1
|
Charrier M, Leroux I, Pichon J, Schleder C, Larcher T, Hamel A, Magot A, Péréon Y, Lamirault G, Tremblay JP, Skuk D, Rouger K. Human MuStem cells are competent to fuse with nonhuman primate myofibers in a clinically relevant transplantation context: A proof-of-concept study. J Neuropathol Exp Neurol 2024; 83:684-694. [PMID: 38752570 DOI: 10.1093/jnen/nlae044] [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] [Indexed: 07/20/2024] Open
Abstract
We previously reported that human muscle-derived stem cells (hMuStem cells) contribute to tissue repair after local administration into injured skeletal muscle or infarcted heart in immunodeficient rodent models. However, extrapolation of these findings to a clinical context is problematic owing to the considerable differences often seen between in vivo findings in humans versus rodents. Therefore, we investigated whether the muscle regenerative behavior of hMuStem cells is maintained in a clinically relevant transplantation context. Human MuStem cells were intramuscularly administered by high-density microinjection matrices into nonhuman primates receiving tacrolimus-based immunosuppression thereby reproducing the protocol that has so far produced the best results in clinical trials of cell therapy in myopathies. Four and 9 weeks after administration, histological analysis of cell injection sites revealed large numbers of hMuStem cell-derived nuclei in all cases. Most graft-derived nuclei were distributed in small myofiber groups in which no signs of a specific immune response were observed. Importantly, hMuStem cells contributed to simian tissue repair by fusing mainly with host myofibers, demonstrating their capacity for myofiber regeneration in this model. Together, these findings obtained in a valid preclinical model provide new insights supporting the potential of hMuStem cells in future cell therapies for muscle diseases.
Collapse
Affiliation(s)
- Marine Charrier
- Oniris, INRAE, PAnTher, Nantes, France
- L'institut du Thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
- Nantes Université, Nantes, France
| | | | | | | | | | - Antoine Hamel
- Service de Chirurgie Infantile, Centre Hospitalier Universitaire (CHU) de Nantes, Nantes, France
| | - Armelle Magot
- Centre de Référence Maladies Neuromusculaires AOC, Filnemus, Euro-NMD, Laboratoire d'Explorations Fonctionnelles, Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | - Yann Péréon
- Centre de Référence Maladies Neuromusculaires AOC, Filnemus, Euro-NMD, Laboratoire d'Explorations Fonctionnelles, Centre Hospitalier Universitaire Hôtel Dieu, Nantes, France
| | | | - Jacques P Tremblay
- Axe Neurosciences, Research Center of the CHU de Quebec-CHUL and Department of Molecular Medicine, School of Medicine, Laval University, Quebec, Quebec, Canada
| | - Daniel Skuk
- Axe Neurosciences, Research Center of the CHU de Quebec-CHUL and Department of Molecular Medicine, School of Medicine, Laval University, Quebec, Quebec, Canada
| | | |
Collapse
|
2
|
Musgrove L, Russell FD, Ventura T. Considerations for cultivated crustacean meat: potential cell sources, potential differentiation and immortalization strategies, and lessons from crustacean and other animal models. Crit Rev Food Sci Nutr 2024:1-25. [PMID: 38733287 DOI: 10.1080/10408398.2024.2342480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2024]
Abstract
Cultivated crustacean meat (CCM) is a means to create highly valued shrimp, lobster, and crab products directly from stem cells, thus removing the need to farm or fish live animals. Conventional crustacean enterprises face increasing pressures in managing overfishing, pollution, and the warming climate, so CCM may provide a way to ensure sufficient supply as global demand for these products grows. To support the development of CCM, this review briefly details crustacean cell culture work to date, before addressing what is presently known about crustacean muscle development, particularly the molecular mechanisms involved, and how this might relate to recent work on cultivated meat production in vertebrate species. Recognizing the current lack of cell lines available to establish CCM cultures, we also consider primary stem cell sources that can be obtained non-lethally including tissues from limbs which are readily released and regrown, and putative stem cells in circulating hemolymph. Molecular approaches to inducing myogenic differentiation and immortalization of putative stem cells are also reviewed. Finally, we assess the current status of tools available to CCM researchers, particularly antibodies, and propose avenues to address existing shortfalls in order to see the field progress.
Collapse
Affiliation(s)
- Lisa Musgrove
- Centre for Bioinnovation, University of the Sunshine Coast (UniSC), Maroochydore, QLD, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast (UniSC), Maroochydore, QLD, Australia
| | - Fraser D Russell
- Centre for Bioinnovation, University of the Sunshine Coast (UniSC), Maroochydore, QLD, Australia
- School of Health, University of the Sunshine Coast (UniSC), Maroochydore, QLD, Australia
| | - Tomer Ventura
- Centre for Bioinnovation, University of the Sunshine Coast (UniSC), Maroochydore, QLD, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast (UniSC), Maroochydore, QLD, Australia
| |
Collapse
|
3
|
Jiang H, Liu B, Lin J, Xue T, Han Y, Lu C, Zhou S, Gu Y, Xu F, Shen Y, Xu L, Sun H. MuSCs and IPCs: roles in skeletal muscle homeostasis, aging and injury. Cell Mol Life Sci 2024; 81:67. [PMID: 38289345 PMCID: PMC10828015 DOI: 10.1007/s00018-023-05096-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024]
Abstract
Skeletal muscle is a highly specialized tissue composed of myofibres that performs crucial functions in movement and metabolism. In response to external stimuli and injuries, a range of stem/progenitor cells, with muscle stem cells or satellite cells (MuSCs) being the predominant cell type, are rapidly activated to repair and regenerate skeletal muscle within weeks. Under normal conditions, MuSCs remain in a quiescent state, but become proliferative and differentiate into new myofibres in response to injury. In addition to MuSCs, some interstitial progenitor cells (IPCs) such as fibro-adipogenic progenitors (FAPs), pericytes, interstitial stem cells expressing PW1 and negative for Pax7 (PICs), muscle side population cells (SPCs), CD133-positive cells and Twist2-positive cells have been identified as playing direct or indirect roles in regenerating muscle tissue. Here, we highlight the heterogeneity, molecular markers, and functional properties of these interstitial progenitor cells, and explore the role of muscle stem/progenitor cells in skeletal muscle homeostasis, aging, and muscle-related diseases. This review provides critical insights for future stem cell therapies aimed at treating muscle-related diseases.
Collapse
Affiliation(s)
- Haiyan Jiang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Junfei Lin
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Tong Xue
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Yimin Han
- Department of Paediatrics, Medical School of Nantong University, Nantong University, Nantong, 226001, People's Republic of China
| | - Chunfeng Lu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China
| | - Feng Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, Nantong, 226001, Jiangsu, People's Republic of China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Lingchi Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, 226001, Jiangsu, People's Republic of China.
| |
Collapse
|
4
|
Roberts TC, Wood MJA, Davies KE. Therapeutic approaches for Duchenne muscular dystrophy. Nat Rev Drug Discov 2023; 22:917-934. [PMID: 37652974 DOI: 10.1038/s41573-023-00775-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 09/02/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a monogenic muscle-wasting disorder and a priority candidate for molecular and cellular therapeutics. Although rare, it is the most common inherited myopathy affecting children and so has been the focus of intense research activity. It is caused by mutations that disrupt production of the dystrophin protein, and a plethora of drug development approaches are under way that aim to restore dystrophin function, including exon skipping, stop codon readthrough, gene replacement, cell therapy and gene editing. These efforts have led to the clinical approval of four exon skipping antisense oligonucleotides, one stop codon readthrough drug and one gene therapy product, with other approvals likely soon. Here, we discuss the latest therapeutic strategies that are under development and being deployed to treat DMD. Lessons from these drug development programmes are likely to have a major impact on the DMD field, but also on molecular and cellular medicine more generally. Thus, DMD is a pioneer disease at the forefront of future drug discovery efforts, with these experimental treatments paving the way for therapies using similar mechanisms of action being developed for other genetic diseases.
Collapse
Affiliation(s)
- Thomas C Roberts
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
- MDUK Oxford Neuromuscular Centre, Oxford, UK.
| | - Matthew J A Wood
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
- MDUK Oxford Neuromuscular Centre, Oxford, UK
| | - Kay E Davies
- MDUK Oxford Neuromuscular Centre, Oxford, UK.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
5
|
De Paolis F, Testa S, Guarnaccia G, Reggio A, Fornetti E, Cicciarelli F, Deodati R, Bernardini S, Peluso D, Baldi J, Biagini R, Bellisari FC, Izzo A, Sgalambro F, Arrigoni F, Rizzo F, Cannata S, Sciarra T, Fuoco C, Gargioli C. Long-term longitudinal study on swine VML model. Biol Direct 2023; 18:42. [PMID: 37518063 PMCID: PMC10388508 DOI: 10.1186/s13062-023-00399-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND Volumetric Muscle Loss (VML), resulting from severe trauma or surgical ablation, is a pathological condition preventing myofibers regeneration, since skeletal muscle owns the remarkable ability to restore tissue damage, but only when limited in size. The current surgical therapies employed in the treatment of this pathology, which particularly affects military personnel, do not yet provide satisfactory results. For this reason, more innovative approaches must be sought, specifically skeletal muscle tissue engineering seems to highlight promising results obtained from preclinical studies in VML mouse model. Despite the great results obtained in rodents, translation into human needs a comparable animal model in terms of size, in order to validate the efficacy of the tissue engineering approach reconstructing larger muscle mass (human-like). In this work we aim to demonstrate the validity of a porcine model, that has underwent a surgical ablation of a large muscle area, as a VML damage model. RESULTS For this purpose, morphological, ultrasound, histological and fluorescence analyses were carried out on the scar tissue formed following the surgical ablation of the peroneus tertius muscle of Sus scrofa domesticus commonly called mini-pig. In particular, the replenishment of the damaged area, the macrophage infiltration and the vascularization at different time-points were evaluated up to the harvesting of the scar upon six months. CONCLUSION Here we demonstrated that following VML damage, there is an extremely poor regenerative process in the swine muscle tissue, while the formation of fibrotic, scar tissue occurs. The analyses performed up to 180 days after the injury revealed the development of a stable, structured and cellularized tissue, provided with vessels and extracellular matrix acquiring the status of granulation tissue like in human.
Collapse
Affiliation(s)
- Francesca De Paolis
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy
- PhD Program in Cellular and Molecular Biology, Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Stefano Testa
- Marseille Medical Genetics, Aix-Marseille University, INSERM, Marseille, MMG, France
| | | | - Alessio Reggio
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Ersilia Fornetti
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Felice Cicciarelli
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Rebecca Deodati
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Sergio Bernardini
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Daniele Peluso
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Jacopo Baldi
- IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | | | - Antonio Izzo
- Department of Clinical Sciences and Applied Biotechnologies (DISCAB), Aquila, Italy
| | - Ferruccio Sgalambro
- Department of Clinical Sciences and Applied Biotechnologies (DISCAB), Aquila, Italy
| | - Francesco Arrigoni
- Department of Clinical Sciences and Applied Biotechnologies (DISCAB), Aquila, Italy
| | - Francesco Rizzo
- Joint Veteran Center, Scientific Department, Army Medical Center, Rome, Italy
| | - Stefano Cannata
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Tommaso Sciarra
- Joint Veteran Center, Scientific Department, Army Medical Center, Rome, Italy
| | - Claudia Fuoco
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy.
| | - Cesare Gargioli
- Department of Biology, University of Rome "Tor Vergata", Rome, 00133, Italy.
| |
Collapse
|
6
|
Johnson AL, Kamal M, Parise G. The Role of Supporting Cell Populations in Satellite Cell Mediated Muscle Repair. Cells 2023; 12:1968. [PMID: 37566047 PMCID: PMC10417507 DOI: 10.3390/cells12151968] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/12/2023] Open
Abstract
Skeletal muscle has a high capacity to repair and remodel in response to damage, largely through the action of resident muscle stem cells, termed satellite cells. Satellite cells are required for the proper repair of skeletal muscle through a process known as myogenesis. Recent investigations have observed relationships between satellite cells and other cell types and structures within the muscle microenvironment. These findings suggest that the crosstalk between inflammatory cells, fibrogenic cells, bone-marrow-derived cells, satellite cells, and the vasculature is essential for the restoration of muscle homeostasis. This review will discuss the influence of the cells and structures within the muscle microenvironment on satellite cell function and muscle repair.
