1
|
Shahin‐Shamsabadi A, Cappuccitti J. In Vivo-Like Scaffold-Free 3D In Vitro Models of Muscular Dystrophies: The Case for Anchored Cell Sheet Engineering in Personalized Medicine. Adv Healthc Mater 2025; 14:e2404465. [PMID: 39718233 PMCID: PMC12057609 DOI: 10.1002/adhm.202404465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Indexed: 12/25/2024]
Abstract
Progress in understanding the underlying mechanisms of muscular dystrophies is hindered by the lack of pathophysiologically relevant in vitro models. Here, an entirely scaffold-free anchored cell sheet engineering platform is used to create patient-specific three-dimensional (3D) skeletal muscle in vitro models. This approach effectively replicates mature muscle phenotypes and tissue- and disease-specific extracellular matric (ECM). Models were developed using primary cells from healthy individuals and patients with Duchenne Muscular Dystrophy and Myotonic Dystrophy Type 1. Through a combination of quantified histological staining (Hematoxylin & Eosin, Movat's Pentachrome, Masson's Trichrome) and immunostaining (desmin, myosin heavy chain, laminin, and dystrophin), it was demonstrated that the models formed mature constructs closely resembling their respective in vivo conditions. Proteomics analysis revealed that the models exhibited appropriate upregulation and downregulation of disease-relevant pathways. Models of diseased tissues accurately reflected key phenotypic features of the diseases, including alterations in muscle fiber integrity and ECM composition. Upon treatment with therapeutically beneficial drugs, significant changes in their proteomic profiles were documented, highlighting the models' potential for drug screening. This novel in vitro modeling approach, unlike other 3D techniques that rely on exogenous biomaterials that interfere with natural cellular behaviors, provides a promising platform for studying muscular dystrophies.
Collapse
|
2
|
Lisowska M, Rowińska M, Suszyńska A, Bearzi C, Łaczmańska I, Hanusek J, Kunik A, Dzianisava V, Rzepecki R, Machowska M, Piekarowicz K. Human iPSC-Derived Muscle Cells as a New Model for Investigation of EDMD1 Pathogenesis. Int J Mol Sci 2025; 26:1539. [PMID: 40004006 PMCID: PMC11855679 DOI: 10.3390/ijms26041539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Emery-Dreifuss muscular dystrophy type 1 (EDMD1) is a rare genetic disease caused by mutations in the EMD gene, which encodes the nuclear envelope protein emerin. Despite understanding the genetic basis of the disease, the molecular mechanism underlying muscle and cardiac pathogenesis remains elusive. Progress is restricted by the limited availability of patient-derived samples; therefore, there is an urgent need for human-specific cellular models. In this study, we present the generation and characterization of induced pluripotent stem cell (iPSC) lines derived from EDMD1 patients carrying EMD mutations that lead to truncated or absent emerin, together with iPSCs from healthy donor. The patient-specific iPSCs exhibit stable karyotypes, maintain appropriate morphology, express pluripotency markers, and demonstrate the ability to differentiate into three germ layers. To model EDMD1, these iPSCs were differentiated into myogenic progenitors, myoblasts, and multinucleated myotubes, which represent all stages of myogenesis. Each developmental stage was validated by the presence of stage-specific markers, ensuring the accuracy of the model. We present the first iPSC-based in vitro platform that captures the complexity of EDMD1 pathogenesis during myogenesis. This model can significantly contribute to understanding disease mechanisms and develop the targeted therapeutic strategies for EDMD1.
Collapse
Affiliation(s)
- Marta Lisowska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, 50-383 Wrocław, Poland
| | - Marta Rowińska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, 50-383 Wrocław, Poland
| | - Aleksandra Suszyńska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, 50-383 Wrocław, Poland
| | - Claudia Bearzi
- Institute for Biomedical Technologies, National Research Council, 20054 Segrate, Milan, Italy
| | - Izabela Łaczmańska
- Department of Genetics, Wrocław Medical University, 50-368 Wrocław, Poland
| | - Julia Hanusek
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, 50-383 Wrocław, Poland
| | - Amanda Kunik
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, 50-383 Wrocław, Poland
| | - Volha Dzianisava
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, 50-383 Wrocław, Poland
| | - Ryszard Rzepecki
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, 50-383 Wrocław, Poland
| | - Magdalena Machowska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, 50-383 Wrocław, Poland
| | - Katarzyna Piekarowicz
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, 50-383 Wrocław, Poland
| |
Collapse
|
3
|
Fernandes VAR, dos Santos GP, Iatecola A, Buchaim DV, Garcia IJF, Reis CHB, Bueno LMM, Pagani BT, Buchaim RL, da Cunha MR. Evaluation of Creatine Monohydrate Supplementation on the Gastrocnemius Muscle of Mice with Muscular Dystrophy: A Preliminary Study. PATHOPHYSIOLOGY 2025; 32:2. [PMID: 39846639 PMCID: PMC11755625 DOI: 10.3390/pathophysiology32010002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025] Open
Abstract
Background/Objectives: Duchenne muscular dystrophy (DMD) is a genetic disease characterized by a lack of dystrophin caused by mutations in the DMD gene, and some minor cases are due to decreased levels of dystrophin, leading to muscle weakness and motor impairment. Creatine supplementation has demonstrated several benefits for the muscle, such as increased strength, enhanced tissue repair, and improved ATP resynthesis. This preliminary study aimed to investigate the effects of creatine on the gastrocnemius muscle in dystrophy muscle (MDX) and healthy C57BL/10 mice. Methods: Twenty MDX and C57Bl/10 mice were organized into groups and supplemented or not with creatine in a dosage of 0.3 mg for 8 weeks. Gastrocnemius tissue was analyzed using histomorphology and histomorphometric techniques. Results: The results demonstrated potential anti-inflammatory effects of creatine, with less observation of inflammatory infiltrates, the preservation of intramuscular glycogen, and reduction in tissue fibrosis in supplemented animals. Conclusions: These findings suggest that creatine may enhance tissue function and slow the progression of DMD. However, further research, with more analysis, is needed to elucidate molecular mechanisms underlying creatine's effects on reducing mononuclear leukocytes and its role in mitigating tissue fibrosis.
Collapse
Affiliation(s)
- Victor Augusto Ramos Fernandes
- Postgraduate Program in Health Sciences, Faculty of Medicine of Jundiaí (FMJ), Jundiaí 13202-550, Brazil;
- Neurobiology Study Group, Nossa Senhora do Patrocínio University Center (CEUNSP), Itu 13300-200, Brazil; (G.P.d.S.); (A.I.)
| | - Gabriela Pereira dos Santos
- Neurobiology Study Group, Nossa Senhora do Patrocínio University Center (CEUNSP), Itu 13300-200, Brazil; (G.P.d.S.); (A.I.)
| | - Amilton Iatecola
- Neurobiology Study Group, Nossa Senhora do Patrocínio University Center (CEUNSP), Itu 13300-200, Brazil; (G.P.d.S.); (A.I.)
| | - Daniela Vieira Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo (FMVZ/USP), Sao Paulo 05508-270, Brazil; (D.V.B.); (R.L.B.)
- Medical and Dentistry School, University Center of Adamantina (FAI), Adamantina 17800-000, Brazil; (I.J.F.G.); (L.M.M.B.)
| | - Ionaly Judith Faria Garcia
- Medical and Dentistry School, University Center of Adamantina (FAI), Adamantina 17800-000, Brazil; (I.J.F.G.); (L.M.M.B.)
| | | | - Lívia Maluf Menegazzo Bueno
- Medical and Dentistry School, University Center of Adamantina (FAI), Adamantina 17800-000, Brazil; (I.J.F.G.); (L.M.M.B.)
| | - Bruna Trazzi Pagani
- Dentistry School, University of Marilia (UNIMAR), Marilia 17525-902, Brazil;
| | - Rogerio Leone Buchaim
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of Sao Paulo (FMVZ/USP), Sao Paulo 05508-270, Brazil; (D.V.B.); (R.L.B.)
