina Markouli
- Department Reproduction and Genetics, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Lise Barbé
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, 94107 CA, United States
| | | | - Pierre Hilven
- Department Reproduction and Genetics, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Geoffrey Duqué
- Department Reproduction and Genetics, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics and the Genetics Institute, University of Florida, College of Medicine, Gainesville, FL 32610, USA
| | - Alexander Gheldof
- Department Reproduction and Genetics, Vrije Universiteit Brussel, Brussels 1090, Belgium.,Center for Medical Genetics, UZ Brussel, Brussels 1090, Belgium
| | - Claudia Spits
- Department Reproduction and Genetics, Vrije Universiteit Brussel, Brussels 1090, Belgium
| | - Karen D Sermon
- Department Reproduction and Genetics, Vrije Universiteit Brussel, Brussels 1090, Belgium
| |
Collapse
|
6
|
Klionsky DJ, Petroni G, Amaravadi RK, Baehrecke EH, Ballabio A, Boya P, Bravo‐San Pedro JM, Cadwell K, Cecconi F, Choi AMK, Choi ME, Chu CT, Codogno P, Colombo M, Cuervo AM, Deretic V, Dikic I, Elazar Z, Eskelinen E, Fimia GM, Gewirtz DA, Green DR, Hansen M, Jäättelä M, Johansen T, Juhász G, Karantza V, Kraft C, Kroemer G, Ktistakis NT, Kumar S, Lopez‐Otin C, Macleod KF, Madeo F, Martinez J, Meléndez A, Mizushima N, Münz C, Penninger JM, Perera R, Piacentini M, Reggiori F, Rubinsztein DC, Ryan K, Sadoshima J, Santambrogio L, Scorrano L, Simon H, Simon AK, Simonsen A, Stolz A, Tavernarakis N, Tooze SA, Yoshimori T, Yuan J, Yue Z, Zhong Q, Galluzzi L, Pietrocola F. Autophagy in major human diseases. EMBO J 2021; 40:e108863. [PMID: 34459017 PMCID: PMC8488577 DOI: 10.15252/embj.2021108863] [Citation(s) in RCA: 926] [Impact Index Per Article: 231.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a core molecular pathway for the preservation of cellular and organismal homeostasis. Pharmacological and genetic interventions impairing autophagy responses promote or aggravate disease in a plethora of experimental models. Consistently, mutations in autophagy-related processes cause severe human pathologies. Here, we review and discuss preclinical data linking autophagy dysfunction to the pathogenesis of major human disorders including cancer as well as cardiovascular, neurodegenerative, metabolic, pulmonary, renal, infectious, musculoskeletal, and ocular disorders.
Collapse
Affiliation(s)
| | - Giulia Petroni
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNYUSA
| | - Ravi K Amaravadi
- Department of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Abramson Cancer CenterUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer BiologyUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Andrea Ballabio
- Telethon Institute of Genetics and MedicinePozzuoliItaly
- Department of Translational Medical SciencesSection of PediatricsFederico II UniversityNaplesItaly
- Department of Molecular and Human GeneticsBaylor College of Medicine, and Jan and Dan Duncan Neurological Research InstituteTexas Children HospitalHoustonTXUSA
| | - Patricia Boya
- Margarita Salas Center for Biological ResearchSpanish National Research CouncilMadridSpain
| | - José Manuel Bravo‐San Pedro
- Faculty of MedicineDepartment Section of PhysiologyComplutense University of MadridMadridSpain
- Center for Networked Biomedical Research in Neurodegenerative Diseases (CIBERNED)MadridSpain
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball InstituteNew York University Grossman School of MedicineNew YorkNYUSA
- Department of MicrobiologyNew York University Grossman School of MedicineNew YorkNYUSA
- Division of Gastroenterology and HepatologyDepartment of MedicineNew York University Langone HealthNew YorkNYUSA
| | - Francesco Cecconi
- Cell Stress and Survival UnitCenter for Autophagy, Recycling and Disease (CARD)Danish Cancer Society Research CenterCopenhagenDenmark
- Department of Pediatric Onco‐Hematology and Cell and Gene TherapyIRCCS Bambino Gesù Children's HospitalRomeItaly
- Department of BiologyUniversity of Rome ‘Tor Vergata’RomeItaly
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care MedicineJoan and Sanford I. Weill Department of MedicineWeill Cornell MedicineNew YorkNYUSA
- New York‐Presbyterian HospitalWeill Cornell MedicineNew YorkNYUSA
| | - Mary E Choi
- New York‐Presbyterian HospitalWeill Cornell MedicineNew YorkNYUSA
- Division of Nephrology and HypertensionJoan and Sanford I. Weill Department of MedicineWeill Cornell MedicineNew YorkNYUSA
| | - Charleen T Chu
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Patrice Codogno
- Institut Necker‐Enfants MaladesINSERM U1151‐CNRS UMR 8253ParisFrance
- Université de ParisParisFrance
| | - Maria Isabel Colombo
- Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia‐Instituto de Histología y Embriología (IHEM)‐Universidad Nacional de CuyoCONICET‐ Facultad de Ciencias MédicasMendozaArgentina
| | - Ana Maria Cuervo
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNYUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineBronxNYUSA
| | - Vojo Deretic
- Autophagy Inflammation and Metabolism (AIMCenter of Biomedical Research ExcellenceUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
- Department of Molecular Genetics and MicrobiologyUniversity of New Mexico Health Sciences CenterAlbuquerqueNMUSA
| | - Ivan Dikic
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt, Frankfurt am MainGermany
- Buchmann Institute for Molecular Life SciencesGoethe UniversityFrankfurt, Frankfurt am MainGermany
| | - Zvulun Elazar
- Department of Biomolecular SciencesThe Weizmann Institute of ScienceRehovotIsrael
| | | | - Gian Maria Fimia
- Department of Molecular MedicineSapienza University of RomeRomeItaly
- Department of EpidemiologyPreclinical Research, and Advanced DiagnosticsNational Institute for Infectious Diseases ‘L. Spallanzani’ IRCCSRomeItaly
| | - David A Gewirtz
- Department of Pharmacology and ToxicologySchool of MedicineVirginia Commonwealth UniversityRichmondVAUSA
| | - Douglas R Green
- Department of ImmunologySt. Jude Children's Research HospitalMemphisTNUSA
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery InstituteProgram of DevelopmentAging, and RegenerationLa JollaCAUSA
| | - Marja Jäättelä
- Cell Death and MetabolismCenter for Autophagy, Recycling & DiseaseDanish Cancer Society Research CenterCopenhagenDenmark
- Department of Cellular and Molecular MedicineFaculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Terje Johansen
- Department of Medical BiologyMolecular Cancer Research GroupUniversity of Tromsø—The Arctic University of NorwayTromsøNorway
| | - Gábor Juhász
- Institute of GeneticsBiological Research CenterSzegedHungary
- Department of Anatomy, Cell and Developmental BiologyEötvös Loránd UniversityBudapestHungary
| | | | - Claudine Kraft
- Institute of Biochemistry and Molecular BiologyZBMZFaculty of MedicineUniversity of FreiburgFreiburgGermany
- CIBSS ‐ Centre for Integrative Biological Signalling StudiesUniversity of FreiburgFreiburgGermany
| | - Guido Kroemer