Collapse
Affiliation(s)
| | | | - Gianni Parise
- Department of Kinesiology, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
7
|
Sokolova AV, Domnina AP, Mikhailov VM. Accumulation of Dystrophin-Positive Muscle Fibers and Improvement of Neuromuscular Junctions in mdx Mouse Muscles after Bone Marrow Transplantation under Different Conditions. Int J Mol Sci 2023; 24:ijms24108892. [PMID: 37240237 DOI: 10.3390/ijms24108892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/13/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscular disorder caused by mutations in the dystrophin gene. It leads to respiratory and cardiac failure and premature death at a young age. Although recent studies have greatly deepened the understanding of the primary and secondary pathogenetic mechanisms of DMD, an effective treatment remains elusive. In recent decades, stem cells have emerged as a novel therapeutic product for a variety of diseases. In this study, we investigated nonmyeloablative bone marrow cell (BMC) transplantation as a method of cell therapy for DMD in an mdx mouse model. By using BMC transplantation from GFP-positive mice, we confirmed that BMCs participate in the muscle restoration of mdx mice. We analyzed both syngeneic and allogeneic BMC transplantation under different conditions. Our data indicated that 3 Gy X-ray irradiation with subsequent BMC transplantation improved dystrophin synthesis and the structure of striated muscle fibers (SMFs) in mdx mice as well as decreasing the death rate of SMFs. In addition, we observed the normalization of neuromuscular junctions (NMJs) in mdx mice after nonmyeloablative BMC transplantation. In conclusion, we demonstrated that nonmyeloablative BMC transplantation could be considered a method for DMD treatment.
Collapse
Affiliation(s)
| | - Alisa P Domnina
- Institute of Cytology, Russian Academy of Sciences, 194064 Saint-Petersburg, Russia
| | | |
Collapse
|
8
|
Rieux M, Alpaugh M, Salem S, Siddu A, Saint-Pierre M, Denis HL, Rohweder H, Herrmann F, Bazenet C, Lacroix S, Cicchetti F. Understanding the role of the hematopoietic niche in Huntington's disease's phenotypic expression: in vivo evidence using a parabiosis model. Neurobiol Dis 2023; 180:106091. [PMID: 36967065 DOI: 10.1016/j.nbd.2023.106091] [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: 01/19/2023] [Revised: 03/06/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
In a previous study, we have shown that parabiotic coupling of a knock-in mouse model (zQ175) of Huntington's disease (HD) to wild-type (WT) littermates resulted in a worsening of the normal phenotype as seen by detection of mutant huntingtin protein (mHTT) aggregates within peripheral organs and the cerebral cortex as well as vascular abnormalities in WT mice. In contrast, parabiosis improved disease features in the zQ175 mice such as reduction of mHTT aggregate number in the liver and cortex, decrease in blood-brain barrier (BBB) permeability and attenuation of mitochondrial impairments. While the shared circulation mediated these effects, no specific factor was identified. To better understand which blood elements were involved in the aforementioned changes, WT and zQ175 mice underwent parabiotic surgery prior to exposing one of the paired animals to irradiation. The irradiation procedure successfully eliminated the hematopoietic niche followed by repopulation with cells originating from the non-irradiated parabiont, as measured by the quantification of mHTT levels in peripheral blood mononuclear cells. Although irradiation of the WT parabiont, causing the loss of healthy hematopoietic cells, did lead to a few alterations in mitochondrial function in the muscle (TOM40 levels), and increased neuroinflammation in the striatum (GFAP levels), most of the changes observed were likely attributable to the irradiation procedure itself (e.g. mHTT aggregates in cortex and liver; cellular stress in peripheral organs). However, factors such as mHTT aggregation in the brain and periphery, and BBB leakage, which were improved in zQ175 mice when paired to WT littermates in the previous parabiosis experiment, were unaffected by perturbation of the hematopoietic niche. It would therefore appear that cells of the hematopoietic stem cell niche are largely uninvolved in the beneficial effects of parabiosis.
Collapse
Affiliation(s)
- Marie Rieux
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de médecine moléculaire, Université Laval, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Melanie Alpaugh
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de psychiatrie & neurosciences, Université Laval, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Shireen Salem
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de médecine moléculaire, Université Laval, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Alberto Siddu
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de psychiatrie & neurosciences, Université Laval, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Martine Saint-Pierre
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Hélèna L Denis
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de psychiatrie & neurosciences, Université Laval, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | | | | | | | - Steve Lacroix
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de médecine moléculaire, Université Laval, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada
| | - Francesca Cicchetti
- Centre de recherche du CHU de Québec - Université Laval, Axe neurosciences, 2705 Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de médecine moléculaire, Université Laval, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada; Département de psychiatrie & neurosciences, Université Laval, 1050 avenue de la Médecine, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
9
|
Zhang HL, Li Z, Cheng QS, Chen X, Zhang C, Zeng T. In vitro myogenesis activation of specific muscle-derived stem cells from patients with Duchenne muscular dystrophy. Transpl Immunol 2023; 77:101796. [PMID: 36764333 DOI: 10.1016/j.trim.2023.101796] [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: 03/22/2022] [Revised: 01/12/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023]
Abstract
BACKGROUND Muscle-derived stem cells (MDSCs) contribute to the repair of injured muscles. However, the myogenicity of MDSCs generated from patients with Duchenne muscular dystrophy (DMD) relative to healthy individuals remains unclear. METHODS A human DMD model was established using the stem cells prepared from muscle derived from patients with DMD (DMD-hMDSCs). The expression of myogenic lineage-specific markers in MDSCs was examined with immunofluorescence, real-time polymerase chain reaction, and western blotting. RESULTS It was demonstrated that, compared with cells from healthy subjects, DMD-hMDSCs are primed to self-differentiate in growth-inducing medium (GM) and robustly differentiate into myotubes in differentiation-inducing medium(DM). This feature was termed "myogenesis activation," and it was speculated that it contributes to the depletion of myogenic progenitors. Furthermore, MDSCs consistently express pax7, but the time-course of this expression does not correlate with the expression of the myogenic lineage-specific markers. CONCLUSIONS The myogenesis activation in DMD-hMDSCs demonstrated in this study may provide novel mechanistic insights into DMD pathogenesis and potential therapies.
Collapse
Affiliation(s)
- Hui-Li Zhang
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; Department of Neurology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China.
| | - Ze Li
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; Department of Neurology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China
| | - Qiu-Sheng Cheng
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; Department of Neurology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China
| | - Xi Chen
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; Department of Neurology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510180, China
| | - Tao Zeng
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China; Department of Neurology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510180, China.
| |
Collapse
|
10
|
Hsiao Y, Wang I, Yang T. Fibrotic remodeling and tissue regeneration mechanisms define the therapeutic potential of human muscular progenitors. Bioeng Transl Med 2023; 8:e10439. [PMID: 36925693 PMCID: PMC10013817 DOI: 10.1002/btm2.10439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 11/27/2022] Open
Abstract
Fibrosis is an intrinsic biological reaction toward the challenges of tissue injury that is implicated in the wound-healing process. Although it is useful to efficiently mitigate the damage, progression of fibrosis is responsible for the morbidity and mortality occurring in a variety of diseases. Because of lacking effective treatments, there is an emerging need for exploring antifibrotic strategies. Cell therapy based on stem/progenitor cells is regarded as a promising approach for treating fibrotic diseases. Appropriate selection of cellular sources is required for beneficial results. Muscle precursor cells (MPCs) are specialized progenitors harvested from skeletal muscle for conducting muscle regeneration. Whether they are also effective in regulating fibrosis has seldom been explored and merits further investigation. MPCs were successfully harvested from all human samples regardless of demographic backgrounds. The extracellular matrices remodeling was enhanced through the paracrine effects mediated by MPCs. The suppression effects on fibrosis were confirmed in vivo when MPCs were transplanted into the diseased animals with oral submucous fibrosis. The data shown here revealed the potential of MPCs to be employed to simultaneously regulate both processes of fibrosis and tissue regeneration, supporting them as the promising cell candidates for development of the cell therapy for antifibrosis and tissue regeneration.
Collapse
Affiliation(s)
- Ya‐Chuan Hsiao
- Department of OphthalmologyTaipei City Hospital, Zhongxing BranchTaipeiTaiwan
- Department of OphthalmologyCollege of Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - I‐Han Wang
- Department of OtolaryngologyNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
| | - Tsung‐Lin Yang
- Department of OtolaryngologyNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan UniversityTaipeiTaiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
11
|
Sahinyan K, Lazure F, Blackburn DM, Soleimani VD. Decline of regenerative potential of old muscle stem cells: contribution to muscle aging. FEBS J 2023; 290:1267-1289. [PMID: 35029021 DOI: 10.1111/febs.16352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/23/2021] [Accepted: 01/11/2022] [Indexed: 01/01/2023]
Abstract
Muscle stem cells (MuSCs) are required for life-long muscle regeneration. In general, aging has been linked to a decline in the numbers and the regenerative potential of MuSCs. Muscle regeneration depends on the proper functioning of MuSCs, which is itself dependent on intricate interactions with its niche components. Aging is associated with both cell-intrinsic and niche-mediated changes, which can be the result of transcriptional, posttranscriptional, or posttranslational alterations in MuSCs or in the components of their niche. The interplay between cell intrinsic alterations in MuSCs and changes in the stem cell niche environment during aging and its impact on the number and the function of MuSCs is an important emerging area of research. In this review, we discuss whether the decline in the regenerative potential of MuSCs with age is the cause or the consequence of aging skeletal muscle. Understanding the effect of aging on MuSCs and the individual components of their niche is critical to develop effective therapeutic approaches to diminish or reverse the age-related defects in muscle regeneration.
Collapse
Affiliation(s)
- Korin Sahinyan
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Felicia Lazure
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Darren M Blackburn
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montréal, QC, Canada.,Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, QC, Canada
| |
Collapse
|
12
|
Ostrovidov S, Ramalingam M, Bae H, Orive G, Fujie T, Shi X, Kaji H. Latest developments in engineered skeletal muscle tissues for drug discovery and development. Expert Opin Drug Discov 2023; 18:47-63. [PMID: 36535280 DOI: 10.1080/17460441.2023.2160438] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION With the advances in skeletal muscle tissue engineering, new platforms have arisen with important applications in biology studies, disease modeling, and drug testing. Current developments highlight the quest for engineering skeletal muscle tissues with higher complexity . These new human skeletal muscle tissue models will be powerful tools for drug discovery and development and disease modeling. AREAS COVERED The authors review the latest advances in in vitro models of engineered skeletal muscle tissues used for testing drugs with a focus on the use of four main cell culture techniques: Cell cultures in well plates, in microfluidics, in organoids, and in bioprinted constructs. Additional information is provided on the satellite cell niche. EXPERT OPINION In recent years, more sophisticated in vitro models of skeletal muscle tissues have been fabricated. Important developments have been made in stem cell research and in the engineering of human skeletal muscle tissue. Some platforms have already started to be used for drug testing, notably those based on the parameters of hypertrophy/atrophy and the contractibility of myotubes. More developments are expected through the use of multicellular types and multi-materials as matrices . The validation and use of these models in drug testing should now increase.
