Journal articles on the topic 'Human Skeletal muscle derived stem cells'

To see the other types of publications on this topic, follow the link: Human Skeletal muscle derived stem cells.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Human Skeletal muscle derived stem cells.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Tchao, Jason, Jong Jin Kim, Bo Lin, Guy Salama, Cecilia W. Lo, Lei Yang, and Kimimasa Tobita. "Engineered Human Muscle Tissue from Skeletal Muscle Derived Stem Cells and Induced Pluripotent Stem Cell Derived Cardiac Cells." International Journal of Tissue Engineering 2013 (December 5, 2013): 1–15. http://dx.doi.org/10.1155/2013/198762.

Full text
Abstract:
During development, cardiac and skeletal muscle share major transcription factors and sarcomere proteins which were generally regarded as specific to either cardiac or skeletal muscle but not both in terminally differentiated adult cardiac or skeletal muscle. Here, we investigated whether artificial muscle constructed from human skeletal muscle derived stem cells (MDSCs) recapitulates developmental similarities between cardiac and skeletal muscle. We constructed 3-dimensional collagen-based engineered muscle tissue (EMT) using MDSCs (MDSC-EMT) and compared the biochemical and contractile properties with EMT using induced pluripotent stem (iPS) cell-derived cardiac cells (iPS-EMT). Both MDSC-EMT and iPS-EMT expressed cardiac specific troponins, fast skeletal muscle myosin heavy chain, and connexin-43 mimicking developing cardiac or skeletal muscle. At the transcriptional level, MDSC-EMT and iPS-EMT upregulated both cardiac and skeletal muscle-specific genes and expressed Nkx2.5 and Myo-D proteins. MDSC-EMT displayed intracellular calcium ion transients and responses to isoproterenol. Contractile force measurements of MDSC-EMT demonstrated functional properties of immature cardiac and skeletal muscle in both tissues. Results suggest that the EMT from MDSCs mimics developing cardiac and skeletal muscle and can serve as a useful in vitro functioning striated muscle model for investigation of stem cell differentiation and therapeutic options of MDSCs for cardiac repair.
APA, Harvard, Vancouver, ISO, and other styles
2

Metzler, Eric, Helena Escobar, Daniele Yumi Sunaga-Franze, Sascha Sauer, Sebastian Diecke, and Simone Spuler. "Generation of hiPSC-Derived Skeletal Muscle Cells: Exploiting the Potential of Skeletal Muscle-Derived hiPSCs." Biomedicines 10, no. 5 (May 23, 2022): 1204. http://dx.doi.org/10.3390/biomedicines10051204.

Full text
Abstract:
Cell therapies for muscle wasting disorders are on the verge of becoming a realistic clinical perspective. Muscle precursor cells derived from human induced pluripotent stem cells (hiPSCs) represent the key to unrestricted cell numbers indispensable for the treatment of generalized muscle wasting such as cachexia or intensive care unit (ICU)-acquired weakness. We asked how the cell of origin influences efficacy and molecular properties of hiPSC-derived muscle progenitor cells. We generated hiPSCs from primary muscle stem cells and from peripheral blood mononuclear cells (PBMCs) of the same donors (n = 4) and compared their molecular profiles, myogenic differentiation potential, and ability to generate new muscle fibers in vivo. We show that reprogramming into hiPSCs from primary muscle stem cells was faster and 35 times more efficient than from blood cells. Global transcriptome comparison revealed significant differences, but differentiation into induced myogenic cells using a directed transgene-free approach could be achieved with muscle- and PBMC-derived hiPSCs, and both cell types generated new muscle fibers in vivo. Differences in myogenic differentiation efficiency were identified with hiPSCs generated from individual donors. The generation of muscle-stem-cell-derived hiPSCs is a fast and economic method to obtain unrestricted cell numbers for cell-based therapies in muscle wasting disorders, and in this aspect are superior to blood-derived hiPSCs.
APA, Harvard, Vancouver, ISO, and other styles
3

Sato, Takahiko. "Induction of Skeletal Muscle Progenitors and Stem Cells from human induced Pluripotent Stem Cells." Journal of Neuromuscular Diseases 7, no. 4 (September 18, 2020): 395–405. http://dx.doi.org/10.3233/jnd-200497.

Full text
Abstract:
Induced pluripotent stem cells (iPSCs) have the potential to differentiate into various types of cells and tissues including skeletal muscle. The approach to convert these stem cells into skeletal muscle cells offers hope for patients afflicted with skeletal muscle diseases such as Duchenne muscular dystrophy (DMD). Several methods have been reported to induce myogenic differentiation with iPSCs derived from myogenic patients. An important point for generating skeletal muscle cells from iPSCs is to understand in vivo myogenic induction in development and regeneration. Current protocols of myogenic induction utilize techniques with overexpression of myogenic transcription factors such as Myod1(MyoD), Pax3, Pax7, and others, using recombinant proteins or small molecules to induce mesodermal cells followed by myogenic progenitors, and adult muscle stem cells. This review summarizes the current approaches used for myogenic induction and highlights recent improvements.
APA, Harvard, Vancouver, ISO, and other styles
4

Pappas, Matthew P., Ning Xie, Jacqueline S. Penaloza, and Sunny S. K. Chan. "Defining the Skeletal Myogenic Lineage in Human Pluripotent Stem Cell-Derived Teratomas." Cells 11, no. 9 (May 9, 2022): 1589. http://dx.doi.org/10.3390/cells11091589.