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of Sao Paulo, Bauru 17012-901, Brazil
| | - Marcelo Rodrigues da Cunha
- Postgraduate Program in Health Sciences, Faculty of Medicine of Jundiaí (FMJ), Jundiaí 13202-550, Brazil;
- Neurobiology Study Group, Nossa Senhora do Patrocínio University Center (CEUNSP), Itu 13300-200, Brazil; (G.P.d.S.); (A.I.)
| |
Collapse
|
4
|
Siemionow M, Bocian K, Bozyk KT, Ziemiecka A, Siemionow K. Chimeric Cell Therapy Transfers Healthy Donor Mitochondria in Duchenne Muscular Dystrophy. Stem Cell Rev Rep 2024; 20:1819-1829. [PMID: 39017908 PMCID: PMC11445288 DOI: 10.1007/s12015-024-10756-w] [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: 06/23/2024] [Indexed: 07/18/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a severe X-linked disorder characterized by dystrophin gene mutations and mitochondrial dysfunction, leading to progressive muscle weakness and premature death of DMD patients. We developed human Dystrophin Expressing Chimeric (DEC) cells, created by the fusion of myoblasts from normal donors and DMD patients, as a foundation for DT-DEC01 therapy for DMD. Our preclinical studies on mdx mouse models of DMD revealed enhanced dystrophin expression and functional improvements in cardiac, respiratory, and skeletal muscles after systemic intraosseous DEC administration. The current study explored the feasibility of mitochondrial transfer and fusion within the created DEC cells, which is crucial for developing new therapeutic strategies for DMD. Following mitochondrial staining with MitoTracker Deep Red and MitoTracker Green dyes, mitochondrial fusion and transfer was assessed by Flow cytometry (FACS) and confocal microscopy. The PEG-mediated fusion of myoblasts from normal healthy donors (MBN/MBN) and normal and DMD-affected donors (MBN/MBDMD), confirmed the feasibility of myoblast and mitochondrial fusion and transfer. The colocalization of the mitochondrial dyes MitoTracker Deep Red and MitoTracker Green confirmed the mitochondrial chimeric state and the creation of chimeric mitochondria, as well as the transfer of healthy donor mitochondria within the created DEC cells. These findings are unique and significant, introducing the potential of DT-DEC01 therapy to restore mitochondrial function in DMD patients and in other diseases where mitochondrial dysfunction plays a critical role.
Collapse
Affiliation(s)
- Maria Siemionow
- Chair and Department of Traumatology, Orthopedics and Surgery of the Hand, Poznan University of Medical Sciences, Poznan, 61‑545, Poland.
- Dystrogen Therapeutics Technology Polska z o.o., Warsaw, 00-777, Poland.
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| | - Katarzyna Bocian
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, 02-096, Poland
- Polish Stem Cell Bank, FamiCord Group, Warsaw, 00-867, Poland
| | - Katarzyna T Bozyk
- Dystrogen Therapeutics Technology Polska z o.o., Warsaw, 00-777, Poland
| | - Anna Ziemiecka
- Dystrogen Therapeutics Technology Polska z o.o., Warsaw, 00-777, Poland
| | - Krzysztof Siemionow
- Dystrogen Therapeutics Technology Polska z o.o., Warsaw, 00-777, Poland
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
5
|
Grigor’eva EV, Karapetyan LV, Malakhova AA, Medvedev SP, Minina JM, Hayrapetyan VH, Vardanyan VS, Zakian SM, Arakelyan A, Zakharyan R. Generation of iPSCs from a Patient with the M694V Mutation in the MEFV Gene Associated with Familial Mediterranean Fever and Their Differentiation into Macrophages. Int J Mol Sci 2024; 25:6102. [PMID: 38892289 PMCID: PMC11173119 DOI: 10.3390/ijms25116102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/21/2024] Open
Abstract
Familial Mediterranean fever (FMF) is a systemic autoinflammatory disorder caused by inherited mutations in the MEFV (Mediterranean FeVer) gene, located on chromosome 16 (16p13.3) and encoding the pyrin protein. Despite the existing data on MEFV mutations, the exact mechanism of their effect on the development of the pathological processes leading to the spontaneous and recurrent autoinflammatory attacks observed in FMF, remains unclear. Induced pluripotent stem cells (iPSCs) are considered an important tool to study the molecular genetic mechanisms of various diseases due to their ability to differentiate into any cell type, including macrophages, which contribute to the development of FMF. In this study, we developed iPSCs from an Armenian patient with FMF carrying the M694V, p.(Met694Val) (c.2080A>G, rs61752717) pathogenic mutation in exon 10 of the MEFV gene. As a result of direct differentiation, macrophages expressing CD14 and CD45 surface markers were obtained. We found that the morphology of macrophages derived from iPSCs of a patient with the MEFV mutation significantly differed from that of macrophages derived from iPSCs of a healthy donor carrying the wild-type MEFV gene.
Collapse
Affiliation(s)
- Elena V. Grigor’eva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, 630055 Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Lana V. Karapetyan
- Department of Bioengineering, Bioinformatics, and Molecular Biology, Institute of Biomedicine and Pharmacy, Russian-Armenian (Slavonic) University, Yerevan 0051, Armenia; (L.V.K.); (V.H.H.); (A.A.)
| | - Anastasia A. Malakhova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, 630055 Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Sergey P. Medvedev
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, 630055 Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Julia M. Minina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
| | - Varduhi H. Hayrapetyan
- Department of Bioengineering, Bioinformatics, and Molecular Biology, Institute of Biomedicine and Pharmacy, Russian-Armenian (Slavonic) University, Yerevan 0051, Armenia; (L.V.K.); (V.H.H.); (A.A.)
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia
| | - Valentina S. Vardanyan
- Department of Rheumatology, Yerevan State Medical University after Mkhitar Heratsi (YSMU), Yerevan 0025, Armenia;
- Department of Rheumatology, “Mikaelyan” Institute of Surgery, Yerevan 0052, Armenia
| | - Suren M. Zakian
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia; (E.V.G.); (A.A.M.); (S.P.M.); (J.M.M.); (S.M.Z.)
- Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, 630055 Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Arsen Arakelyan
- Department of Bioengineering, Bioinformatics, and Molecular Biology, Institute of Biomedicine and Pharmacy, Russian-Armenian (Slavonic) University, Yerevan 0051, Armenia; (L.V.K.); (V.H.H.); (A.A.)
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia
| | - Roksana Zakharyan
- Department of Bioengineering, Bioinformatics, and Molecular Biology, Institute of Biomedicine and Pharmacy, Russian-Armenian (Slavonic) University, Yerevan 0051, Armenia; (L.V.K.); (V.H.H.); (A.A.)
- Institute of Molecular Biology NAS RA, Yerevan 0014, Armenia
| |
Collapse
|
6
|
Dubey AK, Mostafavi E. Biomaterials-mediated CRISPR/Cas9 delivery: recent challenges and opportunities in gene therapy. Front Chem 2023; 11:1259435. [PMID: 37841202 PMCID: PMC10568484 DOI: 10.3389/fchem.2023.1259435] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The use of biomaterials in delivering CRISPR/Cas9 for gene therapy in infectious diseases holds tremendous potential. This innovative approach combines the advantages of CRISPR/Cas9 with the protective properties of biomaterials, enabling accurate and efficient gene editing while enhancing safety. Biomaterials play a vital role in shielding CRISPR/Cas9 components, such as lipid nanoparticles or viral vectors, from immunological processes and degradation, extending their effectiveness. By utilizing the flexibility of biomaterials, tailored systems can be designed to address specific genetic diseases, paving the way for personalized therapeutics. Furthermore, this delivery method offers promising avenues in combating viral illnesses by precisely modifying pathogen genomes, and reducing their pathogenicity. Biomaterials facilitate site-specific gene modifications, ensuring effective delivery to infected cells while minimizing off-target effects. However, challenges remain, including optimizing delivery efficiency, reducing off-target effects, ensuring long-term safety, and establishing scalable production techniques. Thorough research, pre-clinical investigations, and rigorous safety evaluations are imperative for successful translation from the laboratory to clinical applications. In this review, we discussed how CRISPR/Cas9 delivery using biomaterials revolutionizes gene therapy and infectious disease treatment, offering precise and safe editing capabilities with the potential to significantly improve human health and quality of life.