Collapse
Affiliation(s)
- Serge Ostrovidov
- Department of Biomechanics, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science, BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,School of Basic Medical Science, Chengdu University, Chengdu, Sichuan, China.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Republic of Korea.,Department of Metallurgical and Materials Engineering, Atilim University, Ankara, Turkey
| | - Hojae Bae
- KU Convergence Science and Technology Institute, Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Hwayang-dong, Kwangjin-gu, Seoul, Republic of Korea
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Vitoria-Gasteiz, Spain.,Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.,Biomaterials and Nanomedicine (CIBER-BBN), Biomedical Research Networking Centre in Bioengineering, Vitoria-Gasteiz, Spain
| | - Toshinori Fujie
- Department of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Japan
| | - Xuetao Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, China
| | - Hirokazu Kaji
- Department of Biomechanics, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
13
|
Bowles-Welch AC, Jimenez AC, Stevens HY, Frey Rubio DA, Kippner LE, Yeago C, Roy K. Mesenchymal stromal cells for bone trauma, defects, and disease: Considerations for manufacturing, clinical translation, and effective treatments. Bone Rep 2023. [DOI: 10.1016/j.bonr.2023.101656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
14
|
Goodpaster BH, Bergman BC, Brennan AM, Sparks LM. Intermuscular adipose tissue in metabolic disease. Nat Rev Endocrinol 2022; 19:285-298. [PMID: 36564490 DOI: 10.1038/s41574-022-00784-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2022] [Indexed: 12/24/2022]
Abstract
Intermuscular adipose tissue (IMAT) is a distinct adipose depot described in early reports as a 'fatty replacement' or 'muscle fat infiltration' that was linked to ageing and neuromuscular disease. Later studies quantifying IMAT with modern in vivo imaging methods (computed tomography and magnetic resonance imaging) revealed that IMAT is proportionately higher in men and women with type 2 diabetes mellitus and the metabolic syndrome than in people without these conditions and is associated with insulin resistance and poor physical function with ageing. In parallel, agricultural research has provided extensive insight into the role of IMAT and other muscle lipids in muscle (that is, meat) quality. In addition, studies using rodent models have shown that IMAT is a bona fide white adipose tissue depot capable of robust triglyceride storage and turnover. Insight into the importance of IMAT in human biology has been limited by the dearth of studies on its biological properties, that is, the quality of IMAT. However, in the past few years, investigations have begun to determine that IMAT has molecular and metabolic features that distinguish it from other adipose tissue depots. These studies will be critical to further decipher the role of IMAT in health and disease and to better understand its potential as a therapeutic target.
Collapse
Affiliation(s)
| | - Bryan C Bergman
- Division of Endocrinology, Diabetes, and Metabolism, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Andrea M Brennan
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| |
Collapse
|
15
|
Kato S, Kubo T, Fukazawa T. Effective enrichment of stem cells in the regenerating Xenopus laevis tadpole tails using the side population method. Dev Growth Differ 2022; 64:290-296. [PMID: 35839785 DOI: 10.1111/dgd.12797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/27/2022] [Accepted: 07/13/2022] [Indexed: 11/30/2022]
Abstract
Xenopus laevis tadpoles have high regenerative ability and can regenerate their whole tails after tail amputation. Lineage-restricted tissue stem cells are thought to provide sources for the regenerating tissues by producing undifferentiated progenitor cells in response to tail amputation. However, elucidating the behavioral dynamics of tissue stem cells during tail regeneration is difficult because of their rarity, and there are few established methods of isolating these cells in amphibians. Here, to detect and analyze rare tissue stem cells, we attempted to enrich tissue stem cells from tail regeneration buds. High Hoechst dye efflux capacity is thought to be a common characteristic of several types of mammalian tissue stem cells; these stem cells designated as the "side population (SP)" may be enriched by flow cytometry (SP method). To evaluate the effectiveness of stem cell enrichment using the SP method in regenerating X. laevis tadpole tails, we performed single-cell RNA sequencing (scRNA-seq) of SP cells from regeneration buds and analyzed the frequency of satellite cells, which are muscle stem/progenitor cells expressing pax7. The pax7-expressing cells were enriched in the SP compared with whole normal tails and regeneration buds. Furthermore, hes1-expressing cells assumed to be neural stem/progenitor cells were also enriched in the SP. Our findings suggest that the SP method was efficient for successfully enriching tissue stem cells in the regenerating X. laevis tadpole tails, indicating that the combination of the SP method and scRNA-seq was useful for studying tissue stem cells that contribute to tail regeneration.
Collapse
Affiliation(s)
- Sumika Kato
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Takeo Kubo
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Taro Fukazawa
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
16
|
Li J, Shen Z, Chen W, Feng Z, Fang L, Zhao J, Liu C, Du J, Cheng Y. Screening of miRNAs in White Blood Cell as a Radiation Biomarkers for Rapid Assessment of Acute Radiation Injury. Dose Response 2022; 20:15593258221123679. [PMID: 36132708 PMCID: PMC9483971 DOI: 10.1177/15593258221123679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022] Open
Abstract
Accidental radiation exposure is a threat to human health that necessitates
effective clinical diagnosis. Suitable biomarkers are urgently needed for early
assessment of exposure dose. Existing technologies being used to assess the
extent of radiation have notable limitations. As a radiation biomarker, miRNA
has the advantages of simple detection and high throughput. In this study, we
screened for miRNAs with dose and time dependent responses in peripheral blood
leukocytes via miRNA sequencing in establishing the animal model of acute
radiation injury. Four radiation-sensitive and stably expressed miRNAs were
selected out in the 24 h group of leukocyte miRNAs: mmu-miR-130b-5p,
mmu-miR-148b-5p, mmu-miR-184-3p, mmu-miR-26a-2-3p, and five were screened in the
48 h group of leukocyte miRNAs: mmu-miR-130b-5p, mmu-miR-423-5p, mmu-miR-676-3p,
mmu-miR-150-5p, mmu-miR-342-3p.The correlation curves between their expression
and irradiation dose were plotted. Then, the results were validated by RT-qPCR
in mouse peripheral blood. As a result, mmu-miR-150-5p and mmu-miR-342-3p showed
the highest correlation at 48h after irradiation, and mmu-miR-130b-5p showed
good correlation at both 24 h and 48 h after irradiation. In a conclusion, the
miRNAs that are sensitive to ionizing radiation with dose dependent effects were
selected out, which have the potential of forming a rapid assessment scheme for
acute radiation injury.
Collapse
Affiliation(s)
- Jiaxun Li
- College of Basic Medicine, Naval Medical University, Shanghai, China
| | - Zhefan Shen
- College of Basic Medicine, Naval Medical University, Shanghai, China
| | - Wei Chen
- Naval Medical Center, Naval Medical University, Shanghai, China
| | | | - Lan Fang
- Naval Medical University, Shanghai, China
| | | | - Cong Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Jicong Du
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Ying Cheng
- Department of Radiation Medicine, Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| |
Collapse
|
17
|
Pathophysiology and Emerging Molecular Therapeutic Targets in Heterotopic Ossification. Int J Mol Sci 2022; 23:ijms23136983. [PMID: 35805978 PMCID: PMC9266941 DOI: 10.3390/ijms23136983] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 12/23/2022] Open
Abstract
The term heterotopic ossification (HO) describes bone formation in tissues where bone is normally not present. Musculoskeletal trauma induces signalling events that in turn trigger cells, probably of mesenchymal origin, to differentiate into bone. The aetiology of HO includes extremely rare but severe, generalised and fatal monogenic forms of the disease; and as a common complex disorder in response to musculoskeletal, neurological or burn trauma. The resulting bone forms through a combination of endochondral and intramembranous ossification, depending on the aetiology, initiating stimulus and affected tissue. Given the heterogeneity of the disease, many cell types and biological pathways have been studied in efforts to find effective therapeutic strategies for the disorder. Cells of mesenchymal, haematopoietic and neuroectodermal lineages have all been implicated in the pathogenesis of HO, and the emerging dominant signalling pathways are thought to occur through the bone morphogenetic proteins (BMP), mammalian target of rapamycin (mTOR), and retinoic acid receptor pathways. Increased understanding of these disease mechanisms has resulted in the emergence of several novel investigational therapeutic avenues, including palovarotene and other retinoic acid receptor agonists and activin A inhibitors that target both canonical and non-canonical signalling downstream of the BMP type 1 receptor. In this article we aim to illustrate the key cellular and molecular mechanisms involved in the pathogenesis of HO and outline recent advances in emerging molecular therapies to treat and prevent HO that have had early success in the monogenic disease and are currently being explored in the common complex forms of HO.
Collapse
|
18
|
Lambrescu I, Popa A, Manole E, Ceafalan LC, Gaina G. Application of Droplet Digital PCR Technology in Muscular Dystrophies Research. Int J Mol Sci 2022; 23:ijms23094802. [PMID: 35563191 PMCID: PMC9099497 DOI: 10.3390/ijms23094802] [Citation(s) in RCA: 2] [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: 03/01/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 11/25/2022] Open
Abstract
Although they are considered rare disorders, muscular dystrophies have a strong impact on people’s health. Increased disease severity with age, frequently accompanied by the loss of ability to walk in some people, and the lack of treatment, have directed the researchers towards the development of more effective therapeutic strategies aimed to improve the quality of life and life expectancy, slow down the progression, and delay the onset or convert a severe phenotype into a milder one. Improved understanding of the complex pathology of these diseases together with the tremendous advances in molecular biology technologies has led to personalized therapeutic procedures. Different approaches that are currently under extensive investigation require more efficient, sensitive, and less invasive methods. Due to its remarkable analytical sensitivity, droplet digital PCR has become a promising tool for accurate measurement of biomarkers that monitor disease progression and quantification of various therapeutic efficiency and can be considered a tool for non-invasive prenatal diagnosis and newborn screening. Here, we summarize the recent applications of droplet digital PCR in muscular dystrophy research and discuss the factors that should be considered to get the best performance with this technology.
Collapse
Affiliation(s)
- Ioana Lambrescu
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Alexandra Popa
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Department of Animal Production and Public Health, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 050097 Bucharest, Romania
| | - Emilia Manole
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Laura Cristina Ceafalan
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Gisela Gaina
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Correspondence: ; Tel.: +40-21-319-2732
| |
Collapse
|
19
|
Happi Mbakam C, Lamothe G, Tremblay JP. Therapeutic Strategies for Dystrophin Replacement in Duchenne Muscular Dystrophy. Front Med (Lausanne) 2022; 9:859930. [PMID: 35419381 PMCID: PMC8995704 DOI: 10.3389/fmed.2022.859930] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked hereditary disease characterized by progressive muscle wasting due to modifications in the DMD gene (exon deletions, nonsense mutations, intra-exonic insertions or deletions, exon duplications, splice site defects, and deep intronic mutations) that result in a lack of functional dystrophin expression. Many therapeutic approaches have so far been attempted to induce dystrophin expression and improve the patient phenotype. In this manuscript, we describe the relevant updates for some therapeutic strategies for DMD aiming to restore dystrophin expression. We also present and analyze in vitro and in vivo ongoing experimental approaches to treat the disease.
Collapse
Affiliation(s)
- Cedric Happi Mbakam
- Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Gabriel Lamothe
- Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | - Jacques P Tremblay
- Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada.,Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| |
Collapse
|
20
|
Sun C, Kannan S, Choi IY, Lim H, Zhang H, Chen GS, Zhang N, Park SH, Serra C, Iyer SR, Lloyd TE, Kwon C, Lovering RM, Lim SB, Andersen P, Wagner KR, Lee G. Human pluripotent stem cell-derived myogenic progenitors undergo maturation to quiescent satellite cells upon engraftment. Cell Stem Cell 2022; 29:610-619.e5. [PMID: 35395188 PMCID: PMC9000524 DOI: 10.1016/j.stem.2022.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/10/2021] [Accepted: 03/09/2022] [Indexed: 12/19/2022]
Abstract
Human pluripotent stem cell (hPSC)-derived myogenic progenitor cell (MPC) transplantation is a promising therapeutic approach for a variety of degenerative muscle disorders. Here, using an MPC-specific fluorescent reporter system (PAX7::GFP), we demonstrate that hPSC-derived MPCs can contribute to the regeneration of myofibers in mice following local injury and in mice deficient of dystrophin (mdx). We also demonstrate that a subset of PAX7::GFP MPCs engraft within the basal lamina of regenerated myofibers, adopt a quiescent state, and contribute to regeneration upon reinjury and in mdx mouse models. This subset of PAX7::GFP MPCs undergo a maturation process and remodel their molecular characteristics to resemble those of late-stage fetal MPCs/adult satellite cells following in vivo engraftment. These in-vivo-matured PAX7::GFP MPCs retain a cell-autonomous ability to regenerate and can repopulate in the niche of secondary recipient mice, providing a proof of principle for future hPSC-based cell therapy for muscle disorders.