- Centre de Recherche des CordeliersEquipe Labellisée par la Ligue Contre le CancerUniversité de ParisSorbonne UniversitéInserm U1138Institut Universitaire de FranceParisFrance
- Metabolomics and Cell Biology PlatformsInstitut Gustave RoussyVillejuifFrance
- Pôle de BiologieHôpital Européen Georges PompidouAP‐HPParisFrance
- Suzhou Institute for Systems MedicineChinese Academy of Medical SciencesSuzhouChina
- Karolinska InstituteDepartment of Women's and Children's HealthKarolinska University HospitalStockholmSweden
| | | | - Sharad Kumar
- Centre for Cancer BiologyUniversity of South AustraliaAdelaideSAAustralia
- Faculty of Health and Medical SciencesUniversity of AdelaideAdelaideSAAustralia
| | - Carlos Lopez‐Otin
- Departamento de Bioquímica y Biología MolecularFacultad de MedicinaInstituto Universitario de Oncología del Principado de Asturias (IUOPA)Universidad de OviedoOviedoSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)MadridSpain
| | - Kay F Macleod
- The Ben May Department for Cancer ResearchThe Gordon Center for Integrative SciencesW‐338The University of ChicagoChicagoILUSA
- The University of ChicagoChicagoILUSA
| | - Frank Madeo
- Institute of Molecular BiosciencesNAWI GrazUniversity of GrazGrazAustria
- BioTechMed‐GrazGrazAustria
- Field of Excellence BioHealth – University of GrazGrazAustria
| | - Jennifer Martinez
- Immunity, Inflammation and Disease LaboratoryNational Institute of Environmental Health SciencesNIHResearch Triangle ParkNCUSA
| | - Alicia Meléndez
- Biology Department, Queens CollegeCity University of New YorkFlushingNYUSA
- The Graduate Center Biology and Biochemistry PhD Programs of the City University of New YorkNew YorkNYUSA
| | - Noboru Mizushima
- Department of Biochemistry and Molecular BiologyGraduate School of MedicineThe University of TokyoTokyoJapan
| | - Christian Münz
- Viral ImmunobiologyInstitute of Experimental ImmunologyUniversity of ZurichZurichSwitzerland
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA)Vienna BioCenter (VBC)ViennaAustria
- Department of Medical GeneticsLife Sciences InstituteUniversity of British ColumbiaVancouverBCCanada
| | - Rushika M Perera
- Department of AnatomyUniversity of California, San FranciscoSan FranciscoCAUSA
- Department of PathologyUniversity of California, San FranciscoSan FranciscoCAUSA
- Helen Diller Family Comprehensive Cancer CenterUniversity of California, San FranciscoSan FranciscoCAUSA
| | - Mauro Piacentini
- Department of BiologyUniversity of Rome “Tor Vergata”RomeItaly
- Laboratory of Molecular MedicineInstitute of Cytology Russian Academy of ScienceSaint PetersburgRussia
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells & SystemsMolecular Cell Biology SectionUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - David C Rubinsztein
- Department of Medical GeneticsCambridge Institute for Medical ResearchUniversity of CambridgeCambridgeUK
- UK Dementia Research InstituteUniversity of CambridgeCambridgeUK
| | - Kevin M Ryan
- Cancer Research UK Beatson InstituteGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowGlasgowUK
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular MedicineCardiovascular Research InstituteRutgers New Jersey Medical SchoolNewarkNJUSA
| | - Laura Santambrogio
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNYUSA
- Sandra and Edward Meyer Cancer CenterNew YorkNYUSA
- Caryl and Israel Englander Institute for Precision MedicineNew YorkNYUSA
| | - Luca Scorrano
- Istituto Veneto di Medicina MolecolarePadovaItaly
- Department of BiologyUniversity of PadovaPadovaItaly
| | - Hans‐Uwe Simon
- Institute of PharmacologyUniversity of BernBernSwitzerland
- Department of Clinical Immunology and AllergologySechenov UniversityMoscowRussia
- Laboratory of Molecular ImmunologyInstitute of Fundamental Medicine and BiologyKazan Federal UniversityKazanRussia
| | | | - Anne Simonsen
- Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
- Centre for Cancer Cell ReprogrammingInstitute of Clinical MedicineUniversity of OsloOsloNorway
- Department of Molecular Cell BiologyInstitute for Cancer ResearchOslo University Hospital MontebelloOsloNorway
| | - Alexandra Stolz
- Institute of Biochemistry IISchool of MedicineGoethe UniversityFrankfurt, Frankfurt am MainGermany
- Buchmann Institute for Molecular Life SciencesGoethe UniversityFrankfurt, Frankfurt am MainGermany
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and BiotechnologyFoundation for Research and Technology‐HellasHeraklion, CreteGreece
- Department of Basic SciencesSchool of MedicineUniversity of CreteHeraklion, CreteGreece
| | - Sharon A Tooze
- Molecular Cell Biology of AutophagyThe Francis Crick InstituteLondonUK
| | - Tamotsu Yoshimori
- Department of GeneticsGraduate School of MedicineOsaka UniversitySuitaJapan
- Department of Intracellular Membrane DynamicsGraduate School of Frontier BiosciencesOsaka UniversitySuitaJapan
- Integrated Frontier Research for Medical Science DivisionInstitute for Open and Transdisciplinary Research Initiatives (OTRI)Osaka UniversitySuitaJapan
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryChinese Academy of SciencesShanghaiChina
- Department of Cell BiologyHarvard Medical SchoolBostonMAUSA
| | - Zhenyu Yue
- Department of NeurologyFriedman Brain InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Qing Zhong
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationDepartment of PathophysiologyShanghai Jiao Tong University School of Medicine (SJTU‐SM)ShanghaiChina
| | - Lorenzo Galluzzi
- Department of Radiation OncologyWeill Cornell Medical CollegeNew YorkNYUSA
- Sandra and Edward Meyer Cancer CenterNew YorkNYUSA
- Caryl and Israel Englander Institute for Precision MedicineNew YorkNYUSA
- Department of DermatologyYale School of MedicineNew HavenCTUSA
- Université de ParisParisFrance
| | | |
Collapse
|
7
|
Todorow V, Hintze S, Kerr ARW, Hehr A, Schoser B, Meinke P. Transcriptome Analysis in a Primary Human Muscle Cell Differentiation Model for Myotonic Dystrophy Type 1. Int J Mol Sci 2021; 22:8607. [PMID: 34445314 PMCID: PMC8395314 DOI: 10.3390/ijms22168607] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 01/01/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is caused by CTG-repeat expansions leading to a complex pathology with a multisystemic phenotype that primarily affects the muscles and brain. Despite a multitude of information, especially on the alternative splicing of several genes involved in the pathology, information about additional factors contributing to the disease development is still lacking. We performed RNAseq and gene expression analyses on proliferating primary human myoblasts and differentiated myotubes. GO-term analysis indicates that in myoblasts and myotubes, different molecular pathologies are involved in the development of the muscular phenotype. Gene set enrichment for splicing reveals the likelihood of whole, differentiation stage specific, splicing complexes that are misregulated in DM1. These data add complexity to the alternative splicing phenotype and we predict that it will be of high importance for therapeutic interventions to target not only mature muscle, but also satellite cells.