Collapse
Affiliation(s)
- Ankit Kumar Dubey
- Global Research and Publishing Foundation, New Delhi, India
- Institute of Scholars, Bengaluru, Karnataka, India
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
7
|
Heydemann A, Bieganski G, Wachowiak J, Czarnota J, Niezgoda A, Siemionow K, Ziemiecka A, Sikorska MH, Bozyk K, Tullius SG, Siemionow M. Dystrophin Expressing Chimeric (DEC) Cell Therapy for Duchenne Muscular Dystrophy: A First-in-Human Study with Minimum 6 Months Follow-up. Stem Cell Rev Rep 2023:10.1007/s12015-023-10530-4. [PMID: 37000376 PMCID: PMC10366026 DOI: 10.1007/s12015-023-10530-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2023] [Indexed: 04/01/2023]
Abstract
Duchenne Muscular Dystrophy (DMD) is a X-linked progressive lethal muscle wasting disease for which there is no cure. We present first-in-human study assessing safety and efficacy of novel Dystrophin Expressing Chimeric (DEC) cell therapy created by fusion of patient myoblasts with myoblasts of normal donor origin. We report here on safety and functional outcomes of the first 3 DMD patients. No study related adverse events (AE) and no serious adverse events (SAE) were observed up to 14 months after systemic-intraosseous administration of DEC01. Ambulatory patients showed improvements in functional tests (6-Minute Walk Test (6MWT), North Star Ambulatory Assessment (NSAA)) and both, ambulatory and non-ambulatory in PUL, strength and fatigue resistance which correlated with improvement of Electromyography (EMG) parameters. DEC01 therapy does not require immunosuppression, involves no risks of off target mutations, is not dependent upon the causative mutation and is therefore a universal therapy that does not use viral vectors and therefore can be readministered, if needed. This study was approved by the Bioethics Committee (approval No. 46/2019). Mechanism of action of the Dystrophin Expressing Chimeric Cell (DEC) cells created via ex vivo fusion of human myoblast from normal and DMD-affected donors. Following systemic-intraosseous administration, DEC engraft and fuse with the myoblasts of DMD patients, deliver dystrophin and improve muscle strength and function. (Created with BioRender.com).
Collapse
Affiliation(s)
- Ahlke Heydemann
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
- Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Grzegorz Bieganski
- Department of Infectious Diseases and Child Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jacek Wachowiak
- Department of Pediatric Oncology, Hematology and Transplantology, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Adam Niezgoda
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | - Krzysztof Siemionow
- Dystrogen Therapeutics Corp., Chicago, IL, USA
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA
| | | | | | | | - Stefan G Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Siemionow
- Dystrogen Therapeutics Corp., Chicago, IL, USA.
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA.
- Department of Traumatology Orthopedics and Hand Surgery, Poznan University of Medical Sciences, Poznan, Poland.
| |
Collapse
|
8
|
Long-Term Protective Effect of Human Dystrophin Expressing Chimeric (DEC) Cell Therapy on Amelioration of Function of Cardiac, Respiratory and Skeletal Muscles in Duchenne Muscular Dystrophy. Stem Cell Rev Rep 2022; 18:2872-2892. [PMID: 35590083 PMCID: PMC9622520 DOI: 10.1007/s12015-022-10384-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 12/12/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) is a lethal disease caused by mutations in dystrophin encoding gene, causing progressive degeneration of cardiac, respiratory, and skeletal muscles leading to premature death due to cardiac and respiratory failure. Currently, there is no cure for DMD. Therefore, novel therapeutic approaches are needed for DMD patients. We have previously reported functional improvements which correlated with increased dystrophin expression following administration of dystrophin expressing chimeric (DEC) cells of myoblast origin to the mdx mouse models of DMD. In the current study, we confirmed dose-dependent protective effect of human DEC therapy created from myoblasts of normal and DMD-affected donors, on restoration of dystrophin expression and amelioration of cardiac, respiratory, and skeletal muscle function at 180 days after systemic-intraosseous DEC administration to mdx/scid mouse model of DMD. Functional improvements included maintenance of ejection fraction and fractional shortening levels on echocardiography, reduced enhanced pause and expiration time on plethysmography and improved grip strength and maximum stretch induced contraction of skeletal muscles. Improved function was associated with amelioration of mdx muscle pathology revealed by reduced muscle fibrosis, reduced inflammation and improved muscle morphology confirmed by reduced number of centrally nucleated fibers and normalization of muscle fiber diameters. Our findings confirm the long-term systemic effect of DEC therapy in the most severely affected by DMD organs including heart, diaphragm, and long skeletal muscles. These encouraging preclinical data introduces human DEC as a novel therapeutic modality of Advanced Therapy Medicinal Product (ATMP) with the potential to improve or halt the progression of DMD and enhance quality of life of DMD patients.
Collapse
|
9
|
Bourque K, Hawey C, Jiang A, Mazarura GR, Hébert TE. Biosensor-based profiling to track cellular signalling in patient-derived models of dilated cardiomyopathy. Cell Signal 2022; 91:110239. [PMID: 34990783 DOI: 10.1016/j.cellsig.2021.110239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/06/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022]
Abstract
Dilated cardiomyopathies (DCM) represent a diverse group of cardiovascular diseases impacting the structure and function of the myocardium. To better treat these diseases, we need to understand the impact of such cardiomyopathies on critical signalling pathways that drive disease progression downstream of receptors we often target therapeutically. Our understanding of cellular signalling events has progressed substantially in the last few years, in large part due to the design, validation and use of biosensor-based approaches to studying such events in cells, tissues and in some cases, living animals. Another transformative development has been the use of human induced pluripotent stem cells (hiPSCs) to generate disease-relevant models from individual patients. We highlight the importance of going beyond monocellular cultures to incorporate the influence of paracrine signalling mediators. Finally, we discuss the recent coalition of these approaches in the context of DCM. We discuss recent work in generating patient-derived models of cardiomyopathies and the utility of using signalling biosensors to track disease progression and test potential therapeutic strategies that can be later used to inform treatment options in patients.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Alyson Jiang
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Grace R Mazarura
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
10
|
Singh S, Singh T, Kunja C, Dhoat NS, Dhania NK. Gene-editing, immunological and iPSCs based therapeutics for muscular dystrophy. Eur J Pharmacol 2021; 912:174568. [PMID: 34656607 DOI: 10.1016/j.ejphar.2021.174568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/25/2021] [Accepted: 10/11/2021] [Indexed: 10/20/2022]
Abstract
Muscular dystrophy is a well-known genetically heterogeneous group of rare muscle disorders. This progressive disease causes the breakdown of skeletal muscles over time and leads to grave weakness. This breakdown is caused by a diverse pattern of mutations in dystrophin and dystrophin associated protein complex. These mutations lead to the production of altered proteins in response to which, the body stimulates production of various cytokines and immune cells, particularly reactive oxygen species and NFκB. Immune cells display/exhibit a dual role by inducing muscle damage and muscle repair. Various anti-oxidants, anti-inflammatory and glucocorticoid drugs serve as potent therapeutics for muscular dystrophy. Along with the above mentioned therapeutics, induced pluripotent stem cells also serve as a novel approach paving a way for personalized treatment. These pluripotent stem cells allow regeneration of large numbers of regenerative myogenic progenitors that can be administered in muscular dystrophy patients which assist in the recovery of lost muscle fibers. In this review, we have summarized gene-editing, immunological and induced pluripotent stem cell based therapeutics for muscular dystrophy treatment.