Collapse
Affiliation(s)
- Congshan Sun
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Genetic Muscle Disorders, The Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Suraj Kannan
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biomedical Engineering and Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - In Young Choi
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - HoTae Lim
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hao Zhang
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Grace S Chen
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Nancy Zhang
- Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Seong-Hyun Park
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Carlo Serra
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Genetic Muscle Disorders, The Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Shama R Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Thomas E Lloyd
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chulan Kwon
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biomedical Engineering and Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Su Bin Lim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Peter Andersen
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kathryn R Wagner
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Genetic Muscle Disorders, The Kennedy Krieger Institute, Baltimore, MD 21205, USA.
| | - Gabsang Lee
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
21
|
|
22
|
Charrier M, Lorant J, Contreras-Lopez R, Téjédor G, Blanquart C, Lieubeau B, Schleder C, Leroux I, Deshayes S, Fonteneau JF, Babarit C, Hamel A, Magot A, Péréon Y, Viau S, Delorme B, Luz-Crawford P, Lamirault G, Djouad F, Rouger K. Human MuStem cells repress T-cell proliferation and cytotoxicity through both paracrine and contact-dependent pathways. Stem Cell Res Ther 2022; 13:7. [PMID: 35012660 PMCID: PMC8751303 DOI: 10.1186/s13287-021-02681-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/09/2021] [Indexed: 11/23/2022] Open
Abstract
Background Muscular dystrophies (MDs) are inherited diseases in which a dysregulation of the immune response exacerbates disease severity and are characterized by infiltration of various immune cell types leading to muscle inflammation, fiber necrosis and fibrosis. Immunosuppressive properties have been attributed to mesenchymal stem cells (MSCs) that regulate the phenotype and function of different immune cells. However, such properties were poorly considered until now for adult stem cells with myogenic potential and advanced as possible therapeutic candidates for MDs. In the present study, we investigated the immunoregulatory potential of human MuStem (hMuStem) cells, for which we previously demonstrated that they can survive in injured muscle and robustly counteract adverse tissue remodeling. Methods The impact of hMuStem cells or their secretome on the proliferative and phenotypic properties of T-cells was explored by co-culture experiments with either peripheral blood mononucleated cells or CD3-sorted T-cells. A comparative study was produced with the bone marrow (BM)-MSCs. The expression profile of immune cell-related markers on hMuStem cells was determined by flow cytometry while their secretory profile was examined by ELISA assays. Finally, the paracrine and cell contact-dependent effects of hMuStem cells on the T-cell-mediated cytotoxic response were analyzed through IFN-γ expression and lysis activity. Results Here, we show that hMuStem cells have an immunosuppressive phenotype and can inhibit the proliferation and the cytotoxic response of T-cells as well as promote the generation of regulatory T-cells through direct contact and via soluble factors. These effects are associated, in part, with the production of mediators including heme-oxygenase-1, leukemia inhibitory factor and intracellular cell adhesion molecule-1, all of which are produced at significantly higher levels by hMuStem cells than BM-MSCs. While the production of prostaglandin E2 is involved in the suppression of T-cell proliferation by both hMuStem cells and BM-MSCs, the participation of inducible nitric oxide synthase activity appears to be specific to hMuStem cell-mediated one. Conclusions Together, our findings demonstrate that hMuStem cells are potent immunoregulatory cells. Combined with their myogenic potential, the attribution of these properties reinforces the positioning of hMuStem cells as candidate therapeutic agents for the treatment of MDs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02681-3.
Collapse
Affiliation(s)
- Marine Charrier
- INRAE, Oniris, PAnTher, UMR 703, Oniris - Site de La Chantrerie, 101, Route de Gachet, CS. 40706, 44307, Nantes, France.,L'institut du Thorax, INSERM, CNRS, UNIV Nantes, 44007, Nantes, France.,Université de Nantes, Nantes, France
| | - Judith Lorant
- INRAE, Oniris, PAnTher, UMR 703, Oniris - Site de La Chantrerie, 101, Route de Gachet, CS. 40706, 44307, Nantes, France
| | - Rafael Contreras-Lopez
- INSERM U1183 IRMB, Hôpital Saint Eloi, CHRU Montpellier, Université de Montpellier, 80, Rue Augustin Fliche, 34295, Montpellier, France.,Laboratorio de Immunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Las Condes, Chile
| | - Gautier Téjédor
- INSERM U1183 IRMB, Hôpital Saint Eloi, CHRU Montpellier, Université de Montpellier, 80, Rue Augustin Fliche, 34295, Montpellier, France
| | | | | | - Cindy Schleder
- INRAE, Oniris, PAnTher, UMR 703, Oniris - Site de La Chantrerie, 101, Route de Gachet, CS. 40706, 44307, Nantes, France
| | - Isabelle Leroux
- INRAE, Oniris, PAnTher, UMR 703, Oniris - Site de La Chantrerie, 101, Route de Gachet, CS. 40706, 44307, Nantes, France
| | - Sophie Deshayes
- CNRS, INSERM, CRCINA, Université de Nantes, 44000, Nantes, France
| | | | - Candice Babarit
- INRAE, Oniris, PAnTher, UMR 703, Oniris - Site de La Chantrerie, 101, Route de Gachet, CS. 40706, 44307, Nantes, France
| | - Antoine Hamel
- Service de Chirurgie Infantile, Centre Hospitalier Universitaire (CHU) de Nantes, 44093, Nantes, France
| | - Armelle Magot
- Laboratoire d'Explorations Fonctionnelles, Centre de Référence Maladies Neuromusculaires AOC, CHU Nantes, 44093, Nantes, France
| | - Yann Péréon
- Laboratoire d'Explorations Fonctionnelles, Centre de Référence Maladies Neuromusculaires AOC, CHU Nantes, 44093, Nantes, France
| | - Sabrina Viau
- Biotherapy Division, Macopharma, 59420, Mouvaux, France
| | - Bruno Delorme
- Biotherapy Division, Macopharma, 59420, Mouvaux, France
| | - Patricia Luz-Crawford
- Laboratorio de Immunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Las Condes, Chile.,IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | | | - Farida Djouad
- INSERM U1183 IRMB, Hôpital Saint Eloi, CHRU Montpellier, Université de Montpellier, 80, Rue Augustin Fliche, 34295, Montpellier, France.
| | - Karl Rouger
- INRAE, Oniris, PAnTher, UMR 703, Oniris - Site de La Chantrerie, 101, Route de Gachet, CS. 40706, 44307, Nantes, France.
| |
Collapse
|
23
|
Adult Stem Cell Therapy as Regenerative Medicine for End-Stage Liver Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1401:57-72. [DOI: 10.1007/5584_2022_719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
24
|
The Efficacy of Schwann-Like Differentiated Muscle-Derived Stem Cells in Treating Rodent Upper Extremity Peripheral Nerve Injury. Plast Reconstr Surg 2021; 148:787-798. [PMID: 34550935 DOI: 10.1097/prs.0000000000008383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND There is a pressing need to identify alternative mesenchymal stem cell sources for Schwann cell cellular replacement therapy, to improve peripheral nerve regeneration. This study assessed the efficacy of Schwann cell-like cells (induced muscle-derived stem cells) differentiated from muscle-derived stem cells (MDSCs) in augmenting nerve regeneration and improving muscle function after nerve trauma. METHODS The Schwann cell-like nature of induced MDSCs was characterized in vitro using immunofluorescence, flow cytometry, microarray, and reverse-transcription polymerase chain reaction. In vivo, four groups (n = 5 per group) of rats with median nerve injuries were examined: group 1 animals were treated with intraneural phosphate-buffered saline after cold and crush axonotmesis (negative control); group 2 animals were no-injury controls; group 3 animals were treated with intraneural green fluorescent protein-positive MDSCs; and group 4 animals were treated with green fluorescent protein-positive induced MDSCs. All animals underwent weekly upper extremity functional testing. Rats were euthanized 5 weeks after treatment. The median nerve and extrinsic finger flexors were harvested for nerve histomorphometry, myelination, muscle weight, and atrophy analyses. RESULTS In vitro, induced MDSCs recapitulated native Schwann cell gene expression patterns and up-regulated pathways involved in neuronal growth/signaling. In vivo, green fluorescent protein-positive induced MDSCs remained stably transformed 5 weeks after injection. Induced MDSC therapy decreased muscle atrophy after median nerve injury (p = 0.0143). Induced MDSC- and MDSC-treated animals demonstrated greater functional muscle recovery when compared to untreated controls (hand grip after induced MDSC treatment: group 1, 0.91 N; group 4, 3.38 N); p < 0.0001) at 5 weeks after treatment. This may demonstrate the potential beneficial effects of MDSC therapy, regardless of differentiation stage. CONCLUSION Both MDSCs and induced MDSCs decrease denervation muscle atrophy and improve subsequent functional outcomes after upper extremity nerve trauma in rodents.
Collapse
|
25
|
Archacka K, Grabowska I, Mierzejewski B, Graffstein J, Górzyńska A, Krawczyk M, Różycka AM, Kalaszczyńska I, Muras G, Stremińska W, Jańczyk-Ilach K, Walczak P, Janowski M, Ciemerych MA, Brzoska E. Hypoxia preconditioned bone marrow-derived mesenchymal stromal/stem cells enhance myoblast fusion and skeletal muscle regeneration. Stem Cell Res Ther 2021; 12:448. [PMID: 34372911 PMCID: PMC8351116 DOI: 10.1186/s13287-021-02530-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
Background The skeletal muscle reconstruction occurs thanks to unipotent stem cells, i.e., satellite cells. The satellite cells remain quiescent and localized between myofiber sarcolemma and basal lamina. They are activated in response to muscle injury, proliferate, differentiate into myoblasts, and recreate myofibers. The stem and progenitor cells support skeletal muscle regeneration, which could be disturbed by extensive damage, sarcopenia, cachexia, or genetic diseases like dystrophy. Many lines of evidence showed that the level of oxygen regulates the course of cell proliferation and differentiation. Methods In the present study, we analyzed hypoxia impact on human and pig bone marrow-derived mesenchymal stromal cell (MSC) and mouse myoblast proliferation, differentiation, and fusion. Moreover, the influence of the transplantation of human bone marrow-derived MSCs cultured under hypoxic conditions on skeletal muscle regeneration was studied. Results We showed that bone marrow-derived MSCs increased VEGF expression and improved myogenesis under hypoxic conditions in vitro. Transplantation of hypoxia preconditioned bone marrow-derived MSCs into injured muscles resulted in the improved cell engraftment and formation of new vessels. Conclusions We suggested that SDF-1 and VEGF secreted by hypoxia preconditioned bone marrow-derived MSCs played an essential role in cell engraftment and angiogenesis. Importantly, hypoxia preconditioned bone marrow-derived MSCs more efficiently engrafted injured muscles; however, they did not undergo myogenic differentiation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02530-3.