Collapse
Affiliation(s)
- Vanessa Todorow
- Department of Neurology, Friedrich-Baur-Institute, LMU Klinikum, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Stefan Hintze
- Department of Neurology, Friedrich-Baur-Institute, LMU Klinikum, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Alastair R W Kerr
- Cancer Biomarker Centre, CRUK Manchester Institute, University of Manchester, Manchester SK10 4TG, UK
| | - Andreas Hehr
- Centre for Human Genetics, 93047 Regensburg, Germany
| | - Benedikt Schoser
- Department of Neurology, Friedrich-Baur-Institute, LMU Klinikum, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| | - Peter Meinke
- Department of Neurology, Friedrich-Baur-Institute, LMU Klinikum, Ludwig-Maximilians-University Munich, 80336 Munich, Germany
| |
Collapse
|
8
|
Grande V, Hathazi D, O'Connor E, Marteau T, Schara-Schmidt U, Hentschel A, Gourdon G, Nikolenko N, Lochmüller H, Roos A. Dysregulation of GSK3β-Target Proteins in Skin Fibroblasts of Myotonic Dystrophy Type 1 (DM1) Patients. J Neuromuscul Dis 2021; 8:603-619. [PMID: 33682722 DOI: 10.3233/jnd-200558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is the most common monogenetic muscular disorder of adulthood. This multisystemic disease is caused by CTG repeat expansion in the 3'-untranslated region of the DM1 protein kinase gene called DMPK. DMPK encodes a myosin kinase expressed in skeletal muscle cells and other cellular populations such as smooth muscle cells, neurons and fibroblasts. The resultant expanded (CUG)n RNA transcripts sequester RNA binding factors leading to ubiquitous and persistent splicing deregulation. The accumulation of mutant CUG repeats is linked to increased activity of glycogen synthase kinase 3 beta (GSK3β), a highly conserved and ubiquitous serine/threonine kinase with functions in pathways regulating inflammation, metabolism, oncogenesis, neurogenesis and myogenesis. As GSK3β-inhibition ameliorates defects in myogenesis, muscle strength and myotonia in a DM1 mouse model, this kinase represents a key player of DM1 pathobiochemistry and constitutes a promising therapeutic target. To better characterise DM1 patients, and monitor treatment responses, we aimed to define a set of robust disease and severity markers linked to GSK3βby unbiased proteomic profiling utilizing fibroblasts derived from DM1 patients with low (80- 150) and high (2600- 3600) CTG-repeats. Apart from GSK3β increase, we identified dysregulation of nine proteins (CAPN1, CTNNB1, CTPS1, DNMT1, HDAC2, HNRNPH3, MAP2K2, NR3C1, VDAC2) modulated by GSK3β. In silico-based expression studies confirmed expression in neuronal and skeletal muscle cells and revealed a relatively elevated abundance in fibroblasts. The potential impact of each marker in the myopathology of DM1 is discussed based on respective function to inform potential uses as severity markers or for monitoring GSK3β inhibitor treatment responses.
Collapse
Affiliation(s)
- Valentina Grande
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany
| | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany.,Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Emily O'Connor
- Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Theo Marteau
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany
| | - Ulrike Schara-Schmidt
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany
| | - Andreas Hentschel
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Genevieve Gourdon
- Centre de Recherche en Myologie, Association Institut de Myologie, Sorbonne Université, Inserm UMR 974, Paris, France
| | - Nikoletta Nikolenko
- National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Foundation Trust, London, UK
| | - Hanns Lochmüller
- Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada.,Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany.,Centro Nacional de AnálisisGenómico, Center for Genomic Regulation (CNAG-CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Andreas Roos
- Department of Neuropediatrics, University Hospital Essen, Duisburg-Essen University, Germany.,Childrens Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
9
|
Morton SU, Sefton CR, Zhang H, Dai M, Turner DL, Uhler MD, Agrawal PB. microRNA-mRNA Profile of Skeletal Muscle Differentiation and Relevance to Congenital Myotonic Dystrophy. Int J Mol Sci 2021; 22:ijms22052692. [PMID: 33799993 PMCID: PMC7962092 DOI: 10.3390/ijms22052692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/25/2021] [Accepted: 03/04/2021] [Indexed: 01/08/2023] Open
Abstract
microRNAs (miRNAs) regulate messenger RNA (mRNA) abundance and translation during key developmental processes including muscle differentiation. Assessment of miRNA targets can provide insight into muscle biology and gene expression profiles altered by disease. mRNA and miRNA libraries were generated from C2C12 myoblasts during differentiation, and predicted miRNA targets were identified based on presence of miRNA binding sites and reciprocal expression. Seventeen miRNAs were differentially expressed at all time intervals (comparing days 0, 2, and 5) of differentiation. mRNA targets of differentially expressed miRNAs were enriched for functions related to calcium signaling and sarcomere formation. To evaluate this relationship in a disease state, we evaluated the miRNAs differentially expressed in human congenital myotonic dystrophy (CMD) myoblasts and compared with normal control. Seventy-four miRNAs were differentially expressed during healthy human myocyte maturation, of which only 12 were also up- or downregulated in CMD patient cells. The 62 miRNAs that were only differentially expressed in healthy cells were compared with differentiating C2C12 cells. Eighteen of the 62 were conserved in mouse and up- or down-regulated during mouse myoblast differentiation, and their C2C12 targets were enriched for functions related to muscle differentiation and contraction.
Collapse
Affiliation(s)
- Sarah U. Morton
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: (S.U.M.); (P.B.A.)
| | | | - Huanqing Zhang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (H.Z.); (M.D.); (D.L.T.); (M.D.U.)
| | - Manhong Dai
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (H.Z.); (M.D.); (D.L.T.); (M.D.U.)
| | - David L. Turner
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (H.Z.); (M.D.); (D.L.T.); (M.D.U.)
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael D. Uhler
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (H.Z.); (M.D.); (D.L.T.); (M.D.U.)
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pankaj B. Agrawal
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
- Correspondence: (S.U.M.); (P.B.A.)
| |
Collapse
|
10
|
Dong W, Liu Q, Wang ZC, Du XX, Liu LL, Wang N, Weng JF, Peng XP. miR-322/miR-503 clusters regulate defective myoblast differentiation in myotonic dystrophy RNA-toxic by targeting Celf1. Toxicol Res (Camb) 2021; 10:29-39. [PMID: 33613970 DOI: 10.1093/toxres/tfaa096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/05/2020] [Accepted: 11/17/2020] [Indexed: 11/13/2022] Open
Abstract
Myotonic dystrophy (DM) is a genetic disorder featured by muscular dystrophy. It is caused by CUG expansion in the myotonic dystrophy protein kinase gene that leads to aberrant signaling and impaired myocyte differentiation. Many studies have shown that microRNAs are involved in the differentiation process of myoblasts. The purpose of this study was to investigate how the miR-322/miR-503 cluster regulates intracellular signaling to affect cell differentiation. The cell model of DM1 was employed by expressing GFP-CUG200 or CUGBP Elav-like family member 1 (Celf1) in myoblasts. Immunostaining of MF-20 was performed to examine myocyte differentiation. qRT-PCR and western blot were used to determine the levels of Celf1, MyoD, MyoG, Mef2c, miR-322/miR-503, and mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK/ERK) signaling. Dual luciferase assay was performed to validate the interaction between miR-322/miR-503 and Celf1. CUG expansion in myoblasts impaired the cell differentiation, increased the Celf1 level, but it decreased the miR-322/miR-503 levels. miR-322/miR-503 mimics restored the impaired differentiation caused by CUG expansion, while miR-322/miR-503 inhibitors further suppressed. miR-322/miR-503 directly targeted Celf1 and negatively regulated its expression. Knockdown of Celf1 promoted myocyte differentiation. Further, miR-322/miR-503 mimics rescued the impaired differentiation of myocytes caused by CUG expansion or Celf1 overexpression through suppressing of MEK/ERK signaling. miR-322/miR-503 cluster recover the defective myocyte differentiation caused by RNA-toxic via targeting Celf1. Restoring miR-322/miR-503 levels could be an avenue for DM1 therapy.