Collapse
Affiliation(s)
- Shagun Singh
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India
| | - Tejpal Singh
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India
| | - Chaitanya Kunja
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India
| | - Navdeep S Dhoat
- Department of Pediatrics Surgery, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India
| | - Narender K Dhania
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India.
| |
Collapse
|
11
|
Goullée H, Taylor RL, Forrest ARR, Laing NG, Ravenscroft G, Clayton JS. Improved CRISPR/Cas9 gene editing in primary human myoblasts using low confluency cultures on Matrigel. Skelet Muscle 2021; 11:23. [PMID: 34551826 PMCID: PMC8456651 DOI: 10.1186/s13395-021-00278-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 09/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background CRISPR/Cas9 is an invaluable tool for studying cell biology and the development of molecular therapies. However, delivery of CRISPR/Cas9 components into some cell types remains a major hurdle. Primary human myoblasts are a valuable cell model for muscle studies, but are notoriously difficult to transfect. There are currently no commercial lipofection protocols tailored for primary myoblasts, and most generic guidelines simply recommend transfecting healthy cells at high confluency. This study aimed to maximize CRISPR/Cas9 transfection and editing in primary human myoblasts. Methods Since increased cell proliferation is associated with increased transfection efficiency, we investigated two factors known to influence myoblast proliferation: cell confluency, and a basement membrane matrix, Matrigel. CRISPR/Cas9 editing was performed by delivering Cas9 ribonucleoprotein complexes via lipofection into primary human myoblasts, cultured in wells with or without a Matrigel coating, at low (~ 40%) or high (~ 80%) confluency. Results Cells transfected at low confluency on Matrigel-coated wells had the highest levels of transfection, and were most effectively edited across three different target loci, achieving a maximum editing efficiency of 93.8%. On average, editing under these conditions was >4-fold higher compared to commercial recommendations (high confluency, uncoated wells). Conclusion This study presents a simple, effective and economical method of maximizing CRISPR/Cas9-mediated gene editing in primary human myoblasts. This protocol could be a valuable tool for improving the genetic manipulation of cultured human skeletal muscle cells, and potentially be adapted for use in other cell types. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-021-00278-1.
Collapse
Affiliation(s)
- Hayley Goullée
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Harry Perkins Institute of Medical Research, 6 Verdun St, Nedlands, WA, 6009, Australia.,School of Biomedical Science, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Rhonda L Taylor
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Harry Perkins Institute of Medical Research, 6 Verdun St, Nedlands, WA, 6009, Australia.,School of Biomedical Science, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Alistair R R Forrest
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Harry Perkins Institute of Medical Research, 6 Verdun St, Nedlands, WA, 6009, Australia
| | - Nigel G Laing
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Harry Perkins Institute of Medical Research, 6 Verdun St, Nedlands, WA, 6009, Australia
| | - Gianina Ravenscroft
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Harry Perkins Institute of Medical Research, 6 Verdun St, Nedlands, WA, 6009, Australia
| | - Joshua S Clayton
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Nedlands, WA, Australia. .,Harry Perkins Institute of Medical Research, 6 Verdun St, Nedlands, WA, 6009, Australia.
| |
Collapse
|
12
|
Chitena L, Masisi K, Masisi K, Kwape TE, Gaobotse G. Application of Stem Cell Therapy during the treatment of HIV/AIDS and Duchenne Muscular Dystrophy. Curr Stem Cell Res Ther 2021; 17:633-647. [PMID: 35135463 DOI: 10.2174/1574888x16666210810104445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 11/22/2022]
Abstract
Treating diseases such as Muscular dystrophy (MD) and HIV/AIDS poses several challenges to the rapidly evolving field of regenerative medicine. Previously, stem cell therapy has been said to affect the clinical courses of HIV/AIDS and MD, but, in practice, eradication or control of these diseases was not achievable. The introduction of gene editing into stem cell therapy has stimulated HIV/AIDS and MD cell therapy research studies substantially. Here, we review current methods of treating HIV/AIDS and MD using stem cell therapy. This review also details the use of different types of cells and methods in cell therapy and the modeling of new cell-based therapies to treat Duchenne muscular dystrophy. We speculate that the effective use stem cell therapy in conjunction with other treatment therapies such as steroids and rehabilitation could improve livelihood.
Collapse
Affiliation(s)
- Lorraine Chitena
- Department of Biological Sciences and Biotechnology, Botswana International University of Science and Technology, Private Bag 16, Palapye. Botswana
| | - Keletso Masisi
- Department of Biological Sciences and Biotechnology, Botswana International University of Science and Technology, Private Bag 16, Palapye. Botswana
| | - Kabo Masisi
- Department of Biological Sciences and Biotechnology, Botswana International University of Science and Technology, Private Bag 16, Palapye. Botswana
| | - Tebogo E Kwape
- Department of Biological Sciences and Biotechnology, Botswana International University of Science and Technology, Private Bag 16, Palapye. Botswana
| | - Goabaone Gaobotse
- Department of Biological Sciences and Biotechnology, Botswana International University of Science and Technology, Private Bag 16, Palapye. Botswana
| |
Collapse
|
13
|
Basu A, Tiwari VK. Epigenetic reprogramming of cell identity: lessons from development for regenerative medicine. Clin Epigenetics 2021; 13:144. [PMID: 34301318 PMCID: PMC8305869 DOI: 10.1186/s13148-021-01131-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/13/2021] [Indexed: 12/17/2022] Open
Abstract
Epigenetic mechanisms are known to define cell-type identity and function. Hence, reprogramming of one cell type into another essentially requires a rewiring of the underlying epigenome. Cellular reprogramming can convert somatic cells to induced pluripotent stem cells (iPSCs) that can be directed to differentiate to specific cell types. Trans-differentiation or direct reprogramming, on the other hand, involves the direct conversion of one cell type into another. In this review, we highlight how gene regulatory mechanisms identified to be critical for developmental processes were successfully used for cellular reprogramming of various cell types. We also discuss how the therapeutic use of the reprogrammed cells is beginning to revolutionize the field of regenerative medicine particularly in the repair and regeneration of damaged tissue and organs arising from pathological conditions or accidents. Lastly, we highlight some key challenges hindering the application of cellular reprogramming for therapeutic purposes.
Collapse
Affiliation(s)
- Amitava Basu
- Institute of Molecular Biology (IMB), 55128, Mainz, Germany.
| | - Vijay K Tiwari
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queens University Belfast, Belfast, BT9 7BL, UK.
| |
Collapse
|
14
|
Gutiérrez J, Gonzalez D, Escalona-Rivano R, Takahashi C, Brandan E. Reduced RECK levels accelerate skeletal muscle differentiation, improve muscle regeneration, and decrease fibrosis. FASEB J 2021; 35:e21503. [PMID: 33811686 DOI: 10.1096/fj.202001646rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 02/07/2021] [Accepted: 02/19/2021] [Indexed: 12/15/2022]
Abstract
The muscle regeneration process requires a properly assembled extracellular matrix (ECM). Its homeostasis depends on the activity of different matrix-metalloproteinases (MMPs). The reversion-inducing-cysteine-rich protein with kazal motifs (RECK) is a membrane-anchored protein that negatively regulates the activity of different MMPs. However, the role of RECK in the process of skeletal muscle differentiation, regeneration, and fibrosis has not been elucidated. Here, we show that during skeletal muscle differentiation of C2C12 myoblasts and in satellite cells on isolated muscle fibers, RECK is transiently up regulated. C2C12 myoblasts with reduced RECK levels are more prone to enter the differentiation program, showing an accelerated differentiation process. Notch-1 signaling was reduced, while p38 and AKT signaling were augmented in myoblasts with decreased RECK levels. Overexpression of RECK restores the normal differentiation process but diminished the ability to form myotubes. Transient up-regulation of RECK occurs during skeletal muscle regeneration, which was accelerated in RECK-deficient mice (Reck±). RECK, MMPs and ECM proteins augmented in chronically damaged WT muscle, a model of muscle fibrosis. In this model, RECK ± mice showed diminished fibrosis compared to WT. These results strongly suggest that RECK is acting as a potential myogenic repressor during muscle formation and regeneration, emerging as a new player in these processes, and as a potential target to treat individuals with the muscle-wasting disease.