Collapse
Affiliation(s)
- Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Bartosz Mierzejewski
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Joanna Graffstein
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Alicja Górzyńska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Marta Krawczyk
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Anna M Różycka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Ilona Kalaszczyńska
- Department of Histology and Embryology, Medical University of Warsaw, 02-004, Warsaw, Poland.,Laboratory for Cell Research and Application, Medical University of Warsaw, 02-097, Warsaw, Poland
| | - Gabriela Muras
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Władysława Stremińska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Katarzyna Jańczyk-Ilach
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Piotr Walczak
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Warszawska 30 St, 10-082, Olsztyn, Poland.,Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, the Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mirosław Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, 21201, USA.,NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5 St, 02-106, Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096, Warsaw, Poland.
| |
Collapse
|
26
|
Boyer O, Butler-Browne G, Chinoy H, Cossu G, Galli F, Lilleker JB, Magli A, Mouly V, Perlingeiro RCR, Previtali SC, Sampaolesi M, Smeets H, Schoewel-Wolf V, Spuler S, Torrente Y, Van Tienen F. Myogenic Cell Transplantation in Genetic and Acquired Diseases of Skeletal Muscle. Front Genet 2021; 12:702547. [PMID: 34408774 PMCID: PMC8365145 DOI: 10.3389/fgene.2021.702547] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/16/2021] [Indexed: 01/04/2023] Open
Abstract
This article will review myogenic cell transplantation for congenital and acquired diseases of skeletal muscle. There are already a number of excellent reviews on this topic, but they are mostly focused on a specific disease, muscular dystrophies and in particular Duchenne Muscular Dystrophy. There are also recent reviews on cell transplantation for inflammatory myopathies, volumetric muscle loss (VML) (this usually with biomaterials), sarcopenia and sphincter incontinence, mainly urinary but also fecal. We believe it would be useful at this stage, to compare the same strategy as adopted in all these different diseases, in order to outline similarities and differences in cell source, pre-clinical models, administration route, and outcome measures. This in turn may help to understand which common or disease-specific problems have so far limited clinical success of cell transplantation in this area, especially when compared to other fields, such as epithelial cell transplantation. We also hope that this may be useful to people outside the field to get a comprehensive view in a single review. As for any cell transplantation procedure, the choice between autologous and heterologous cells is dictated by a number of criteria, such as cell availability, possibility of in vitro expansion to reach the number required, need for genetic correction for many but not necessarily all muscular dystrophies, and immune reaction, mainly to a heterologous, even if HLA-matched cells and, to a minor extent, to the therapeutic gene product, a possible antigen for the patient. Finally, induced pluripotent stem cell derivatives, that have entered clinical experimentation for other diseases, may in the future offer a bank of immune-privileged cells, available for all patients and after a genetic correction for muscular dystrophies and other myopathies.
Collapse
Affiliation(s)
- Olivier Boyer
- Department of Immunology & Biotherapy, Rouen University Hospital, Normandy University, Inserm U1234, Rouen, France
| | - Gillian Butler-Browne
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Hector Chinoy
- Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Giulio Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, The University of Manchester, Manchester, United Kingdom
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Berlin, Germany
- InSpe and Division of Neuroscience, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Francesco Galli
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - James B. Lilleker
- Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal NHS Foundation Trust, Salford, United Kingdom
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, United Kingdom
| | - Alessandro Magli
- Department of Medicine, Lillehei Heart Institute, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Rita C. R. Perlingeiro
- Department of Medicine, Lillehei Heart Institute, Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| | - Stefano C. Previtali
- InSpe and Division of Neuroscience, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Human Anatomy Unit, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Hubert Smeets
- Department of Toxicogenomics, Maastricht University Medical Centre, Maastricht, Netherlands
- School for Mental Health and Neurosciences (MHeNS), Maastricht University, Maastricht, Netherlands
- School for Developmental Biology and Oncology (GROW), Maastricht University, Maastricht, Netherlands
| | - Verena Schoewel-Wolf
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, a Cooperation Between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Yvan Torrente
- Unit of Neurology, Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Centro Dino Ferrari, Università degli Studi di Milano, Fondazione Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Florence Van Tienen
- Department of Toxicogenomics, Maastricht University Medical Centre, Maastricht, Netherlands
- School for Mental Health and Neurosciences (MHeNS), Maastricht University, Maastricht, Netherlands
| |
Collapse
|
27
|
Gaina G, Popa (Gruianu) A. Muscular dystrophy: Experimental animal models and therapeutic approaches (Review). Exp Ther Med 2021; 21:610. [PMID: 33936267 PMCID: PMC8082581 DOI: 10.3892/etm.2021.10042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
The muscular dystrophies are a heterogeneous group of genetically inherited diseases characterized by muscle weakness and progressive wasting, which can cause premature death in severe forms. Although >30 years have passed since the identification of the first protein involved in a type of muscular dystrophy, there is no effective treatment for these disabling disorders. In the last decade, several novel therapeutic approaches have been developed and investigated as promising therapeutic approaches aimed to ameliorate the dystrophic phenotype either by restoring dystrophin expression or by compensating for dystrophin deficiency. Concurrently, with the development of therapeutic approaches, in addition to naturally occurring animal models, a wide range of genetically engineered animal models has been generated. The use of animals as models of muscular dystrophies has greatly improved the understanding of the pathogenicity of these diseases and has proven useful in gene therapy studies. In this review, we summarize these latest innovative therapeutic approaches to muscular dystrophies and the usefulness of the various most common experimental animal models.
Collapse
Affiliation(s)
- Gisela Gaina
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Alexandra Popa (Gruianu)
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, ‘Victor Babes’ National Institute of Pathology, 050096 Bucharest, Romania
- Department of Animal Production and Public Health, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 050097 Bucharest, Romania
| |
Collapse
|
28
|
Cwykiel J, Madajka-Niemeyer M, Siemionow M. Development of Donor Recipient Chimeric Cells of bone marrow origin as a novel approach for tolerance induction in transplantation. Stem Cell Investig 2021; 8:8. [PMID: 33969113 DOI: 10.21037/sci-2020-044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/03/2021] [Indexed: 12/14/2022]
Abstract
Background Cell therapies and chimerism-based strategies are currently the most successful approach for tolerance induction in transplantation. This study aimed to establish and characterize novel Donor Recipient Chimeric Ccell (DRCC) therapy of bone marrow (BM) origin presenting donor-recipient phenotype to support tolerance induction. Methods Ex vivo fusions of fully MHC-mismatched BM cells from ACI (RT1a) and Lewis (RT1l) rats were performed using polyethylene-glycol (PEG). The creation of rat DRCC was tested by flow cytometry (FC), confocal microscopy and PCR. FC characterized DRCC's phenotype (CD3, CD4, CD8, CD45, CD90, CD11b/c, CD45RA, OX-82, or CD4/CD25) and apoptosis, while mixed lymphocyte reaction assessed DRCC's immunogenicity and colony forming unit assay tested DRCC's differentiation and proliferation. DRCC's polyploidy was evaluated using Hoechst33342 staining and COMET assay tested genotoxicity of fusion procedure. ELISA analyzed the secretion of IL-2, IL-4, IL-10, TGFß1, IFNγ and TNFα by DRCC at day 1, 5 and 14 post-fusion. The DRCC's phenotype after long-term culturing was assessed by reverse-transcription PCR. Results The chimeric state of DRCC was confirmed. Fusion did not change the expression of hematopoietic markers compared to BM controls. Although an increased number of early and late apoptotic (Annexin V+/Sytox blue- and Annexin V+/Sytox blue+, respectively) DRCC was detected at 24h post-fusion, the number significantly decreased at day 5 (38.4%±3.1% and 22.6%±2.5%, vs. 28.3%±2.5% and 13.9%±2.6%, respectively, P<0.05). DRCC presented decreased immunogenicity, increased expression of IL-10 and TGFβ1 and proliferative potential comparable to BM controls. The average percentage of tetraploid DRCC was 3.1%±0.2% compared to 0.96%±0.1% in BM controls. The lack of damage to the DRCC's DNA content supported the DRCC's safety. In culture, DRCC maintained proliferation for up to 28 days while preserving hematopoietic profile. Conclusions This study confirmed feasibility of DRCC creation via ex vivo PEG mediated fusion. The created DRCC revealed pro-tolerogenic properties indicating potential immunomodulatory effect of DRCC therapy when applied in vivo to support tolerance induction in solid organ and vascularized composite allograft transplantation.
Collapse
Affiliation(s)
- Joanna Cwykiel
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA.,Department of Plastic Surgery, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Maria Siemionow
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL, USA.,Department of Plastic Surgery, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Surgery, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
29
|
Kindler V, Paccaud J, Hannouche D, Laumonier T. Human myoblasts differentiate in various mesenchymal lineages and inhibit allogeneic T cell proliferation through an indolamine 2,3 dioxygenase dependent pathway. Exp Cell Res 2021; 403:112586. [PMID: 33839146 DOI: 10.1016/j.yexcr.2021.112586] [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: 09/16/2020] [Revised: 03/16/2021] [Accepted: 03/28/2021] [Indexed: 12/12/2022]
Abstract
Muscle stem cells (MuSC) are considered as a reliable source of therapeutic cells to restore diseased muscles. However in most cases, injected MuSC-derived myoblasts are rapidly destroyed by the host immune response, which impairs the beneficial effect. By contrast, human mesenchymal stromal cells (MSC), have been reported to exhibit potent immune regulatory functions. Thus, we investigated, in vitro, the multipotent differentiation- and immunosuppressive capacities of human myoblasts and compared these features with those of human MSC. Myoblasts shared numerous cell surface markers with MSC, including CD73, CD90, CD105 and CD146. Both cell type were negative for HLA-DR and CD45, CD34 and CD31. CD56, a myogenic marker, was expressed by myoblasts exclusively. Myoblasts displayed multipotent potential capabilities with differentiation in chondrocytes, adipocytes and osteoblasts in vitro. Myoblasts also inhibited allogenic T cell proliferation in vitro in a dose dependent manner, very similarly to MSC. This effect was partly mediated via the activation of indolamine 2,3 dioxygenase enzyme (IDO) after IFNγ exposure. Altogether, these data demonstrate that human myoblasts can differentiate in various mesenchymal linages and exhibit powerful immunosuppressive properties in vitro. Such features may open new therapeutic strategies for MuSC-derived myoblasts.
Collapse
Affiliation(s)
- Vincent Kindler
- Department of Orthopedic Surgery, Geneva University Hospitals & Faculty of Medicine, Geneva, Switzerland
| | - Joris Paccaud
- Department of Orthopedic Surgery, Geneva University Hospitals & Faculty of Medicine, Geneva, Switzerland
| | - Didier Hannouche
- Department of Orthopedic Surgery, Geneva University Hospitals & Faculty of Medicine, Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland
| | - Thomas Laumonier
- Department of Orthopedic Surgery, Geneva University Hospitals & Faculty of Medicine, Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, Geneva, Switzerland.
| |
Collapse
|
30
|
Khatkar H, See A. Stem Cell Therapy in the Management of Fracture Non-Union - Evaluating Cellular Mechanisms and Clinical Progress. Cureus 2021; 13:e13869. [PMID: 33859917 PMCID: PMC8038927 DOI: 10.7759/cureus.13869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Bone, as a physiological and anatomical construct, displays remarkable intrinsic healing capacity. The overwhelming majority of fractures will heal satisfactorily, if aligned anatomically, compressed and immobilised appropriately. Of the 10% of fractures that do not heal, even under ideal mechanical and biological conditions, further consideration must be given to augment bone healing. Management strategies for non-union pose a significant clinical challenge to the practicing orthopaedic surgeon. Stem cell therapy is beginning to demonstrate significant potential for augmented bone repair in the context of non-union. This review attempts to contextualise the function of stem cells within this clinical setting, reviewing the relevant cellular mechanisms and clinical applications. From evaluating the literature base, there is a lack of high-quality evidence examining the role of mesenchymal stem cells (MSCs) within this research focus. Appropriately designed randomised controlled trials are required to evaluate this research area further, with a view to guiding future treatment options for the practicing orthopaedic surgeon.