Collapse
Affiliation(s)
- Wei Dong
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province 330006, P. R. China
| | - Qian Liu
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province 330006, P. R. China
| | - Zhi-Chao Wang
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province 330006, P. R. China
| | - Xing-Xiang Du
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province 330006, P. R. China
| | - Lei-Lei Liu
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province 330006, P. R. China
| | - Nan Wang
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province 330006, P. R. China
| | - Jun-Fei Weng
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province 330006, P. R. China
| | - Xiao-Ping Peng
- Department of Cardiovascular, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi Province 330006, P. R. China
| |
Collapse
|
11
|
Yanay N, Rabie M, Nevo Y. Impaired Regeneration in Dystrophic Muscle-New Target for Therapy. Front Mol Neurosci 2020; 13:69. [PMID: 32523512 PMCID: PMC7261890 DOI: 10.3389/fnmol.2020.00069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
Muscle stem cells (MuSCs), known as satellite cells (SCs) have an incredible ability to regenerate, which enables the maintenance and growth of muscle tissue. In response to damaging stimuli, SCs are activated, proliferate, differentiate, and fuse to repair or generate a new muscle fiber. However, dystrophic muscles are characterized by poor muscle regeneration along with chronic inflammation and fibrosis. Indications for SC involvement in muscular dystrophy pathologies are accumulating, but their contribution to muscle pathophysiology is not precisely understood. In congenital muscular dystrophy type 1A (LAMA2-CMD), mutations in Lama2 gene cause either complete or partial absence in laminin-211 protein. Laminin-211 functions as a link between muscle extracellular matrix (ECM) and two adhesion systems in the sarcolemma; one is the well-known dystrophin-glycoprotein complex (DGC), and the second is the integrin complex. Because of its protein interactions and location, laminin-211 has a crucial role in muscle function and survival by maintaining sarcolemma integrity. In addition, laminin-211 is expressed in SCs and suggested to have a role in SC proliferation and differentiation. Downstream to the primary defect in laminin-211, several secondary genes and pathways accelerate disease mechanism, while at the same time there are unsuccessful attempts to regenerate as compensation for the dystrophic process. Lately, next-generation sequencing platforms have advanced our knowledge about the secondary events occurring in various diseases, elucidate the pathophysiology, and characterize new essential targets for development of new treatment strategies. This review will mainly focus on SC contribution to impaired regeneration in muscular dystrophies and specifically new findings suggesting SC involvement in LAMA2-CMD pathology.
Collapse
Affiliation(s)
- Nurit Yanay
- Felsenstein Medical Research Center (FMRC), Tel-Aviv University, Tel-Aviv, Israel.,Institute of Neurology, Schneider Children's Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | - Malcolm Rabie
- Felsenstein Medical Research Center (FMRC), Tel-Aviv University, Tel-Aviv, Israel.,Institute of Neurology, Schneider Children's Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | - Yoram Nevo
- Felsenstein Medical Research Center (FMRC), Tel-Aviv University, Tel-Aviv, Israel.,Institute of Neurology, Schneider Children's Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
12
|
Abstract
Skeletal muscle fibres are multinucleated cells that contain postmitotic nuclei (i.e. they are no longer able to divide) and perform muscle contraction. They are formed by fusion of muscle precursor cells, and grow into elongating myofibres by the addition of further precursor cells, called satellite cells, which are also responsible for regeneration following injury. Skeletal muscle regeneration occurs in most muscular dystrophies in response to necrosis of muscle fibres. However, the complex environment within dystrophic skeletal muscle, which includes inflammatory cells, fibroblasts and fibro-adipogenic cells, together with the genetic background of the in vivo model and the muscle being studied, complicates the interpretation of laboratory studies on muscular dystrophies. Many genes are expressed in satellite cells and in other tissues, which makes it difficult to determine the molecular cause of various types of muscular dystrophies. Here, and in the accompanying poster, we discuss our current knowledge of the cellular mechanisms that govern the growth and regeneration of skeletal muscle, and highlight the defects in satellite cell function that give rise to muscular dystrophies. Summary: The mechanisms of skeletal muscle development, growth and regeneration are described. We discuss whether these processes are dysregulated in inherited muscle diseases and identify pathways that may represent therapeutic targets.
Collapse
Affiliation(s)
- Jennifer Morgan
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK .,National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Terence Partridge
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.,National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK.,Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Ave NW, Washington, DC 20010, USA
| |
Collapse
|
13
|
Dosage effect of multiple genes accounts for multisystem disorder of myotonic dystrophy type 1. Cell Res 2019; 30:133-145. [PMID: 31853004 PMCID: PMC7015062 DOI: 10.1038/s41422-019-0264-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 11/09/2019] [Indexed: 12/19/2022] Open
Abstract
Multisystem manifestations in myotonic dystrophy type 1 (DM1) may be due to dosage reduction in multiple genes induced by aberrant expansion of CTG repeats in DMPK, including DMPK, its neighboring genes (SIX5 or DMWD) and downstream MBNL1. However, direct evidence is lacking. Here, we develop a new strategy to generate mice carrying multigene heterozygous mutations to mimic dosage reduction in one step by injection of haploid embryonic stem cells with mutant Dmpk, Six5 and Mbnl1 into oocytes. The triple heterozygous mutant mice exhibit adult-onset DM1 phenotypes. With the additional mutation in Dmwd, the quadruple heterozygous mutant mice recapitulate many major manifestations in congenital DM1. Moreover, muscle stem cells in both models display reduced stemness, providing a unique model for screening small molecules for treatment of DM1. Our results suggest that the complex symptoms of DM1 result from the reduced dosage of multiple genes.
Collapse
|
14
|
Sabater-Arcis M, Bargiela A, Furling D, Artero R. miR-7 Restores Phenotypes in Myotonic Dystrophy Muscle Cells by Repressing Hyperactivated Autophagy. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 19:278-292. [PMID: 31855836 PMCID: PMC6926285 DOI: 10.1016/j.omtn.2019.11.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 11/08/2019] [Accepted: 11/08/2019] [Indexed: 12/16/2022]
Abstract
Unstable CTG expansions in the 3’ UTR of the DMPK gene are responsible for myotonic dystrophy type 1 (DM1) condition. Muscle dysfunction is one of the main contributors to DM1 mortality and morbidity. Pathways by which mutant DMPK trigger muscle defects, however, are not fully understood. We previously reported that miR-7 was downregulated in a DM1 Drosophila model and in biopsies from patients. Here, using DM1 and normal muscle cells, we investigated whether miR-7 contributes to the muscle phenotype by studying the consequences of replenishing or blocking miR-7, respectively. Restoration of miR-7 with agomiR-7 was sufficient to rescue DM1 myoblast fusion defects and myotube growth. Conversely, oligonucleotide-mediated blocking of miR-7 in normal myoblasts led to fusion and myotube growth defects. miR-7 was found to regulate autophagy and the ubiquitin-proteasome system in human muscle cells. Thus, low levels of miR-7 promoted both processes, and high levels of miR-7 repressed them. Furthermore, we uncovered that the mechanism by which miR-7 improves atrophy-related phenotypes is independent of MBNL1, thus suggesting that miR-7 acts downstream or in parallel to MBNL1. Collectively, these results highlight an unknown function for miR-7 in muscle dysfunction through autophagy- and atrophy-related pathways and support that restoration of miR-7 levels is a candidate therapeutic target for counteracting muscle dysfunction in DM1.
Collapse
Affiliation(s)
- Maria Sabater-Arcis
- Translational Genomics Group, Incliva Health Research Institute, Valencia 46100, Spain; Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), University of Valencia, Valencia 46100, Spain; CIPF-INCLIVA Joint Unit, Valencia 46012, Spain
| | - Ariadna Bargiela
- Translational Genomics Group, Incliva Health Research Institute, Valencia 46100, Spain; Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), University of Valencia, Valencia 46100, Spain; CIPF-INCLIVA Joint Unit, Valencia 46012, Spain.
| | - Denis Furling
- Sorbonne Université, Inserm, Association Institut de Myologie, Centre de Recherche en Myologie, Paris 75013, France
| | - Ruben Artero
- Translational Genomics Group, Incliva Health Research Institute, Valencia 46100, Spain; Interdisciplinary Research Structure for Biotechnology and Biomedicine (ERI BIOTECMED), University of Valencia, Valencia 46100, Spain; CIPF-INCLIVA Joint Unit, Valencia 46012, Spain
| |
Collapse
|
15
|
Renna LV, Bosè F, Brigonzi E, Fossati B, Meola G, Cardani R. Aberrant insulin receptor expression is associated with insulin resistance and skeletal muscle atrophy in myotonic dystrophies. PLoS One 2019; 14:e0214254. [PMID: 30901379 PMCID: PMC6430513 DOI: 10.1371/journal.pone.0214254] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/08/2019] [Indexed: 12/26/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) and type 2 (DM2) are autosomal dominant multisystemic disorders linked to two different genetic loci and characterized by several features including myotonia, muscle atrophy and insulin resistance. The aberrant alternative splicing of insulin receptor (IR) gene and post-receptor signalling abnormalities have been associated with insulin resistance, however the precise molecular defects that cause metabolic dysfunctions are still unknown. Thus, the aims of this study were to investigate in DM skeletal muscle biopsies if beyond INSR missplicing, altered IR protein expression could play a role in insulin resistance and to verify if the lack of insulin pathway activation could contribute to skeletal muscle wasting. Our analysis showed that DM skeletal muscle exhibits a lower expression of the insulin receptor in type 1 fibers which can contribute to the defective activation of the insulin pathway. Moreover, the aberrant insulin signalling activation leads to a lower activation of mTOR and to an increase in MuRF1 and Atrogin-1/MAFbx expression, possible explaining DM skeletal muscle fiber atrophy. Taken together our data indicate that the defective insulin signalling activation can contribute to skeletal muscle features in DM patients and are probably linked to an aberrant specific-fiber type expression of the insulin receptor.