Collapse
Affiliation(s)
- Jaime Gutiérrez
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile.,Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David Gonzalez
- Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Escalona-Rivano
- Cellular Signaling and Differentiation Laboratory (CSDL), School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile
| | - Chiaki Takahashi
- Oncology and Molecular Biology, Cancer and Stem Cell Research Program, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Enrique Brandan
- Centro de Regeneración y Envejecimiento (CARE), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Fundación Ciencia & Vida, Santiago, Chile
| |
Collapse
|
15
|
Florczyk-Soluch U, Polak K, Dulak J. The multifaceted view of heart problem in Duchenne muscular dystrophy. Cell Mol Life Sci 2021; 78:5447-5468. [PMID: 34091693 PMCID: PMC8257522 DOI: 10.1007/s00018-021-03862-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/29/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022]
Abstract
Dystrophin is a large protein serving as local scaffolding repetitively bridging cytoskeleton and the outside of striated muscle cell. As such dystrophin is a critical brick primarily in dystrophin-associated protein complex (DAGC) and in a larger submembranous unit, costamere. Accordingly, the lack of functional dystrophin laying at the root of Duchenne muscular dystrophy (DMD) drives sarcolemma instability. From this point on, the cascade inevitably leading to the death of myocyte begins. In cardiomyocytes, intracellular calcium overload and related mitochondrial-mediated cell death mainly contribute to myocardial dysfunction and dilation while other protein dysregulation and/or mislocalization may affect electrical conduction system and favor arrhythmogenesis. Although clinically DMD manifests as progressive muscle weakness and skeletal muscle symptoms define characteristic of DMD, it is the heart problem the biggest challenge that most often develop in the form of dilated cardiomyopathy (DCM). Current standards of treatment and recent progress in respiratory care, introduced in most settings in the 1990s, have improved quality of life and median life expectancy to 4th decade of patient's age. At the same time, cardiac causes of death related to DMD increases. Despite preventive and palliative cardiac treatments available, the prognoses remain poor. Direct therapeutic targeting of dystrophin deficiency is critical, however, hindered by the large size of the dystrophin cDNA and/or stochastic, often extensive genetic changes in DMD gene. The correlation between cardiac involvement and mutations affecting specific dystrophin isoforms, may provide a mutation-specific cardiac management and novel therapeutic approaches for patients with CM. Nonetheless, the successful cardiac treatment poses a big challenge and may require combined therapy to combat dystrophin deficiency and its after-effects (critical in DMD pathogenesis). This review locates the multifaceted heart problem in the course of DMD, balancing the insights into basic science, translational efforts and clinical manifestation of dystrophic heart disease.
Collapse
Affiliation(s)
- Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
16
|
Cappella M, Elouej S, Biferi MG. The Potential of Induced Pluripotent Stem Cells to Test Gene Therapy Approaches for Neuromuscular and Motor Neuron Disorders. Front Cell Dev Biol 2021; 9:662837. [PMID: 33937264 PMCID: PMC8080375 DOI: 10.3389/fcell.2021.662837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
The reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) represents a major advance for the development of human disease models. The emerging of this technique fostered the concept of "disease in a dish," which consists into the generation of patient-specific models in vitro. Currently, iPSCs are used to study pathological molecular mechanisms caused by genetic mutations and they are considered a reliable model for high-throughput drug screenings. Importantly, precision-medicine approaches to treat monogenic disorders exploit iPSCs potential for the selection and validation of lead candidates. For example, antisense oligonucleotides (ASOs) were tested with promising results in myoblasts or motor neurons differentiated from iPSCs of patients affected by either Duchenne muscular dystrophy or Amyotrophic lateral sclerosis. However, the use of iPSCs needs additional optimization to ensure translational success of the innovative strategies based on gene delivery through adeno associated viral vectors (AAV) for these diseases. Indeed, to establish an efficient transduction of iPSCs with AAV, several aspects should be optimized, including viral vector serotype, viral concentration and timing of transduction. This review will outline the use of iPSCs as a model for the development and testing of gene therapies for neuromuscular and motor neuron disorders. It will then discuss the advantages for the use of this versatile tool for gene therapy, along with the challenges associated with the viral vector transduction of iPSCs.
Collapse
Affiliation(s)
- Marisa Cappella
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, Paris, France
| | - Sahar Elouej
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, Paris, France
| | - Maria Grazia Biferi
- Sorbonne University, INSERM, Institute of Myology, Center of Research in Myology, Paris, France
| |
Collapse
|
17
|
Starosta A, Konieczny P. Therapeutic aspects of cell signaling and communication in Duchenne muscular dystrophy. Cell Mol Life Sci 2021; 78:4867-4891. [PMID: 33825942 PMCID: PMC8233280 DOI: 10.1007/s00018-021-03821-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/26/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating chromosome X-linked disease that manifests predominantly in progressive skeletal muscle wasting and dysfunctions in the heart and diaphragm. Approximately 1/5000 boys and 1/50,000,000 girls suffer from DMD, and to date, the disease is incurable and leads to premature death. This phenotypic severity is due to mutations in the DMD gene, which result in the absence of functional dystrophin protein. Initially, dystrophin was thought to be a force transducer; however, it is now considered an essential component of the dystrophin-associated protein complex (DAPC), viewed as a multicomponent mechanical scaffold and a signal transduction hub. Modulating signal pathway activation or gene expression through epigenetic modifications has emerged at the forefront of therapeutic approaches as either an adjunct or stand-alone strategy. In this review, we propose a broader perspective by considering DMD to be a disease that affects myofibers and muscle stem (satellite) cells, as well as a disorder in which abrogated communication between different cell types occurs. We believe that by taking this systemic view, we can achieve safe and holistic treatments that can restore correct signal transmission and gene expression in diseased DMD tissues.
Collapse
Affiliation(s)
- Alicja Starosta
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland
| | - Patryk Konieczny
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| |
Collapse
|
18
|
The Role of Z-disc Proteins in Myopathy and Cardiomyopathy. Int J Mol Sci 2021; 22:ijms22063058. [PMID: 33802723 PMCID: PMC8002584 DOI: 10.3390/ijms22063058] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/07/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
The Z-disc acts as a protein-rich structure to tether thin filament in the contractile units, the sarcomeres, of striated muscle cells. Proteins found in the Z-disc are integral for maintaining the architecture of the sarcomere. They also enable it to function as a (bio-mechanical) signalling hub. Numerous proteins interact in the Z-disc to facilitate force transduction and intracellular signalling in both cardiac and skeletal muscle. This review will focus on six key Z-disc proteins: α-actinin 2, filamin C, myopalladin, myotilin, telethonin and Z-disc alternatively spliced PDZ-motif (ZASP), which have all been linked to myopathies and cardiomyopathies. We will summarise pathogenic variants identified in the six genes coding for these proteins and look at their involvement in myopathy and cardiomyopathy. Listing the Minor Allele Frequency (MAF) of these variants in the Genome Aggregation Database (GnomAD) version 3.1 will help to critically re-evaluate pathogenicity based on variant frequency in normal population cohorts.
Collapse
|
19
|
Matsumoto K, Luther KB, Haltiwanger RS. Diseases related to Notch glycosylation. Mol Aspects Med 2020; 79:100938. [PMID: 33341260 DOI: 10.1016/j.mam.2020.100938] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 12/15/2022]
Abstract
The Notch receptors are a family of transmembrane proteins that mediate direct cell-cell interactions and control numerous cell-fate specifications in humans. The extracellular domains of mammalian Notch proteins contain 29-36 tandem epidermal growth factor-like (EGF) repeats, most of which have O-linked glycan modifications: O-glucose added by POGLUT1, O-fucose added by POFUT1 and elongated by Fringe enzymes, and O-GlcNAc added by EOGT. The extracellular domain is also N-glycosylated. Mutations in the glycosyltransferases modifying Notch have been identified in several diseases, including Dowling-Degos Disease (haploinsufficiency of POFUT1 or POGLUT1), a form of limb-girdle muscular dystrophy (autosomal recessive mutations in POGLUT1), Spondylocostal Dysostosis 3 (autosomal recessive mutations in LFNG), Adams-Oliver syndrome (autosomal recessive mutations in EOGT), and some cancers (amplification, gain or loss-of-function of POFUT1, Fringe enzymes, POGLUT1, MGAT3). Here we review the characteristics of these diseases and potential molecular mechanisms.