Collapse
Affiliation(s)
- Harman Khatkar
- Trauma and Orthopaedics, Royal Berkshire Hospital, Reading, GBR.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, GBR
| | - Abbas See
- Trauma and Orthopaedics, Kettering General Hospital, Kettering, GBR
| |
Collapse
|
31
|
Biressi S, Filareto A, Rando TA. Stem cell therapy for muscular dystrophies. J Clin Invest 2021; 130:5652-5664. [PMID: 32946430 DOI: 10.1172/jci142031] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Muscular dystrophies are a heterogeneous group of genetic diseases, characterized by progressive degeneration of skeletal and cardiac muscle. Despite the intense investigation of different therapeutic options, a definitive treatment has not been developed for this debilitating class of pathologies. Cell-based therapies in muscular dystrophies have been pursued experimentally for the last three decades. Several cell types with different characteristics and tissues of origin, including myogenic stem and progenitor cells, stromal cells, and pluripotent stem cells, have been investigated over the years and have recently entered in the clinical arena with mixed results. In this Review, we do a roundup of the past attempts and describe the updated status of cell-based therapies aimed at counteracting the skeletal and cardiac myopathy present in dystrophic patients. We present current challenges, summarize recent progress, and make recommendations for future research and clinical trials.
Collapse
Affiliation(s)
- Stefano Biressi
- Department of Cellular, Computational and Integrative Biology (CIBIO) and.,Dulbecco Telethon Institute, University of Trento, Povo, Italy
| | - Antonio Filareto
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Conneticut, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences and.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California, USA.,Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
32
|
Abstract
The resident stem cell for skeletal muscle is the satellite cell. On the 50th anniversary of its discovery in 1961, we described the history of skeletal muscle research and the seminal findings made during the first 20 years in the life of the satellite cell (Scharner and Zammit 2011, doi: 10.1186/2044-5040-1-28). These studies established the satellite cell as the source of myoblasts for growth and regeneration of skeletal muscle. Now on the 60th anniversary, we highlight breakthroughs in the second phase of satellite cell research from 1980 to 2000. These include technical innovations such as isolation of primary satellite cells and viable muscle fibres complete with satellite cells in their niche, together with generation of many useful reagents including genetically modified organisms and antibodies still in use today. New methodologies were combined with description of endogenous satellite cells markers, notably Pax7. Discovery of the muscle regulatory factors Myf5, MyoD, myogenin, and MRF4 in the late 1980s revolutionized understanding of the control of both developmental and regerenative myogenesis. Emergence of genetic lineage markers facilitated identification of satellite cells in situ, and also empowered transplantation studies to examine satellite cell function. Finally, satellite cell heterogeneity and the supportive role of non-satellite cell types in muscle regeneration were described. These major advances in methodology and in understanding satellite cell biology provided further foundations for the dramatic escalation of work on muscle stem cells in the 21st century.
Collapse
Affiliation(s)
- Elise N Engquist
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, SE1 1UL, UK
| |
Collapse
|
33
|
"Betwixt Mine Eye and Heart a League Is Took": The Progress of Induced Pluripotent Stem-Cell-Based Models of Dystrophin-Associated Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21196997. [PMID: 32977524 PMCID: PMC7582534 DOI: 10.3390/ijms21196997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
The ultimate goal of precision disease modeling is to artificially recreate the disease of affected people in a highly controllable and adaptable external environment. This field has rapidly advanced which is evident from the application of patient-specific pluripotent stem-cell-derived precision therapies in numerous clinical trials aimed at a diverse set of diseases such as macular degeneration, heart disease, spinal cord injury, graft-versus-host disease, and muscular dystrophy. Despite the existence of semi-adequate treatments for tempering skeletal muscle degeneration in dystrophic patients, nonischemic cardiomyopathy remains one of the primary causes of death. Therefore, cardiovascular cells derived from muscular dystrophy patients' induced pluripotent stem cells are well suited to mimic dystrophin-associated cardiomyopathy and hold great promise for the development of future fully effective therapies. The purpose of this article is to convey the realities of employing precision disease models of dystrophin-associated cardiomyopathy. This is achieved by discussing, as suggested in the title echoing William Shakespeare's words, the settlements (or "leagues") made by researchers to manage the constraints ("betwixt mine eye and heart") distancing them from achieving a perfect precision disease model.
Collapse
|
34
|
Testa S, Riera CS, Fornetti E, Riccio F, Fuoco C, Bernardini S, Baldi J, Costantini M, Foddai ML, Cannata S, Gargioli C. Skeletal Muscle-Derived Human Mesenchymal Stem Cells: Influence of Different Culture Conditions on Proliferative and Myogenic Capabilities. Front Physiol 2020; 11:553198. [PMID: 33041857 PMCID: PMC7526461 DOI: 10.3389/fphys.2020.553198] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/12/2020] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle tissue is characterized by restrained self-regenerative capabilities, being ineffective in relation to trauma extension both in time span (e.g., chronic diseases) and in size (e.g., large trauma). For these reasons, tissue engineering and/or cellular therapies represent a valuable solution in the cases where the physiological healing process failed. Satellite cells, the putative skeletal muscle stem cells, have been the first solution explored to remedy the insufficient self-regeneration capacity. Nevertheless, some limitation related to donor age, muscle condition, expansion hitch, and myogenic potentiality maintenance have limited their use as therapeutic tool. To overcome this hindrance, different stem cells population with myogenic capabilities have been investigated to evaluate their real potentiality for therapeutic approaches, but, as of today, the perfect cell candidate has not been identified yet. In this work, we analyze the characteristics of skeletal muscle-derived human Mesenchymal Stem Cells (hMSCs), showing the maintenance/increment of myogenic activity upon differential culture conditions. In particular, we investigate the influence of a commercial enriched growth medium (Cyto-Grow), and of a medium enriched with either human-derived serum (H.S.) or human Platelet-rich Plasma (PrP), in order to set up a culture protocol useful for employing this cell population in clinical therapeutic strategies. The presented results reveal that both the enriched medium (Cyto-Grow) and the human-derived supplements (H.S. and PrP) have remarkable effects on hMSCs proliferation and myogenic differentiation compared to standard condition, uncovering the real possibility to exploit these human derivatives to ameliorate stem cells yield and efficacy.
Collapse
Affiliation(s)
- Stefano Testa
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Ersilia Fornetti
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Federica Riccio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Claudia Fuoco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Jacopo Baldi
- IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | | | - Stefano Cannata
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
35
|
Tamaki T. Biomedical applications of muscle-derived stem cells: from bench to bedside. Expert Opin Biol Ther 2020; 20:1361-1371. [PMID: 32643444 DOI: 10.1080/14712598.2020.1793953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Skeletal muscle-derived stem cells (Sk-MDSCs) are considered promising sources of adult stem cell therapy. Skeletal muscle comprises approximately 40-50% of the total body mass with marked potential for postnatal adaptive response, such as muscle hypertrophy, hyperplasia, atrophy, and regenerative capacity. This strongly suggests that skeletal muscle contains various stem/progenitor cells related to muscle-nerve-vascular tissues, which would support the above postnatal events even in adulthood. AREA COVERED The focus of this review is the therapeutic potential of the Sk-MDSCs as an adult stem cell autograft. For this purpose, the validity of cell isolation and purification, tissue reconstitution capacity in vivo after transplantation, comparison of the results of basic mouse and preclinical human studies, potential problematic and beneficial aspects, and effective usage have been discussed following the history of clinical applications. EXPERT OPINION Although the clinical application of Sk-MDSCs began as a therapy for the systemic disease of Duchenne muscular dystrophy, here, through the unique local injection method, therapy for severely damaged peripheral nerves, particularly the long-gap nerve transection, has been introduced. The beneficial aspects of the use of Sk-MDSCs as the source of local tissue transplantation therapy have also been discussed.
Collapse
Affiliation(s)
- Tetsuro Tamaki
- Muscle Physiology and Cell Biology Unit, Department of Physiology, Tokai University School of Medicine , Isehara, Kanagawa ,Japan
| |
Collapse
|
36
|
He L, Nguyen NB, Ardehali R, Zhou B. Heart Regeneration by Endogenous Stem Cells and Cardiomyocyte Proliferation: Controversy, Fallacy, and Progress. Circulation 2020; 142:275-291. [PMID: 32687441 DOI: 10.1161/circulationaha.119.045566] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Ischemic heart disease is the leading cause of death worldwide. Myocardial infarction results in an irreversible loss of cardiomyocytes with subsequent adverse remodeling and heart failure. Identifying new sources for cardiomyocytes and promoting their formation represents a goal of cardiac biology and regenerative medicine. Within the past decade, many types of putative cardiac stem cells (CSCs) have been reported to regenerate the injured myocardium by differentiating into new cardiomyocytes. Some of these CSCs have been translated from bench to bed with reported therapeutic effectiveness. However, recent basic research studies on stem cell tracing have begun to question their fundamental biology and mechanisms of action, raising serious concerns over the myogenic potential of CSCs. We review the history of different types of CSCs within the past decade and provide an update of recent cell tracing studies that have challenged the origin and existence of CSCs. In addition to the potential role of CSCs in heart regeneration, proliferation of preexisting cardiomyocytes has recently gained more attention. This review will also evaluate the methodologic and technical aspects of past and current studies on CSCs and cardiomyocyte proliferation, with emphasis on technical strengths, advantages, and potential limitations of research approaches. While our understanding of cardiomyocyte generation and regeneration continues to evolve, it is important to address the shortcomings and inaccuracies in this field. This is best achieved by embracing technological advancements and improved methods to label single cardiomyocytes/progenitors and accurately investigate their developmental potential and fate/lineage commitment.
Collapse
Affiliation(s)
- Lingjuan He
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China (L.H., B.Z.)
| | - Ngoc B Nguyen
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine (N.B.N., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (N.B.N., R.A.), University of California, Los Angeles
| | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine (N.B.N., R.A.), University of California, Los Angeles.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research (N.B.N., R.A.), University of California, Los Angeles
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China (L.H., B.Z.).,School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.).,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China (B.Z.).,Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China (B.Z.)
| |
Collapse
|
37
|
Brzoska E, Kalkowski L, Kowalski K, Michalski P, Kowalczyk P, Mierzejewski B, Walczak P, Ciemerych MA, Janowski M. Muscular Contribution to Adolescent Idiopathic Scoliosis from the Perspective of Stem Cell-Based Regenerative Medicine. Stem Cells Dev 2020; 28:1059-1077. [PMID: 31170887 DOI: 10.1089/scd.2019.0073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a relatively frequent disease within a range 0.5%-5.0% of population, with higher frequency in females. While a resultant spinal deformity is usually medically benign condition, it produces far going psychosocial consequences, which warrant attention. The etiology of AIS is unknown and current therapeutic approaches are symptomatic only, and frequently inconvenient or invasive. Muscular contribution to AIS is widely recognized, although it did not translate to clinical routine as yet. Muscle asymmetry has been documented by pathological examinations as well as systemic muscle disorders frequently leading to scoliosis. It has been also reported numerous genetic, metabolic and radiological alterations in patients with AIS, which are linked to muscular and neuromuscular aspects. Therefore, muscles might be considered an attractive and still insufficiently exploited therapeutic target for AIS. Stem cell-based regenerative medicine is rapidly gaining momentum based on the tremendous progress in understanding of developmental biology. It comes also with a toolbox of various stem cells such as satellite cells or mesenchymal stem cells, which could be transplanted; also, the knowledge acquired in research on regenerative medicine can be applied to manipulation of endogenous stem cells to obtain desired therapeutic goals. Importantly, paravertebral muscles are located relatively superficially; therefore, they can be an easy target for minimally invasive approaches to treatment of AIS. It comes in pair with a fast progress in image guidance, which allows for precise delivery of therapeutic agents, including stem cells to various organs such as brain, muscles, and others. Summing up, it seems that there is a link between AIS, muscles, and stem cells, which might be worth of further investigations with a long-term goal of setting foundations for eventual bench-to-bedside translation.