Collapse
Affiliation(s)
- Laura Valentina Renna
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Francesca Bosè
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Elisa Brigonzi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Barbara Fossati
- Department of Neurology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Giovanni Meola
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Department of Neurology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Rosanna Cardani
- Laboratory of Muscle Histopathology and Molecular Biology, IRCCS-Policlinico San Donato, San Donato Milanese, Milan, Italy
- * E-mail:
| |
Collapse
|
16
|
Morriss GR, Rajapakshe K, Huang S, Coarfa C, Cooper TA. Mechanisms of skeletal muscle wasting in a mouse model for myotonic dystrophy type 1. Hum Mol Genet 2019; 27:2789-2804. [PMID: 29771332 DOI: 10.1093/hmg/ddy192] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 05/14/2018] [Indexed: 12/18/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a multi-systemic disease resulting in severe muscle weakening and wasting. DM1 is caused by expansion of CTG repeats in the 3' untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. We have developed an inducible, skeletal muscle-specific mouse model of DM1 (CUG960) that expresses 960 CUG repeat-expressing animals (CUG960) in the context of human DMPK exons 11-15. CUG960 RNA-expressing mice induced at postnatal day 1, as well as adult-onset animals, show clear, measurable muscle wasting accompanied by severe histological defects including central myonuclei, reduced fiber cross-sectional area, increased percentage of oxidative myofibers, the presence of nuclear RNA foci that colocalize with Mbnl1 protein, and increased Celf1 protein in severely affected muscles. Importantly, muscle loss, histological abnormalities and RNA foci are reversible, demonstrating recovery upon removal of toxic RNA. RNA-seq and protein array analysis indicate that the balance between anabolic and catabolic pathways that normally regulate muscle mass may be disrupted by deregulation of platelet derived growth factor receptor β signaling and the PI3K/AKT pathways, along with prolonged activation of AMP-activated protein kinase α signaling. Similar changes were detected in DM1 skeletal muscle compared with unaffected controls. The mouse model presented in this paper shows progressive skeletal muscle wasting and has been used to identify potential molecular mechanisms underlying skeletal muscle loss. The reversibility of the phenotype establishes a baseline response for testing therapeutic approaches.
Collapse
Affiliation(s)
- Ginny R Morriss
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Dan L. Duncan Cancer Center, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Thomas A Cooper
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
17
|
Maury Y, Poydenot P, Brinon B, Lesueur L, Gide J, Roquevière S, Côme J, Polvèche H, Auboeuf D, Alexandre Denis J, Pietu G, Furling D, Lechuga M, Baghdoyan S, Peschanski M, Martinat C. Pluripotent Stem Cell-Based Drug Screening Reveals Cardiac Glycosides as Modulators of Myotonic Dystrophy Type 1. iScience 2019; 11:258-271. [PMID: 30639849 PMCID: PMC6327858 DOI: 10.1016/j.isci.2018.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/14/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023] Open
Abstract
There is currently no treatment for myotonic dystrophy type 1 (DM1), the most frequent myopathy of genetic origin. This progressive neuromuscular disease is caused by nuclear-retained RNAs containing expanded CUG repeats. These toxic RNAs alter the activities of RNA splicing factors, resulting in alternative splicing misregulation. By combining human mutated pluripotent stem cells and phenotypic drug screening, we revealed that cardiac glycosides act as modulators for both upstream nuclear aggregations of DMPK mRNAs and several downstream alternative mRNA splicing defects. However, these occurred at different drug concentration ranges. Similar biological effects were recorded in a DM1 mouse model. At the mechanistic level, we demonstrated that this effect was calcium dependent and was synergic with inhibition of the ERK pathway. These results further underscore the value of stem-cell-based assays for drug discovery in monogenic diseases. Myotonic dystrophy type 1 hPSCs were adapted for high content screening FDA-approved cardiac glycosides normalize in vitro and in vivo DM1 biological markers Cardiac glycosides synergize with the ERK pathway to normalize DM1 biomarkers This study emphasizes the value of human pluripotent stem cells for drug discovery
Collapse
Affiliation(s)
- Yves Maury
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | | | | | - Lea Lesueur
- INSERM, UMR 861, UEVE, ISTEM, AFM, 91100 Corbeil-Essonnes, France
| | | | | | - Julien Côme
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | | | | | | | - Geneviève Pietu
- INSERM, UMR 861, UEVE, ISTEM, AFM, 91100 Corbeil-Essonnes, France
| | - Denis Furling
- Sorbonne Universités UPMC Univ Paris 06, INSERM, Centre de Recherche en Myologie - UMRS974, Institut de Myologie, 75013 Paris, France
| | - Marc Lechuga
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France
| | | | - Marc Peschanski
- CECS, I-STEM, AFM, 91100 Corbeil-Essonnes, France; INSERM, UMR 861, UEVE, ISTEM, AFM, 91100 Corbeil-Essonnes, France
| | - Cécile Martinat
- INSERM, UMR 861, UEVE, ISTEM, AFM, 91100 Corbeil-Essonnes, France.
| |
Collapse
|
18
|
Activation of the interferon type I response rather than autophagy contributes to myogenesis inhibition in congenital DM1 myoblasts. Cell Death Dis 2018; 9:1071. [PMID: 30341284 PMCID: PMC6195593 DOI: 10.1038/s41419-018-1080-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/16/2022]
Abstract
Congenital myotonic dystrophy type 1 (CDM1) is characterized by severe symptoms that affect patients from birth, with 40% mortality in the neonatal period and impaired skeletal muscle development. In this paper, we examined the relationship between autophagy and abnormal myogenic differentiation of CDM1 myoblasts. We investigated these pathological features at both ultrastructural and molecular levels, utilizing two CDM1 foetal myoblasts, CDM13 and CDM15, with 1800 and 3200 repeats, respectively. The congenital nature of these CDM1 myoblasts was confirmed by the high methylation level at the DMPK locus. Our results indicated that abnormal autophagy was independent of myogenic differentiation, as CDM13 myoblasts differentiated as well as control myoblasts but underwent autophagy like CDM15, displaying impaired differentiation. miRNA expression profiles revealed that CDM15 myoblasts failed to upregulate the complex network of myo-miRNAs under MYOD and MEF2A control, while this network was upregulated in CDM13 myoblasts. Interestingly, the abnormal differentiation of CDM15 myoblasts was associated with cellular stress accompanied by the induction of the interferon type 1 pathway (innate immune response). Indeed, inhibition of the interferon (IFN) type I pathway restores myogenic differentiation of CDM15 myoblasts, suggesting that the inappropriate activation of the innate immune response might contribute to impaired myogenic differentiation and severe muscle symptoms observed in some CDM1 patients. These findings open up the possibility of new therapeutic approaches to treat CDM1.