Collapse
Affiliation(s)
- Kenjiroo Matsumoto
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Kelvin B Luther
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA
| | - Robert S Haltiwanger
- Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA.
| |
Collapse
|
20
|
Combined Cell Therapy in the Treatment of Neurological Disorders. Biomedicines 2020; 8:biomedicines8120613. [PMID: 33333803 PMCID: PMC7765161 DOI: 10.3390/biomedicines8120613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 02/07/2023] Open
Abstract
Cell therapy of neurological diseases is gaining momentum. Various types of stem/progenitor cells and their derivatives have shown positive therapeutic results in animal models of neurological disorders and in clinical trials. Each tested cell type proved to have its advantages and flaws and unique cellular and molecular mechanism of action, prompting the idea to test combined transplantation of two or more types of cells (combined cell therapy). This review summarizes the results of combined cell therapy of neurological pathologies reported up to this point. The number of papers describing experimental studies or clinical trials addressing this subject is still limited. However, its successful application to the treatment of neurological pathologies including stroke, spinal cord injury, neurodegenerative diseases, Duchenne muscular dystrophy, and retinal degeneration has been reported in both experimental and clinical studies. The advantages of combined cell therapy can be realized by simple summation of beneficial effects of different cells. Alternatively, one kind of cells can support the survival and functioning of the other by enhancing the formation of optimum environment or immunomodulation. No significant adverse events were reported. Combined cell therapy is a promising approach for the treatment of neurological disorders, but further research needs to be conducted.
Collapse
|
21
|
Tijore A, Lee BH, Salila Vijayalal Mohan HK, Li H, Tan LP. Bioactive micropatterned platform to engineer myotube-like cells from stem cells. Biofabrication 2020; 13. [PMID: 33285529 DOI: 10.1088/1758-5090/abd157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/07/2020] [Indexed: 11/12/2022]
Abstract
Skeletal muscle has the capacity to repair and heal itself after injury. However, this self-healing ability is diminished in the event of severe injuries and myopathies. In such conditions, stem cell-based regenerative treatments can play an important part in post injury restoration. We herein report the development of a bioactive (integrin-β1 antibody immobilized) gold micropatterned platform to promote human mesenchymal stem cells (hMSCs) differentiation into the myotube-like cells. hMSCs grown on bioactive micropattern differentiated into the myotube-like cells within two weeks. Further, up-regulation of myogenic markers, multi-nucleated state with continuous actin cytoskeleton and absence of proliferation marker confirmed the formation of myotube-like cells on bioactive micropattern. Prominent expression of elongated integrin-β1 focal adhesions (ITG-β1 FAs) and development of anisotropic stress fibres in those differentiated cells elucidated their importance in stem cell myogenesis. Together these findings delineate the synergistic role of engineered cell anisotropy and ITG-β1 mediated signaling in the development of myotube-like cells from hMSCs.
Collapse
Affiliation(s)
- Ajay Tijore
- National University of Singapore, Mechanobiology Institute, Singapore, 119260, SINGAPORE
| | - Bae Hoon Lee
- Nanyang Technological University, School of Materials Science and Engineering, Singapore, Singapore, 639798, SINGAPORE
| | | | - Holden Li
- Nanyang Technological University, School of Mechanical and Aerospace Engineering, Singapore, Singapore, 639798, SINGAPORE
| | - Lay Poh Tan
- Nanyang Technological University, School of Materials Science and Engineering, Singapore, Singapore, 639798, SINGAPORE
| |
Collapse
|
22
|
Péladeau C, Jasmin BJ. Targeting IRES-dependent translation as a novel approach for treating Duchenne muscular dystrophy. RNA Biol 2020; 18:1238-1251. [PMID: 33164678 DOI: 10.1080/15476286.2020.1847894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Internal-ribosomal entry sites (IRES) are translational elements that allow the initiation machinery to start protein synthesis via internal initiation. IRESs promote tissue-specific translation in stress conditions when conventional cap-dependent translation is inhibited. Since many IRES-containing mRNAs are relevant to diseases, this cellular mechanism is emerging as an attractive therapeutic target for pharmacological and genetic modulations. Indeed, there has been growing interest over the past years in determining the therapeutic potential of IRESs for several disease conditions such as cancer, neurodegeneration and neuromuscular diseases including Duchenne muscular dystrophy (DMD). IRESs relevant for DMD have been identified in several transcripts whose protein product results in functional improvements in dystrophic muscles. Together, these converging lines of evidence indicate that activation of IRES-mediated translation of relevant transcripts in DMD muscle represents a novel and appropriate therapeutic strategy for DMD that warrants further investigation, particularly to identify agents that can modulate their activity.
Collapse
Affiliation(s)
- Christine Péladeau
- Department of Cellular and Molecular Medicine, and the Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, and the Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
23
|
Priester C, MacDonald A, Dhar M, Bow A. Examining the Characteristics and Applications of Mesenchymal, Induced Pluripotent, and Embryonic Stem Cells for Tissue Engineering Approaches across the Germ Layers. Pharmaceuticals (Basel) 2020; 13:E344. [PMID: 33114710 PMCID: PMC7692540 DOI: 10.3390/ph13110344] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
The field of regenerative medicine utilizes a wide array of technologies and techniques for repairing and restoring function to damaged tissues. Among these, stem cells offer one of the most potent and promising biological tools to facilitate such goals. Implementation of mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) offer varying advantages based on availability and efficacy in the target tissue. The focus of this review is to discuss characteristics of these three subset stem cell populations and examine their utility in tissue engineering. In particular, the development of therapeutics that utilize cell-based approaches, divided by germinal layer to further assess research targeting specific tissues of the mesoderm, ectoderm, and endoderm. The combinatorial application of MSCs, iPSCs, and ESCs with natural and synthetic scaffold technologies can enhance the reparative capacity and survival of implanted cells. Continued efforts to generate more standardized approaches for these cells may provide improved study-to-study variations on implementation, thereby increasing the clinical translatability of cell-based therapeutics. Coupling clinically translatable research with commercially oriented methods offers the potential to drastically advance medical treatments for multiple diseases and injuries, improving the quality of life for many individuals.
Collapse
Affiliation(s)
- Caitlin Priester
- Department of Animal Science, University of Tennessee, Knoxville, TN 37998, USA;
| | - Amber MacDonald
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Madhu Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| | - Austin Bow
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, 2407 River Drive, Knoxville, TN 37996, USA; (A.M.); (M.D.)
| |
Collapse
|
24
|
Xu D, Zhao L, Jiang J, Li S, Sun Z, Huang X, Li C, Wang T, Sun L, Li X, Jiang Z, Zhang L. A potential therapeutic effect of catalpol in Duchenne muscular dystrophy revealed by binding with TAK1. J Cachexia Sarcopenia Muscle 2020; 11:1306-1320. [PMID: 32869445 PMCID: PMC7567147 DOI: 10.1002/jcsm.12581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/29/2020] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a progressive muscle disease caused by the loss of dystrophin, which results in inflammation, fibrosis, and the inhibition of myoblast differentiation in skeletal muscle. Catalpol, an iridoid glycoside, improves skeletal muscle function by enhancing myogenesis; it has potential to treat DMD. We demonstrate the positive effects of catalpol in dystrophic skeletal muscle. METHODS mdx (loss of dystrophin) mice (n = 18 per group) were treated with catalpol (200 mg/kg) for six consecutive weeks. Serum analysis, skeletal muscle performance and histology, muscle contractile function, and gene and protein expression were performed. Molecular docking and ligand-target interactions, RNA interference, immunofluorescence, and plasmids transfection were utilized to explore the protective mechanism in DMD by which catalpol binding with transforming growth factor-β-activated kinase 1 (TAK1) in skeletal muscle. RESULTS Six weeks of catalpol treatment improved whole-body muscle health in mdx mice, which was characterized by reduced plasma creatine kinase (n = 18, -35.1%, P < 0.05) and lactic dehydrogenase (n = 18, -10.3%, P < 0.05) activity. These effects were accompanied by enhanced grip strength (n = 18, +25.4%, P < 0.05) and reduced fibrosis (n = 18, -29.0% for hydroxyproline content, P < 0.05). Moreover, catalpol treatment protected against muscle fatigue and promoted muscle recovery in the tibialis anterior (TA) and diaphragm (DIA) muscles (n = 6, +69.8%, P < 0.05 and + 74.8%, P < 0.001, respectively), which was accompanied by enhanced differentiation in primary myoblasts from DMD patients (n = 6, male, mean age: 4.7 ± 1.9 years) and mdx mice. In addition, catalpol eliminated p-TAK1 overexpression in mdx mice (n = 12, -21.3%, P < 0.05) and primary myoblasts. The catalpol-induced reduction in fibrosis and increased myoblast differentiation resulted from the inhibition of TAK1 phosphorylation, leading to reduced myoblast trans-differentiation into myofibroblasts. Catalpol inhibited the phosphorylation of TAK1 by binding to TAK1, possibly at Asp-206, Thr-208, Asn-211, Glu-297, Lys-294, and Tyr-293. CONCLUSIONS Our findings show that catalpol and TAK1 inhibitors substantially improve whole-body muscle health and the function of dystrophic skeletal muscles and may provide a novel therapy for DMD.