Collapse
Affiliation(s)
- Edyta Brzoska
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Lukasz Kalkowski
- 2Department of Neurology and Neurosurgery, Faculty of Medical Sciences, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Kamil Kowalski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Pawel Michalski
- 3Spine Surgery Department, Institute of Mother and Child, Warsaw, Poland
| | - Pawel Kowalczyk
- 4Department of Neurosurgery, Children's Memorial Health Institute, Warsaw, Poland
| | - Bartosz Mierzejewski
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Piotr Walczak
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Maria A Ciemerych
- 1Department of Cytology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Miroslaw Janowski
- 5Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,6Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
38
|
Archacka K, Bem J, Brzoska E, Czerwinska AM, Grabowska I, Kasprzycka P, Hoinkis D, Siennicka K, Pojda Z, Bernas P, Binkowski R, Jastrzebska K, Kupiec A, Malesza M, Michalczewska E, Soszynska M, Ilach K, Streminska W, Ciemerych MA. Beneficial Effect of IL-4 and SDF-1 on Myogenic Potential of Mouse and Human Adipose Tissue-Derived Stromal Cells. Cells 2020; 9:cells9061479. [PMID: 32560483 PMCID: PMC7349575 DOI: 10.3390/cells9061479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Under physiological conditions skeletal muscle regeneration depends on the satellite cells. After injury these cells become activated, proliferate, and differentiate into myofibers reconstructing damaged tissue. Under pathological conditions satellite cells are not sufficient to support regeneration. For this reason, other cells are sought to be used in cell therapies, and different factors are tested as a tool to improve the regenerative potential of such cells. Many studies are conducted using animal cells, omitting the necessity to learn about human cells and compare them to animal ones. Here, we analyze and compare the impact of IL-4 and SDF-1, factors chosen by us on the basis of their ability to support myogenic differentiation and cell migration, at mouse and human adipose tissue-derived stromal cells (ADSCs). Importantly, we documented that mouse and human ADSCs differ in certain reactions to IL-4 and SDF-1. In general, the selected factors impacted transcriptome of ADSCs and improved migration and fusion ability of cells in vitro. In vivo, after transplantation into injured muscles, mouse ADSCs more eagerly participated in new myofiber formation than the human ones. However, regardless of the origin, ADSCs alleviated immune response and supported muscle reconstruction, and cytokine treatment enhanced these effects. Thus, we documented that the presence of ADSCs improves skeletal muscle regeneration and this influence could be increased by cell pretreatment with IL-4 and SDF-1.
Collapse
Affiliation(s)
- Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Joanna Bem
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Edyta Brzoska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Areta M. Czerwinska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Iwona Grabowska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Paulina Kasprzycka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Dzesika Hoinkis
- Intelliseq Ltd., Stanisława Konarskiego 42/13, 30-046 Krakow, Poland;
| | - Katarzyna Siennicka
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, W.K. Roentgena 5, 02-781 Warsaw, Poland; (K.S.); (Z.P.)
| | - Zygmunt Pojda
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, W.K. Roentgena 5, 02-781 Warsaw, Poland; (K.S.); (Z.P.)
| | - Patrycja Bernas
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Robert Binkowski
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Kinga Jastrzebska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Aleksandra Kupiec
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Malgorzata Malesza
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Emilia Michalczewska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Marta Soszynska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Katarzyna Ilach
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Wladyslawa Streminska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
| | - Maria A. Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Ilji Miecznikowa 1, 02-096 Warsaw, Poland; (K.A.); (J.B.); (E.B.); (A.M.C.); (I.G.); (P.K.); (P.B.); (R.B.); (K.J.); (A.K.); (M.M.); (E.M.); (M.S.); (K.I.); (W.S.)
- Correspondence: ; Tel.: +48-22-55-42-216
| |
Collapse
|
39
|
Pulik Ł, Mierzejewski B, Ciemerych MA, Brzóska E, Łęgosz P. The Survey of Cells Responsible for Heterotopic Ossification Development in Skeletal Muscles-Human and Mouse Models. Cells 2020; 9:cells9061324. [PMID: 32466405 PMCID: PMC7349686 DOI: 10.3390/cells9061324] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/16/2020] [Accepted: 05/21/2020] [Indexed: 12/18/2022] Open
Abstract
Heterotopic ossification (HO) manifests as bone development in the skeletal muscles and surrounding soft tissues. It can be caused by injury, surgery, or may have a genetic background. In each case, its development might differ, and depending on the age, sex, and patient's conditions, it could lead to a more or a less severe outcome. In the case of the injury or surgery provoked ossification development, it could be, to some extent, prevented by treatments. As far as genetic disorders are concerned, such prevention approaches are highly limited. Many lines of evidence point to the inflammatory process and abnormalities in the bone morphogenetic factor signaling pathway as the molecular and cellular backgrounds for HO development. However, the clear targets allowing the design of treatments preventing or lowering HO have not been identified yet. In this review, we summarize current knowledge on HO types, its symptoms, and possible ways of prevention and treatment. We also describe the molecules and cells in which abnormal function could lead to HO development. We emphasize the studies involving animal models of HO as being of great importance for understanding and future designing of the tools to counteract this pathology.
Collapse
Affiliation(s)
- Łukasz Pulik
- Department of Orthopaedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland;
| | - Bartosz Mierzejewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland; (B.M.); (M.A.C.)
| | - Maria A. Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland; (B.M.); (M.A.C.)
| | - Edyta Brzóska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1 St, 02-096 Warsaw, Poland; (B.M.); (M.A.C.)
- Correspondence: (E.B.); (P.Ł.); Tel.: +48-22-5542-203 (E.B.); +48-22-5021-514 (P.Ł.)
| | - Paweł Łęgosz
- Department of Orthopaedics and Traumatology, Medical University of Warsaw, Lindley 4 St, 02-005 Warsaw, Poland;
- Correspondence: (E.B.); (P.Ł.); Tel.: +48-22-5542-203 (E.B.); +48-22-5021-514 (P.Ł.)
| |
Collapse
|
40
|
Forcina L, Cosentino M, Musarò A. Mechanisms Regulating Muscle Regeneration: Insights into the Interrelated and Time-Dependent Phases of Tissue Healing. Cells 2020; 9:E1297. [PMID: 32456017 PMCID: PMC7290814 DOI: 10.3390/cells9051297] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Despite a massive body of knowledge which has been produced related to the mechanisms guiding muscle regeneration, great interest still moves the scientific community toward the study of different aspects of skeletal muscle homeostasis, plasticity, and regeneration. Indeed, the lack of effective therapies for several physiopathologic conditions suggests that a comprehensive knowledge of the different aspects of cellular behavior and molecular pathways, regulating each regenerative stage, has to be still devised. Hence, it is important to perform even more focused studies, taking the advantage of robust markers, reliable techniques, and reproducible protocols. Here, we provide an overview about the general aspects of muscle regeneration and discuss the different approaches to study the interrelated and time-dependent phases of muscle healing.
Collapse
Affiliation(s)
| | | | - Antonio Musarò
- Laboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Via Antonio Scarpa, 14, 00161 Rome, Italy; (L.F.); (M.C.)
| |
Collapse
|
41
|
IL-4 and SDF-1 Increase Adipose Tissue-Derived Stromal Cell Ability to Improve Rat Skeletal Muscle Regeneration. Int J Mol Sci 2020; 21:ijms21093302. [PMID: 32392778 PMCID: PMC7246596 DOI: 10.3390/ijms21093302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022] Open
Abstract
Skeletal muscle regeneration depends on the satellite cells, which, in response to injury, activate, proliferate, and reconstruct damaged tissue. However, under certain conditions, such as large injuries or myopathies, these cells might not sufficiently support repair. Thus, other cell populations, among them adipose tissue-derived stromal cells (ADSCs), are tested as a tool to improve regeneration. Importantly, the pro-regenerative action of such cells could be improved by various factors. In the current study, we tested whether IL-4 and SDF-1 could improve the ability of ADSCs to support the regeneration of rat skeletal muscles. We compared their effect at properly regenerating fast-twitch EDL and poorly regenerating slow-twitch soleus. To this end, ADSCs subjected to IL-4 and SDF-1 were analyzed in vitro and also in vivo after their transplantation into injured muscles. We tested their proliferation rate, migration, expression of stem cell markers and myogenic factors, their ability to fuse with myoblasts, as well as their impact on the mass, structure and function of regenerating muscles. As a result, we showed that cytokine-pretreated ADSCs had a beneficial effect in the regeneration process. Their presence resulted in improved muscle structure and function, as well as decreased fibrosis development and a modulated immune response.
Collapse
|
42
|
Multi-Spheroid-Loaded Human Acellular Dermal Matrix Carrier Preserves Its Spheroid Shape and Improves In Vivo Adipose-Derived Stem Cell Delivery and Engraftment. Tissue Eng Regen Med 2020; 17:271-283. [PMID: 32314311 DOI: 10.1007/s13770-020-00252-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/09/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Current in vivo adult stem cell delivery presents limited clinical effects due to poor engraftment and survival. To overcome current challenges in cell delivery and promote surgical cell delivery for soft tissue repair, a multi-spheroid-loaded thin sectioned acellular dermal matrix (tsADM) carrier which preserves loaded spheroids' three-dimensional (3D) structure, was developed. METHODS Adipose-derived stem cells (ASCs) were used for spheroid delivery. After generating spheroids in 3D cell culture dishes, spheroid plasticity and survival in-between coverslips were evaluated. Spheroids were loaded onto tsADM, their shape changes were followed up for 14 days, and then imaged. Spheroid adhesion stability to tsADM against shear stress was also evaluated. Finally, cell delivery efficacy was compared with cell-seeded tsADM by in vivo implantation and histological evaluation. RESULTS Spheroids withstood cyclic compression stress and maintained their 3D shape without fusion after 48 h of culture in-between coverslips. Cell survival improved when spheroids were cultured on tsADM in-between the coverslips. Spheroid-loaded tsADM with coverslips maintained their spheroid outline for 14 days of culture whereas without coverslips, the group lost their outline due to spreading after 4 days in culture. Spheroids loaded onto tsADMs were more stable after six rather than 3 days in culture. Spheroid-loaded tsADMs showed about a 2.96-fold higher ASCs transplantation efficacy than cell-seeded tsADMs after 2 weeks of in vivo transplantation. CONCLUSION These results indicate that transplantation of spheroid-loaded tsADMs significantly improved cell delivery. These findings suggest that a combined approach with other cells, drugs, and nanoparticles may improve cell delivery and therapeutic efficacy.
Collapse
|
43
|
Barisic D, Erb M, Follo M, Al-Mudaris D, Rolauffs B, Hart ML. Lack of a skeletal muscle phenotype in adult human bone marrow stromal cells following xenogeneic-free expansion. Stem Cell Res Ther 2020; 11:79. [PMID: 32087752 PMCID: PMC7036219 DOI: 10.1186/s13287-020-1587-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 01/22/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background Many studies have elegantly shown that murine and rat bone marrow-derived mesenchymal stromal cells (bmMSCs) contribute to muscle regeneration and improve muscle function. Yet, the ability of transplanted human bmMSCs to manifest myogenic potential shows conflicting results. While human adipose- and umbilical cord-derived MSCs can be differentiated into a skeletal muscle phenotype using horse serum (HS), bmMSCs have only been shown to differentiate towards the skeletal muscle lineage using a complex mixture of cytokines followed by transfection with notch intracellular domain. Methods Since xenogeneic-free growth supplements are increasingly being used in the expansion of bmMSCs in clinical trials, we investigated the effects of human plasma and platelet lysate (P/PL) on the expression of neuromuscular markers and whether P/PL-expanded human bmMSCs could be differentiated towards a skeletal myogenic phenotype. Neuromuscular markers were measured using the highly sensitive droplet digital polymerase chain reaction for measuring the expression of Myf5, MyoD, MyoG, ACTA1, Desmin, GAP-43, and Coronin 1b transcripts, by performing immunofluorescence for the expression of Desmin, GAP-43, and MEF2, and flow cytometry for the expression of CD56/neural cell adhesion molecule (NCAM). Results Despite that bmMSCs expressed the myogenic regulatory factor (MRF) MEF2 after expansion in P/PL, bmMSCs cultured under such conditions did not express other essential MRFs including Myf5, MyoD, MyoG, or ACTA1 needed for myogenesis. Moreover, HS did not induce myogenesis of bmMSCs and hence did not induce the expression of any of these myogenic markers. P/PL, however, did lead to a significant increase in neurogenic GAP-43, as well as Desmin expression, and resulted in a high baseline expression of the neurogenic gene Coronin 1b which was sustained under further P/PL or HS culture conditions. Fetal bovine serum resulted in equally high levels of GAP-43 and Coronin 1b. Moreover, the proportion of CD56/NCAM-positive bmMSCs cultured in P/PL was 5.9 ± 2.1. Conclusions These data suggest that P/PL may prime a small portion of bmMSCs towards an early neural precursor cell type. Collectively, this shows that P/PL partially primes the cells towards a neurogenic phenotype, but does not prime adult human bmMSCs towards the skeletal muscle lineage.