Collapse
|
19
|
Dial AG, Ng SY, Manta A, Ljubicic V. The Role of AMPK in Neuromuscular Biology and Disease. Trends Endocrinol Metab 2018; 29:300-312. [PMID: 29572064 DOI: 10.1016/j.tem.2018.02.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 12/22/2022]
Abstract
AMP-activated protein kinase (AMPK) is a primary regulator of cellular metabolism. Recent studies have revealed that AMPK also mediates the maintenance and plasticity of α-motoneurons, the neuromuscular junction, and skeletal muscle. Furthermore, AMPK stimulation by either genetic, pharmacological, or physiological approaches elicits beneficial phenotypic remodeling in neuromuscular disorders (NMDs). Here, we review the role of AMPK as a governor of neuromuscular biology, and present evidence for AMPK as an effective molecular target for therapeutic pursuit in the context of the most prevalent NMDs, including Duchenne muscular dystrophy, spinal muscular atrophy, and myotonic dystrophy type 1. This information may be useful for engineering AMPK-targeted pharmacological- or lifestyle-based strategies to treat disorders of the neuromuscular system.
Collapse
Affiliation(s)
- Athan G Dial
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Sean Y Ng
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Alexander Manta
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Vladimir Ljubicic
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
20
|
Castets P, Frank S, Sinnreich M, Rüegg MA. "Get the Balance Right": Pathological Significance of Autophagy Perturbation in Neuromuscular Disorders. J Neuromuscul Dis 2018; 3:127-155. [PMID: 27854220 PMCID: PMC5271579 DOI: 10.3233/jnd-160153] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent research has revealed that autophagy, a major catabolic process in cells, is dysregulated in several neuromuscular diseases and contributes to the muscle wasting caused by non-muscle disorders (e.g. cancer cachexia) or during aging (i.e. sarcopenia). From there, the idea arose to interfere with autophagy or manipulate its regulatory signalling to help restore muscle homeostasis and attenuate disease progression. The major difficulty for the development of therapeutic strategies is to restore a balanced autophagic flux, due to the dynamic nature of autophagy. Thus, it is essential to better understand the mechanisms and identify the signalling pathways at play in the control of autophagy in skeletal muscle. A comprehensive analysis of the autophagic flux and of the causes of its dysregulation is required to assess the pathogenic role of autophagy in diseased muscle. Furthermore, it is essential that experiments distinguish between primary dysregulation of autophagy (prior to disease onset) and impairments as a consequence of the pathology. Of note, in most muscle disorders, autophagy perturbation is not caused by genetic modification of an autophagy-related protein, but rather through indirect alteration of regulatory signalling or lysosomal function. In this review, we will present the mechanisms involved in autophagy, and those ensuring its tight regulation in skeletal muscle. We will then discuss as to how autophagy dysregulation contributes to the pathogenesis of neuromuscular disorders and possible ways to interfere with this process to limit disease progression.
Collapse
Affiliation(s)
| | - Stephan Frank
- Institute of Pathology, Division of Neuropathology Basel University Hospital, Basel, Switzerland
| | - Michael Sinnreich
- Neuromuscular Research Center, Departments of Neurology and Biomedicine, Pharmazentrum, Basel, Switzerland
| | | |
Collapse
|
21
|
Brockhoff M, Rion N, Chojnowska K, Wiktorowicz T, Eickhorst C, Erne B, Frank S, Angelini C, Furling D, Rüegg MA, Sinnreich M, Castets P. Targeting deregulated AMPK/mTORC1 pathways improves muscle function in myotonic dystrophy type I. J Clin Invest 2017; 127:549-563. [PMID: 28067669 DOI: 10.1172/jci89616] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022] Open
Abstract
Myotonic dystrophy type I (DM1) is a disabling multisystemic disease that predominantly affects skeletal muscle. It is caused by expanded CTG repeats in the 3'-UTR of the dystrophia myotonica protein kinase (DMPK) gene. RNA hairpins formed by elongated DMPK transcripts sequester RNA-binding proteins, leading to mis-splicing of numerous pre-mRNAs. Here, we have investigated whether DM1-associated muscle pathology is related to deregulation of central metabolic pathways, which may identify potential therapeutic targets for the disease. In a well-characterized mouse model for DM1 (HSALR mice), activation of AMPK signaling in muscle was impaired under starved conditions, while mTORC1 signaling remained active. In parallel, autophagic flux was perturbed in HSALR muscle and in cultured human DM1 myotubes. Pharmacological approaches targeting AMPK/mTORC1 signaling greatly ameliorated muscle function in HSALR mice. AICAR, an AMPK activator, led to a strong reduction of myotonia, which was accompanied by partial correction of misregulated alternative splicing. Rapamycin, an mTORC1 inhibitor, improved muscle relaxation and increased muscle force in HSALR mice without affecting splicing. These findings highlight the involvement of AMPK/mTORC1 deregulation in DM1 muscle pathophysiology and may open potential avenues for the treatment of this disease.
Collapse
|
22
|
Bargiela A, Cerro-Herreros E, Fernandez-Costa JM, Vilchez JJ, Llamusi B, Artero R. Increased autophagy and apoptosis contribute to muscle atrophy in a myotonic dystrophy type 1 Drosophila model. Dis Model Mech 2016; 8:679-90. [PMID: 26092529 PMCID: PMC4486854 DOI: 10.1242/dmm.018127] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Muscle mass wasting is one of the most debilitating symptoms of myotonic dystrophy type 1 (DM1) disease, ultimately leading to immobility, respiratory defects, dysarthria, dysphagia and death in advanced stages of the disease. In order to study the molecular mechanisms leading to the degenerative loss of adult muscle tissue in DM1, we generated an inducible Drosophila model of expanded CTG trinucleotide repeat toxicity that resembles an adult-onset form of the disease. Heat-shock induced expression of 480 CUG repeats in adult flies resulted in a reduction in the area of the indirect flight muscles. In these model flies, reduction of muscle area was concomitant with increased apoptosis and autophagy. Inhibition of apoptosis or autophagy mediated by the overexpression of DIAP1, mTOR (also known as Tor) or muscleblind, or by RNA interference (RNAi)-mediated silencing of autophagy regulatory genes, achieved a rescue of the muscle-loss phenotype. In fact, mTOR overexpression rescued muscle size to a size comparable to that in control flies. These results were validated in skeletal muscle biopsies from DM1 patients in which we found downregulated autophagy and apoptosis repressor genes, and also in DM1 myoblasts where we found increased autophagy. These findings provide new insights into the signaling pathways involved in DM1 disease pathogenesis.
Collapse
Affiliation(s)
- Ariadna Bargiela
- Translational Genomics Group, Department of Genetics, University of Valencia, Burjassot 46100, Spain INCLIVA Health Research Institute, Valencia 46010, Spain
| | - Estefanía Cerro-Herreros
- Translational Genomics Group, Department of Genetics, University of Valencia, Burjassot 46100, Spain INCLIVA Health Research Institute, Valencia 46010, Spain
| | - Juan M Fernandez-Costa
- Translational Genomics Group, Department of Genetics, University of Valencia, Burjassot 46100, Spain
| | - Juan J Vilchez
- Neurology Section, Hospital Universitari La Fe, Valencia 46026, Spain Department of Internal Medicine, University of Valencia, Valencia 46010, Spain Centro de Investigaciones Biomedicas en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Institute Carlos III, Ministry of Economy and Competitiveness, Madrid 28049, Spain
| | | | - Ruben Artero
- Translational Genomics Group, Department of Genetics, University of Valencia, Burjassot 46100, Spain INCLIVA Health Research Institute, Valencia 46010, Spain
| |
Collapse
|
23
|
Randolph ME, Pavlath GK. A muscle stem cell for every muscle: variability of satellite cell biology among different muscle groups. Front Aging Neurosci 2015; 7:190. [PMID: 26500547 PMCID: PMC4595652 DOI: 10.3389/fnagi.2015.00190] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/21/2015] [Indexed: 12/22/2022] Open
Abstract
The human body contains approximately 640 individual skeletal muscles. Despite the fact that all of these muscles are composed of striated muscle tissue, the biology of these muscles and their associated muscle stem cell populations are quite diverse. Skeletal muscles are affected differentially by various muscular dystrophies (MDs), such that certain genetic mutations specifically alter muscle function in only a subset of muscles. Additionally, defective muscle stem cells have been implicated in the pathology of some MDs. The biology of muscle stem cells varies depending on the muscles with which they are associated. Here we review the biology of skeletal muscle stem cell populations of eight different muscle groups. Understanding the biological variation of skeletal muscles and their resident stem cells could provide valuable insight into mechanisms underlying the susceptibility of certain muscles to myopathic disease.