Collapse
Affiliation(s)
- Dengqiu Xu
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Lei Zhao
- Department of NeurologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Jingwei Jiang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Sijia Li
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Zeren Sun
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Xiaofei Huang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Chunjie Li
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Tao Wang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Jiangsu Center for Pharmacodynamics Research and EvaluationChina Pharmaceutical UniversityNanjingChina
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| | - Xihua Li
- Department of NeurologyChildren's Hospital of Fudan UniversityShanghaiChina
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Key Laboratory of Drug Quality Control and PharmacovigilanceChina Pharmaceutical UniversityNanjingChina
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Center for Drug Research and DevelopmentGuangdong Pharmaceutical UniversityGuangzhouChina
- Key Laboratory of Drug Quality Control and PharmacovigilanceChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
25
|
May V, Arnold AA, Pagad S, Somagutta MR, Sridharan S, Nanthakumaran S, Malik BH. Duchenne's Muscular Dystrophy: The Role of Induced Pluripotent Stem Cells and Genomic Editing on Muscle Regeneration. Cureus 2020; 12:e10600. [PMID: 33123420 PMCID: PMC7584317 DOI: 10.7759/cureus.10600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
There are two types of well-known muscular dystrophies: Duchenne's muscular dystrophy (DMD) and Becker's muscular dystrophy. This article focuses on the X-linked recessive disorder of Duchenne's muscular dystrophy, which primarily affects children at age four, with a shortened life span of up to 40 years. A defective dystrophin protein lacking the gene dystrophin is the primary cause of the disease pathophysiology. This defect causes cardiac and skeletal muscle down-regulation of dystrophin, leading to weak and fibrotic muscles. The disease is currently untreatable, so most kids die due to cardiac failure in their late 30's. This review presents current treatment options, based on previous studies conducted over the last five years. We used the PubMed database to analyze and review the most important investigations. We also included an analysis of induced pluripotent stem cell therapy vs. genetic therapy using the mdx mouse model. We have discovered promising results on mdx mouse models to date and excited about the potential for where further clinical human trials can go.
Collapse
Affiliation(s)
- Vanessa May
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ashley A Arnold
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sukrut Pagad
- Department of Internal Medicine, Larkin Community Hospital, Hialeah, USA
| | - Manoj R Somagutta
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Saijanakan Sridharan
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Saruja Nanthakumaran
- Department of Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Bilal Haider Malik
- Department of Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
26
|
Sengupta K, Mishra MK, Loro E, Spencer MJ, Pyle AD, Khurana TS. Genome Editing-Mediated Utrophin Upregulation in Duchenne Muscular Dystrophy Stem Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:500-509. [PMID: 33230452 PMCID: PMC7554652 DOI: 10.1016/j.omtn.2020.08.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/25/2020] [Indexed: 12/26/2022]
Abstract
Utrophin upregulation is considered a promising therapeutic strategy for Duchenne muscular dystrophy (DMD). A number of microRNAs (miRNAs) post-transcriptionally regulate utrophin expression by binding their cognate sites in the 3′ UTR. Previously we have shown that miRNA: UTRN repression can be alleviated using miRNA let-7c site blocking oligonucleotides (SBOs) to achieve utrophin upregulation and functional improvement in mdx mice. Here, we used CRISPR/Cas9-mediated genome editing to delete five miRNA binding sites (miR-150, miR-296-5p, miR-133b, let-7c, miR-196b) clustered in a 500 bp inhibitory miRNA target region (IMTR) within the UTRN 3′ UTR, for achieving higher expression of endogenous utrophin. Deleting the UTRN IMTR in DMD patient-derived human induced pluripotent stem cells (DMD-hiPSCs) resulted in ca. 2-fold higher levels of utrophin protein. Differentiation of the UTRN edited DMD-hiPSCs (UTRNΔIMTR) by MyoD overexpression resulted in increased sarcolemmal α-sarcoglycan staining consistent with improved dystrophin glycoprotein complex (DGC) restoration. These results demonstrate that CRISPR/Cas9-based UTRN genome editing offers a novel utrophin upregulation therapeutic strategy applicable to all DMD patients, irrespective of the dystrophin mutation status.
Collapse
Affiliation(s)
- Kasturi Sengupta
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Manoj K Mishra
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emanuele Loro
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Melissa J Spencer
- Molecular Biology Institute, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Department of Neurology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - April D Pyle
- Molecular Biology Institute, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.,Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tejvir S Khurana
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
27
|
Zullo L, Bozzo M, Daya A, Di Clemente A, Mancini FP, Megighian A, Nesher N, Röttinger E, Shomrat T, Tiozzo S, Zullo A, Candiani S. The Diversity of Muscles and Their Regenerative Potential across Animals. Cells 2020; 9:cells9091925. [PMID: 32825163 PMCID: PMC7563492 DOI: 10.3390/cells9091925] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
Cells with contractile functions are present in almost all metazoans, and so are the related processes of muscle homeostasis and regeneration. Regeneration itself is a complex process unevenly spread across metazoans that ranges from full-body regeneration to partial reconstruction of damaged organs or body tissues, including muscles. The cellular and molecular mechanisms involved in regenerative processes can be homologous, co-opted, and/or evolved independently. By comparing the mechanisms of muscle homeostasis and regeneration throughout the diversity of animal body-plans and life cycles, it is possible to identify conserved and divergent cellular and molecular mechanisms underlying muscle plasticity. In this review we aim at providing an overview of muscle regeneration studies in metazoans, highlighting the major regenerative strategies and molecular pathways involved. By gathering these findings, we wish to advocate a comparative and evolutionary approach to prompt a wider use of “non-canonical” animal models for molecular and even pharmacological studies in the field of muscle regeneration.