Collapse
Affiliation(s)
- Dominik Barisic
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marita Erb
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marie Follo
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dahlia Al-Mudaris
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bernd Rolauffs
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie L Hart
- G.E.R.N. Center for Tissue Replacement, Regeneration and Neogenesis, Department of Orthopaedics and Trauma Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
44
|
Etienne J, Liu C, Skinner CM, Conboy MJ, Conboy IM. Skeletal muscle as an experimental model of choice to study tissue aging and rejuvenation. Skelet Muscle 2020; 10:4. [PMID: 32033591 PMCID: PMC7007696 DOI: 10.1186/s13395-020-0222-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/12/2020] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle is among the most age-sensitive tissues in mammal organisms. Significant changes in its resident stem cells (i.e., satellite cells, SCs), differentiated cells (i.e., myofibers), and extracellular matrix cause a decline in tissue homeostasis, function, and regenerative capacity. Based on the conservation of aging across tissues and taking advantage of the relatively well-characterization of the myofibers and associated SCs, skeletal muscle emerged as an experimental system to study the decline in function and maintenance of old tissues and to explore rejuvenation strategies. In this review, we summarize the approaches for understanding the aging process and for assaying the success of rejuvenation that use skeletal muscle as the experimental system of choice. We further discuss (and exemplify with studies of skeletal muscle) how conflicting results might be due to variations in the techniques of stem cell isolation, differences in the assays of functional rejuvenation, or deciding on the numbers of replicates and experimental cohorts.
Collapse
Affiliation(s)
- Jessy Etienne
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Chao Liu
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Colin M Skinner
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA.
| |
Collapse
|
45
|
Mierzejewski B, Archacka K, Grabowska I, Florkowska A, Ciemerych MA, Brzoska E. Human and mouse skeletal muscle stem and progenitor cells in health and disease. Semin Cell Dev Biol 2020; 104:93-104. [PMID: 32005567 DOI: 10.1016/j.semcdb.2020.01.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/15/2020] [Accepted: 01/15/2020] [Indexed: 12/25/2022]
Abstract
The proper functioning of tissues and organs depends on their ability to self-renew and repair. Some of the tissues, like epithelia, renew almost constantly while in the others this process is induced by injury or diseases. The stem or progenitor cells responsible for tissue homeostasis have been identified in many organs. Some of them, such as hematopoietic or intestinal epithelium stem cells, are multipotent and can differentiate into various cell types. Others are unipotent. The skeletal muscle tissue does not self-renew spontaneously, however, it presents unique ability to regenerate in response to the injury or disease. Its repair almost exclusively relies on unipotent satellite cells. However, multiple lines of evidence document that some progenitor cells present in the muscle can be supportive for skeletal muscle regeneration. Here, we summarize the current knowledge on the complicated landscape of stem and progenitor cells that exist in skeletal muscle and support its regeneration. We compare the cells from two model organisms, i.e., mouse and human, documenting their similarities and differences and indicating methods to test their ability to undergo myogenic differentiation.
Collapse
Affiliation(s)
- Bartosz Mierzejewski
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Karolina Archacka
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Iwona Grabowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Anita Florkowska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Maria Anna Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland
| | - Edyta Brzoska
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1St, 02-096 Warsaw, Poland.
| |
Collapse
|
46
|
Tey SR, Robertson S, Lynch E, Suzuki M. Coding Cell Identity of Human Skeletal Muscle Progenitor Cells Using Cell Surface Markers: Current Status and Remaining Challenges for Characterization and Isolation. Front Cell Dev Biol 2019; 7:284. [PMID: 31828070 PMCID: PMC6890603 DOI: 10.3389/fcell.2019.00284] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/01/2019] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle progenitor cells (SMPCs), also called myogenic progenitors, have been studied extensively in recent years because of their promising therapeutic potential to preserve and recover skeletal muscle mass and function in patients with cachexia, sarcopenia, and neuromuscular diseases. SMPCs can be utilized to investigate the mechanisms of natural and pathological myogenesis via in vitro modeling and in vivo experimentation. While various types of SMPCs are currently available from several sources, human pluripotent stem cells (PSCs) offer an efficient and cost-effective method to derive SMPCs. As human PSC-derived cells often display varying heterogeneity in cell types, cell enrichment using cell surface markers remains a critical step in current procedures to establish a pure population of SMPCs. Here we summarize the cell surface markers currently being used to detect human SMPCs, describing their potential application for characterizing, identifying and isolating human PSC-derived SMPCs. To date, several positive and negative markers have been used to enrich human SMPCs from differentiated PSCs by cell sorting. A careful analysis of current findings can broaden our understanding and reveal potential uses for these surface markers with SMPCs.
Collapse
Affiliation(s)
- Sin-Ruow Tey
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States
| | - Eileen Lynch
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States
| | - Masatoshi Suzuki
- Department of Comparative Biosciences, University of Wisconsin, Madison, WI, United States.,The Stem Cell and Regenerative Medicine Center, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
47
|
Sun C, Serra C, Lee G, Wagner KR. Stem cell-based therapies for Duchenne muscular dystrophy. Exp Neurol 2019; 323:113086. [PMID: 31639376 DOI: 10.1016/j.expneurol.2019.113086] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 02/08/2023]
Abstract
Muscular dystrophies are a group of genetic muscle disorders that cause progressive muscle weakness and degeneration. Within this group, Duchenne muscular dystrophy (DMD) is the most common and one of the most severe. DMD is an X chromosome linked disease that occurs to 1 in 3500 to 1 in 5000 boys. The cause of DMD is a mutation in the dystrophin gene, whose encoded protein provides both structural support and cell signaling capabilities. So far, there are very limited therapeutic options available and there is no cure for this disease. In this review, we discuss the existing cell therapy research, especially stem cell-based, which utilize myoblasts, satellite cells, bone marrow cells, mesoangioblasts and CD133+ cells. Finally, we focus on human pluripotent stem cells (hPSCs) which hold great potential in treating DMD. hPSCs can be used for autologous transplantation after being specified to a myogenic lineage. Over the last few years, there has been a rapid development of isolation, as well as differentiation, techniques in order to achieve effective transplantation results of myogenic cells specified from hPSCs. In this review, we summarize the current methods of hPSCs myogenic commitment/differentiation, and describe the current status of hPSC-derived myogenic cell transplantation.
Collapse
Affiliation(s)
- Congshan Sun
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, MD 21205, USA.
| | - Carlo Serra
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kathryn R Wagner
- Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA; Center for Genetic Muscle Disorders, Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, MD 21205, USA
| |
Collapse
|
48
|
Peng D, Yuan H, Liu T, Wang T, Reed-Maldonado AB, Kang N, Banie L, Wang G, Tang Y, He L, Lin G, Lue TF. Smooth Muscle Differentiation of Penile Stem/Progenitor Cells Induced by Microenergy Acoustic Pulses In Vitro. J Sex Med 2019; 16:1874-1884. [PMID: 31585805 DOI: 10.1016/j.jsxm.2019.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/08/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Modulating tissue-resident stem and progenitor cells with a non-invasive, mechanobiological intervention is an optimal approach for tissue regeneration. Stem cell antigen-1 (Sca-1) has been identified as a stem cell marker within many organs but never within the penis. AIM To localize and isolate penile stem/progenitor cells (PSPCs) and to evaluate cellular differentiation after exposure to induction medium and microenergy acoustic pulse (MAP) therapy. METHODS Six male Sprague-Dawley rats were used to isolate PSPCs. Isolation was followed by stem cell characterization and differentiation assays. The PSPCs were then treated with MAP (0.033 mJ/mm2, 1 Hz) at various dosages (25, 50, 100, and 200 pulses) and for different durations (1, 2, 4, 6, or 8 hours) in vitro. MAIN OUTCOME MEASURE The PSPCs (Sca-1-positive cells) were isolated using the magnetic-activated cell sorting system. PSPC cellular differentiation was assessed after induction with induction medium and with MAP in vitro. Wnt/β-catenin signaling was also assayed. RESULTS The PSPCs were successfully localized within the penile subtunic and perisinusoidal spaces, and they were successfully isolated using magnetic-activated cell sorting. The stemness of the cells was confirmed by stem cell marker characterization and by multiple differentiation into smooth muscle cells, endothelial cells, adipocytes, and neurons. MAP-induced PSPCs differentiated into smooth muscle cells by activating the Wnt/β-catenin signaling pathway in a time- and dosage-dependent manner. CLINICAL IMPLICATIONS By modulating resident PSPCs, MAP may have utility in the treatment of erectile dysfunction (ED). STRENGTHS & LIMITATIONS This study provides solid evidence in support of microenergy therapies, including both MAP and low-intensity extracorporeal shock wave therapy, for the treatment of ED. Additional studies are needed and should include additional stem cells markers. Furthermore, studies exploring the underling mechanisms for PSPC activation and differentiation are required. CONCLUSION PSPCs were successfully identified, localized, and isolated. Additionally, MAP provoked PSPCs to differentiate into smooth muscle cells via the Wnt/β-catenin signaling pathway. As such, MAP provides a novel method for activating endogenous tissue-resident stem/progenitor cells and might facilitate stem cell regenerative therapy targeting ED. Peng D, Yuan H, Liu T, et al. Smooth Muscle Differentiation of Penile Stem/Progenitor Cells Induced by Microenergy Acoustic Pulses In Vitro. J Sex Med 2019; 16:1874-1884.
Collapse
Affiliation(s)
- Dongyi Peng
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA; Department of Urology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Huixing Yuan
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Tianshu Liu
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Tianyu Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Amanda B Reed-Maldonado
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Ning Kang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Yuxin Tang
- Department of Urology, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Leye He
- Department of Urology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|
49
|
Enhancement of meat production by environmental manipulations in embryo and young broilers. WORLD POULTRY SCI J 2019. [DOI: 10.1017/s0043933906001103] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
50
|
Potential Therapies Using Myogenic Stem Cells Combined with Bio-Engineering Approaches for Treatment of Muscular Dystrophies. Cells 2019; 8:cells8091066. [PMID: 31514443 PMCID: PMC6769835 DOI: 10.3390/cells8091066] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/06/2019] [Accepted: 09/10/2019] [Indexed: 12/31/2022] Open
Abstract
Muscular dystrophies (MDs) are a group of heterogeneous genetic disorders caused by mutations in the genes encoding the structural components of myofibres. The current state-of-the-art treatment is oligonucleotide-based gene therapy that restores disease-related protein. However, this therapeutic approach has limited efficacy and is unlikely to be curative. While the number of studies focused on cell transplantation therapy has increased in the recent years, this approach remains challenging due to multiple issues related to the efficacy of engrafted cells, source of myogenic cells, and systemic injections. Technical innovation has contributed to overcoming cell source challenges, and in recent studies, a combination of muscle resident stem cells and gene editing has shown promise as a novel approach. Furthermore, improvement of the muscular environment both in cultured donor cells and in recipient MD muscles may potentially facilitate cell engraftment. Artificial skeletal muscle generated by myogenic cells and muscle resident cells is an alternate approach that may enable the replacement of damaged tissues. Here, we review the current status of myogenic stem cell transplantation therapy, describe recent advances, and discuss the remaining obstacles that exist in the search for a cure for MD patients.
Collapse
|