Collapse
|
24
|
Mateos-Aierdi AJ, Goicoechea M, Aiastui A, Fernández-Torrón R, Garcia-Puga M, Matheu A, López de Munain A. Muscle wasting in myotonic dystrophies: a model of premature aging. Front Aging Neurosci 2015. [PMID: 26217220 PMCID: PMC4496580 DOI: 10.3389/fnagi.2015.00125] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1 or Steinert’s disease) and type 2 (DM2) are multisystem disorders of genetic origin. Progressive muscular weakness, atrophy and myotonia are the most prominent neuromuscular features of these diseases, while other clinical manifestations such as cardiomyopathy, insulin resistance and cataracts are also common. From a clinical perspective, most DM symptoms are interpreted as a result of an accelerated aging (cataracts, muscular weakness and atrophy, cognitive decline, metabolic dysfunction, etc.), including an increased risk of developing tumors. From this point of view, DM1 could be described as a progeroid syndrome since a notable age-dependent dysfunction of all systems occurs. The underlying molecular disorder in DM1 consists of the existence of a pathological (CTG) triplet expansion in the 3′ untranslated region (UTR) of the Dystrophia Myotonica Protein Kinase (DMPK) gene, whereas (CCTG)n repeats in the first intron of the Cellular Nucleic acid Binding Protein/Zinc Finger Protein 9(CNBP/ZNF9) gene cause DM2. The expansions are transcribed into (CUG)n and (CCUG)n-containing RNA, respectively, which form secondary structures and sequester RNA-binding proteins, such as the splicing factor muscleblind-like protein (MBNL), forming nuclear aggregates known as foci. Other splicing factors, such as CUGBP, are also disrupted, leading to a spliceopathy of a large number of downstream genes linked to the clinical features of these diseases. Skeletal muscle regeneration relies on muscle progenitor cells, known as satellite cells, which are activated after muscle damage, and which proliferate and differentiate to muscle cells, thus regenerating the damaged tissue. Satellite cell dysfunction seems to be a common feature of both age-dependent muscle degeneration (sarcopenia) and muscle wasting in DM and other muscle degenerative diseases. This review aims to describe the cellular, molecular and macrostructural processes involved in the muscular degeneration seen in DM patients, highlighting the similarities found with muscle aging.
Collapse
Affiliation(s)
- Alba Judith Mateos-Aierdi
- Neuroscience Area, Biodonostia Health Research Institute San Sebastián, Spain ; CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain
| | - Maria Goicoechea
- Neuroscience Area, Biodonostia Health Research Institute San Sebastián, Spain ; CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain
| | - Ana Aiastui
- CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain ; Cell Culture Platform, Biodonostia Health Research Institute, San Sebastián Spain
| | - Roberto Fernández-Torrón
- Neuroscience Area, Biodonostia Health Research Institute San Sebastián, Spain ; CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain ; Department of Neurology, Hospital Universitario Donostia, San Sebastián Spain
| | - Mikel Garcia-Puga
- Oncology Area, Biodonostia Health Research Institute San Sebastián, Spain
| | - Ander Matheu
- Oncology Area, Biodonostia Health Research Institute San Sebastián, Spain
| | - Adolfo López de Munain
- Neuroscience Area, Biodonostia Health Research Institute San Sebastián, Spain ; CIBERNED, Instituto Carlos III, Ministerio de Economía y Competitividad Madrid, Spain ; Department of Neurology, Hospital Universitario Donostia, San Sebastián Spain ; Department of Neuroscience, Universidad del País Vasco UPV-EHU San Sebastián, Spain
| |
Collapse
|
25
|
Effect of itraconazole on mouse mesencephalic neurons. Int J Dev Neurosci 2015; 44:75-83. [DOI: 10.1016/j.ijdevneu.2015.06.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/13/2015] [Accepted: 06/06/2015] [Indexed: 01/14/2023] Open
|
26
|
Girolamo F, Lia A, Amati A, Strippoli M, Coppola C, Virgintino D, Roncali L, Toscano A, Serlenga L, Trojano M. Overexpression of autophagic proteins in the skeletal muscle of sporadic inclusion body myositis. Neuropathol Appl Neurobiol 2014; 39:736-49. [PMID: 23452291 DOI: 10.1111/nan.12040] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 02/22/2013] [Indexed: 01/01/2023]
Abstract
AIMS Sporadic inclusion body myositis (s-IBM) is characterized by rimmed vacuole formation and misfolded protein accumulation. Intracellular protein aggregates are cleared by autophagy. When autophagy is blocked aggregates accumulate, resulting in abnormal rimmed vacuole formation. This study investigated the autophagy-lysosome pathway contribution to rimmed vacuole accumulation. METHODS Autophagy was studied in muscle biopsy specimens obtained from eleven s-IBM patients, one suspected hereditary IBM patient, nine patients with other inflammatory myopathies and nine non-myopathic patients as controls. The analysis employed morphometric methods applied to immunohistochemistry using the endosome marker Clathrin, essential proteins of the autophagic cascade such as AuTophaGy-related protein ATG5, splicing variants of microtubule-associated protein light chain 3a (LC3a) and LC3b, compared with Beclin 1, the major autophagy regulator of both the initiation phase and late endosome/lysosome fusion of the autophagy-lysosome pathway. RESULTS In muscle biopsies of s-IBM patients, an increased expression of Clathrin, ATG5, LC3a, LC3b and Beclin 1 was shown. Moreover, the inflammatory components of the disease, essentially lymphocytes, were preferentially distributed around the Beclin 1(+) myofibres. These affected myofibres also showed a moderate sarcoplasmic accumulation of SMI-31(+) phospho-tau paired helical filaments. CONCLUSION The overexpression of autophagy markers linked to the decreased clearance of misfolded proteins, including SMI-31, and rimmed vacuoles accumulation may exhaust cellular resources and lead to cell death.
Collapse
Affiliation(s)
- F Girolamo
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari School of Medicine, Bari, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Lin Y, Tang C, He H, Duan R. Activation of mTOR ameliorates fragile X premutation rCGG repeat-mediated neurodegeneration. PLoS One 2013; 8:e62572. [PMID: 23626835 PMCID: PMC3633886 DOI: 10.1371/journal.pone.0062572] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/23/2013] [Indexed: 01/25/2023] Open
Abstract
Fragile X associated tremor/ataxia syndrome (FXTAS) is a late onset neurodegenerative disorder caused by aberrant expansion of CGG repeats in 5′ UTR of FMR1 gene. The elevated mRNA confers a toxic gain-of-function thought to be the critical event of pathogenesis. Expressing rCGG90 repeats of the human FMR1 5′UTR in Drosophila is sufficient to induce neurodegeneration. Rapamycin has been demonstrated to attenuate neurotoxicity by inducing autophagy in various animal models of neurodegenerative diseases. Surprisingly, we observed rapamycin exacerbated rCGG90-induced neurodegenerative phenotypes through an autophagy-independent mechanism. CGG90 expression levels of FXTAS flies exposed to rapamycin presented no significant differences. We further demonstrated that activation of the mammalian target of rapamycin (mTOR) signaling could suppress neurodegeneration of FXTAS. These findings indicate that rapamycin will exacerbate neurodegeneration, and that enhancing autophagy is insufficient to alleviate neurotoxicity in FXTAS. Moreover, these results suggest mTOR and its downstream molecules as new therapeutic targets for FXTAS by showing significant protection against neurodegeneration.