Collapse
Affiliation(s)
- Letizia Zullo
- Istituto Italiano di Tecnologia, Center for Micro-BioRobotics & Center for Synaptic Neuroscience and Technology (NSYN), 16132 Genova, Italy;
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Correspondence: (L.Z.); (A.Z.)
| | - Matteo Bozzo
- Laboratory of Developmental Neurobiology, Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy; (M.B.); (S.C.)
| | - Alon Daya
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel; (A.D.); (N.N.); (T.S.)
| | - Alessio Di Clemente
- Istituto Italiano di Tecnologia, Center for Micro-BioRobotics & Center for Synaptic Neuroscience and Technology (NSYN), 16132 Genova, Italy;
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 3, 16132 Genova, Italy
| | | | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy;
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy
| | - Nir Nesher
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel; (A.D.); (N.N.); (T.S.)
| | - Eric Röttinger
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, CNRS, INSERM, 06107 Nice, France;
| | - Tal Shomrat
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret 40297, Israel; (A.D.); (N.N.); (T.S.)
| | - Stefano Tiozzo
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer (LBDV), Sorbonne Université, CNRS, 06230 Paris, France;
| | - Alberto Zullo
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy;
- Correspondence: (L.Z.); (A.Z.)
| | - Simona Candiani
- Laboratory of Developmental Neurobiology, Department of Earth, Environment and Life Sciences, University of Genova, Viale Benedetto XV 5, 16132 Genova, Italy; (M.B.); (S.C.)
| |
Collapse
|
28
|
The approved gene therapy drugs worldwide: from 1998 to 2019. Biotechnol Adv 2020; 40:107502. [PMID: 31887345 DOI: 10.1016/j.biotechadv.2019.107502] [Citation(s) in RCA: 224] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
|
29
|
Gogou M, Pavlou E, Haidopoulou K. Therapies that are available and under development for Duchenne muscular dystrophy: What about lung function? Pediatr Pulmonol 2020; 55:300-315. [PMID: 31834673 DOI: 10.1002/ppul.24605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Respiratory failure is the principal source of morbidity and mortality among patients with Duchenne muscular dystrophy exerting a negative influence on their total quality of life. The aim of this review is to provide systematically current literature evidence about the effects of different treatment options (available or under development) for Duchenne muscular dystrophy on the pulmonary function of these patients. METHODS A comprehensive search was undertaken using multiple health-related databases, while two independent reviewers assessed the eligibility of studies. A third person addressed any disagreements between reviewers. The quality of the methodology of the included studies was also assessed. RESULTS A total of 19 original research papers (nine evaluating the role of steroids, six idebenone, three eteplirsen, one stem-cell therapy, and one ataluren) were found to fulfill our selection criteria with the majority of them (14 of 19) being prospective studies, not always including a control group. Endpoints mainly used in these studies were values of pulmonary function tests. Current and under development treatments proved to be safe and no significant adverse events were reported. A beneficial impact on pulmonary function was described by authors in the majority of these studies. The principal effect was slowing of lung disease progress, as expressed by spirometric values. However, the risk of bias was introduced in many of the above studies, while high heterogeneity in terms of treatment protocols and outcome measures limits the comparability of the results. CONCLUSION Glucocorticoids remain the best-studied pharmacologic therapy for Duchenne muscular dystrophy and very likely delay the expected decline in lung function. With regard to new therapeutic agents, initial study results are encouraging. However, larger clinical trials are needed that minimize the risk of study bias, optimize the comparability of treatment groups, examine clinically meaningful pulmonary outcome measures, and include long-term follow up.
Collapse
Affiliation(s)
- Maria Gogou
- Second Department of Pediatrics, University General Hospital AHEPA, Thessaloniki, Greece
| | - Evangelos Pavlou
- Second Department of Pediatrics, University General Hospital AHEPA, Thessaloniki, Greece
| | - Katerina Haidopoulou
- Second Department of Pediatrics, University General Hospital AHEPA, Thessaloniki, Greece
| |
Collapse
|
30
|
Lazzarin MC, Quintana HT, Baptista VIDA, Oliveira FD. Lack of dystrophin influences muscle inflammation but not myogenic regulatory factors after eccentric exercise in mdx mice. MOTRIZ: REVISTA DE EDUCACAO FISICA 2020. [DOI: 10.1590/s1980-6574202000030228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
31
|
Gois Beghini D, Iwao Horita S, Monteiro da Fonseca Cardoso L, Anastacio Alves L, Nagaraju K, Henriques-Pons A. A Promising Future for Stem-Cell-Based Therapies in Muscular Dystrophies-In Vitro and In Vivo Treatments to Boost Cellular Engraftment. Int J Mol Sci 2019; 20:ijms20215433. [PMID: 31683627 PMCID: PMC6861917 DOI: 10.3390/ijms20215433] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/28/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023] Open
Abstract
Muscular dystrophies (MD) are a group of genetic diseases that lead to skeletal muscle wasting and may affect many organs (multisystem). Unfortunately, no curative therapies are available at present for MD patients, and current treatments mainly address the symptoms. Thus, stem-cell-based therapies may present hope for improvement of life quality and expectancy. Different stem cell types lead to skeletal muscle regeneration and they have potential to be used for cellular therapies, although with several limitations. In this review, we propose a combination of genetic, biochemical, and cell culture treatments to correct pathogenic genetic alterations and to increase proliferation, dispersion, fusion, and differentiation into new or hybrid myotubes. These boosted stem cells can also be injected into pretreate recipient muscles to improve engraftment. We believe that this combination of treatments targeting the limitations of stem-cell-based therapies may result in safer and more efficient therapies for MD patients. Matricryptins have also discussed.
Collapse
Affiliation(s)
- Daniela Gois Beghini
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | - Samuel Iwao Horita
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | | | - Luiz Anastacio Alves
- Laboratório de Comunicação Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| | - Kanneboyina Nagaraju
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York, NY 13902, USA.
| | - Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro (RJ) 21040-900, Brazil.
| |
Collapse
|
32
|
Mueller AL, Bloch RJ. Skeletal muscle cell transplantation: models and methods. J Muscle Res Cell Motil 2019; 41:297-311. [PMID: 31392564 DOI: 10.1007/s10974-019-09550-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023]
Abstract
Xenografts of skeletal muscle are used to study muscle repair and regeneration, mechanisms of muscular dystrophies, and potential cell therapies for musculoskeletal disorders. Typically, xenografting involves using an immunodeficient host that is pre-injured to create a niche for human cell engraftment. Cell type and method of delivery to muscle depend on the specific application, but can include myoblasts, satellite cells, induced pluripotent stem cells, mesangioblasts, immortalized muscle precursor cells, and other multipotent cell lines delivered locally or systemically. Some studies follow cell engraftment with interventions to enhance cell proliferation, migration, and differentiation into mature muscle fibers. Recently, several advances in xenografting human-derived muscle cells have been applied to study and treat Duchenne muscular dystrophy and Facioscapulohumeral muscular dystrophy. Here, we review the vast array of techniques available to aid researchers in designing future experiments aimed at creating robust muscle xenografts in rodent hosts.
Collapse
Affiliation(s)
- Amber L Mueller
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA.
| |
Collapse
|
33
|
Chiappalupi S, Salvadori L, Luca G, Riuzzi F, Calafiore R, Donato R, Sorci G. Do porcine Sertoli cells represent an opportunity for Duchenne muscular dystrophy? Cell Prolif 2019; 52:e12599. [PMID: 30912260 PMCID: PMC6536415 DOI: 10.1111/cpr.12599] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/24/2019] [Accepted: 02/09/2019] [Indexed: 12/13/2022] Open
Abstract
Sertoli cells (SeC) are responsible for the immunoprivileged status of the testis thanks to which allogeneic or xenogeneic engraftments can survive without pharmacological immune suppression if co‐injected with SeC. This peculiar ability of SeC is dependent on secretion of a plethora of factors including maturation factors, hormones, growth factors, cytokines and immunomodulatory factors. The anti‐inflammatory and trophic properties of SeC have been largely exploited in several experimental models of diseases, diabetes being the most studied. Duchenne muscular dystrophy (DMD) is a lethal X‐linked recessive pathology in which lack of functional dystrophin leads to progressive muscle degeneration culminating in loss of locomotion and premature death. Despite a huge effort to find a cure, DMD patients are currently treated with anti‐inflammatory steroids. Recently, encapsulated porcine SeC (MC‐SeC) have been injected ip in the absence of immunosuppression in an animal model of DMD resulting in reduction of muscle inflammation and amelioration of muscle morphology and functionality, thus opening an additional avenue in the treatment of DMD. The novel protocol is endowed with the advantage of being potentially applicable to all the cohort of DMD patients regardless of the mutation. This mini‐review addresses several issues linked to the possible use of MC‐SeC injected ip in dystrophic people.
Collapse
Affiliation(s)
- Sara Chiappalupi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy
| | - Laura Salvadori
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Francesca Riuzzi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy
| | | | - Rosario Donato
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy.,Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia, Italy
| | - Guglielmo Sorci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology (IIM), Perugia, Italy.,Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia, Italy
| |
Collapse
|