Collapse
Affiliation(s)
- Yunting Lin
- The State Key Laboratory of Medical Genetics, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Chengyuan Tang
- The State Key Laboratory of Medical Genetics, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Hua He
- The State Key Laboratory of Medical Genetics, Xiangya Medical School, Central South University, Changsha, Hunan, China
| | - Ranhui Duan
- The State Key Laboratory of Medical Genetics, Xiangya Medical School, Central South University, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
28
|
Denis JA, Gauthier M, Rachdi L, Aubert S, Giraud-Triboult K, Poydenot P, Benchoua A, Champon B, Maury Y, Baldeschi C, Scharfmann R, Piétu G, Peschanski M, Martinat C. mTOR-dependent proliferation defect in human ES-derived neural stem cells affected by myotonic dystrophy type 1. J Cell Sci 2013; 126:1763-72. [PMID: 23444380 DOI: 10.1242/jcs.116285] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Patients with myotonic dystrophy type 1 exhibit a diversity of symptoms that affect many different organs. Among these are cognitive dysfunctions, the origin of which has remained elusive, partly because of the difficulty in accessing neural cells. Here, we have taken advantage of pluripotent stem cell lines derived from embryos identified during a pre-implantation genetic diagnosis for mutant-gene carriers, to produce early neuronal cells. Functional characterization of these cells revealed reduced proliferative capacity and increased autophagy linked to mTOR signaling pathway alterations. Interestingly, loss of function of MBNL1, an RNA-binding protein whose function is defective in DM1 patients, resulted in alteration of mTOR signaling, whereas gain-of-function experiments rescued the phenotype. Collectively, these results provide a mechanism by which DM1 mutation might affect a major signaling pathway and highlight the pertinence of using pluripotent stem cells to study neuronal defects.
Collapse
Affiliation(s)
- Jérôme Alexandre Denis
- INSERM/UEVE U-861, I-STEM, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 5 rue Henri Desbruères, 91030 Evry cedex, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Itraconazole inhibits HMEC-1 angiogenesis. Biomed Pharmacother 2012; 66:312-7. [DOI: 10.1016/j.biopha.2011.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 11/21/2011] [Indexed: 12/12/2022] Open
|
30
|
Abstract
Since 1929, when it was discovered that ATP is a substrate for muscle contraction, the knowledge about this purine nucleotide has been greatly expanded. Many aspects of cell metabolism revolve around ATP production and consumption. It is important to understand the concepts of glucose and oxygen consumption in aerobic and anaerobic life and to link bioenergetics with the vast amount of reactions occurring within cells. ATP is universally seen as the energy exchange factor that connects anabolism and catabolism but also fuels processes such as motile contraction, phosphorylations, and active transport. It is also a signalling molecule in the purinergic signalling mechanisms. In this review, we will discuss all the main mechanisms of ATP production linked to ADP phosphorylation as well the regulation of these mechanisms during stress conditions and in connection with calcium signalling events. Recent advances regarding ATP storage and its special significance for purinergic signalling will also be reviewed.
Collapse
|
31
|
Baraibar MA, Hyzewicz J, Rogowska-Wrzesinska A, Ladouce R, Roepstorff P, Mouly V, Friguet B. Oxidative stress-induced proteome alterations target different cellular pathways in human myoblasts. Free Radic Biol Med 2011; 51:1522-32. [PMID: 21810466 DOI: 10.1016/j.freeradbiomed.2011.06.032] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 06/27/2011] [Accepted: 06/27/2011] [Indexed: 11/20/2022]
Abstract
Although increased oxidative stress has been associated with the impairment of proliferation and function of adult human muscle stem cells, proteins either involved in the stress response or damaged by oxidation have not been identified. A parallel proteomics approach was performed for analyzing the protein expression profile as well as proteins preferentially oxidized upon hydrogen peroxide-induced oxidative stress. Fifteen proteins involved in the oxidative stress response were identified. Among them, protein spots identified as peroxiredoxins 1 and 6, glyceraldehyde-3-phosphate dehydrogenase, and α-enolase were shifted to a more acidic isoelectric point upon oxidative stress, indicating posttranslational modifications. Oxidized proteins were evidenced by immunodetection of derivatized carbonyl groups followed by identification by mass spectrometry. The carbonylated proteins identified are mainly cytosolic and involved in carbohydrate metabolism, cellular assembly, cellular homeostasis, and protein synthesis and degradation. Pathway analysis revealed skeletal and muscular disorders, cell death, and cancer-related as the main molecular networks altered. Interestingly, these pathways were focused on two distinct proteins: p53 for altered protein expression and huntingtin for increased protein carbonylation. This study emphasizes the importance of performing analysis addressing different aspects of the cellular proteome to have a more accurate view of their changes upon stress.
Collapse
Affiliation(s)
- Martin A Baraibar
- Laboratoire de Biologie Cellulaire du Vieillissement, UR4, Université Pierre et Marie Curie-Paris 6, 75252 Paris Cedex 05, France
| | | | | | | | | | | | | |
Collapse
|
32
|
Bareford MD, Park MA, Yacoub A, Hamed HA, Tang Y, Cruickshanks N, Eulitt P, Hubbard N, Tye G, Burow ME, Fisher PB, Moran RG, Nephew KP, Grant S, Dent P. Sorafenib enhances pemetrexed cytotoxicity through an autophagy-dependent mechanism in cancer cells. Cancer Res 2011; 71:4955-67. [PMID: 21622715 PMCID: PMC3139015 DOI: 10.1158/0008-5472.can-11-0898] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pemetrexed (ALIMTA, Lilly) is a folate antimetabolite that has been approved by the U.S. Food and Drug Administration for the treatment of non-small cell lung cancer and has been shown to stimulate autophagy. In the present study, we sought to further understand the role of autophagy in response to pemetrexed and to test if combination therapy could enhance the level of toxicity through altered autophagy in tumor cells. The multikinase inhibitor sorafenib (Nexavar, Bayer), used in the treatment of renal and hepatocellular carcinoma, suppresses tumor angiogenesis and promotes autophagy in tumor cells. We found that sorafenib interacted in a greater than additive fashion with pemetrexed to increase autophagy and to kill a diverse array of tumor cell types. Tumor cell types that displayed high levels of cell killing after combination treatment showed elevated levels of AKT, p70 S6K, and/or phosphorylated mTOR, in addition to class III receptor tyrosine kinases such as platelet-derived growth factor receptor beta and VEGF receptors, known in vivo targets of sorafenib. In xenograft and in syngeneic animal models of mammary carcinoma and glioblastoma, the combination of sorafenib and pemetrexed suppressed tumor growth without deleterious effects on normal tissues or animal body mass. Taken together, the data suggest that premexetred and sorafenib act synergistically to enhance tumor killing via the promotion of a toxic form of autophagy that leads to activation of the intrinsic apoptosis pathway, and predict that combination treatment represents a future therapeutic option in the treatment of solid tumors.
Collapse
Affiliation(s)
- M. Danielle Bareford
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Margaret A. Park
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Adly Yacoub
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Hossein A. Hamed
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Yong Tang
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Nichola Cruickshanks
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Patrick Eulitt
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Nisan Hubbard
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Gary Tye
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Matthew E. Burow
- Section of Hematology & Oncology, Tulane University School of Medicine, New Orleans LA 70112
| | - Paul B. Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
- Virginia Institute of Molecular Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Richard G. Moran
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Kenneth P. Nephew
- Department of Medical Sciences, Indiana University, School of Medicine, Bloomington, IN 47405
| | - Steven Grant
- Department of Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
- Virginia Institute of Molecular Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| | - Paul Dent
- Department of Neurosurgery, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
- Virginia Institute of Molecular Medicine, Virginia Commonwealth University, 401 College St., Richmond, VA 23298
| |
Collapse
|