Full text
Abstract:
Skeletal muscle stem cells are essential to muscle homeostasis and regeneration after injury, and have emerged as a promising cell source for treating skeletal disorders. An attractive approach to obtain these cells utilizes differentiation of pluripotent stem cells (PSCs). We recently reported that teratomas derived from mouse PSCs are a rich source of skeletal muscle stem cells. Here, we showed that teratoma formation is also capable of producing skeletal myogenic progenitors from human PSCs. Using single-cell transcriptomics, we discovered several distinct skeletal myogenic subpopulations that represent progressive developmental stages of the skeletal myogenic lineage and recapitulate human embryonic skeletal myogenesis. We further discovered that ERBB3 and CD82 are effective surface markers for prospective isolation of the skeletal myogenic lineage in human PSC-derived teratomas. Therefore, teratoma formation provides an accessible model for obtaining human skeletal myogenic progenitors from PSCs.
APA, Harvard, Vancouver, ISO, and other styles
5

Jiwlawat, Nunnapas, Eileen Lynch, Jeremy Jeffrey, Jonathan M. Van Dyke, and Masatoshi Suzuki. "Current Progress and Challenges for Skeletal Muscle Differentiation from Human Pluripotent Stem Cells Using Transgene-Free Approaches." Stem Cells International 2018 (2018): 1–18. http://dx.doi.org/10.1155/2018/6241681.

Full text
Abstract:
Neuromuscular diseases are caused by functional defects of skeletal muscles, directly via muscle pathology or indirectly via disruption of the nervous system. Extensive studies have been performed to improve the outcomes of therapies; however, effective treatment strategies have not been fully established for any major neuromuscular disease. Human pluripotent stem cells have a great capacity to differentiate into myogenic progenitors and skeletal myocytes for use in treating and modeling neuromuscular diseases. Recent advances have allowed the creation of patient-derived stem cells, which can be used as a unique platform for comprehensive study of disease mechanisms, in vitro drug screening, and potential new cell-based therapies. In the last decade, a number of methods have been developed to derive skeletal muscle cells from human pluripotent stem cells. By controlling the process of myogenesis using transcription factors and signaling molecules, human pluripotent stem cells can be directed to differentiate into cell types observed during muscle development. In this review, we highlight signaling pathways relevant to the formation of muscle tissue during embryonic development. We then summarize current methods to differentiate human pluripotent stem cells toward the myogenic lineage, specifically focusing on transgene-free approaches. Lastly, we discuss existing challenges for deriving skeletal myocytes and myogenic progenitors from human pluripotent stem cells.
APA, Harvard, Vancouver, ISO, and other styles
6

Bisleri, C., C. Alessandri, G. Invernici, A. Negri, J. Manfredi, A. Caruso, and C. Muneretto. "HUMAN SKELETAL MUSCLE-DERIVED STEM CELLS FOR MYOCAR-DIAL REGENERATION." ASAIO Journal 50, no. 2 (March 2004): 171. http://dx.doi.org/10.1097/00002480-200403000-00239.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Stern-Straeter, Jens, Juritz Stephanie, Gregor Bran, Frank Riedel, Haneen Sadick, Karl Hörmann, and Ulrich R. Goessler. "Skeletal Muscle Regeneration: MSC versus Satellite Cells." Otolaryngology–Head and Neck Surgery 139, no. 2_suppl (August 2008): P86. http://dx.doi.org/10.1016/j.otohns.2008.05.484.

Full text
Abstract:
Problem Differentiating stem cells into the myogenic linage in order to create functional muscle tissue is a challenging endeavour. In this work, adipose-derived mesenchymal stem cells (MSC) and satellite cells derived from muscle biopsies were compared regarding proliferation and myogenic differentiation potential under standardized cell culture conditions. This data was obtained in order to discover the most promising type of stem cell for regeneration of muscle tissue and to determine the optimal culture conditions for later clinical use. Methods Human MSC were isolated from adipose tissue, and primary human skeletal myoblasts were extracted from muscle biopsies by enzymatic digestion. Proliferation was analysed using the AlamarBlue® assay. Gene expression of marker genes – such as Myogenin, Myo D, Myf 5 and MHC – were analysed by RT-PCR. Immunostainings against desmin and sarcomeric-actin were performed as differentiation markers. Results MSC cell cultures showed a greater proliferation rate compared with satellite cell cultures. In both stem cell cultures, myogenic differentiation/heritage could be verified by immunostainings against the muscle-specific marker desmin. Gene expression and protein analysis revealed a more stable differentiation of human satellite cell cultures. Conclusion Characterization of both human MSC cultures and satellite cell cultures – and thereby an understanding of myogenesis – might lead to their clinical usage in skeletal muscle tissue engineering. The results in this study appear to indicate that human satellite cell cultures have a more stable differentiation under in vitro conditions and that they might offer a greater potential for skeletal muscle tissue engineering purposes. Significance Our study contributes to the understanding of myogenic differentiation of MSC and satellite cells and helps to improve culture systems for later clinical utilization.
APA, Harvard, Vancouver, ISO, and other styles
8

Xie, Ning, Sabrina N. Chu, Cassandra B. Schultz, and Sunny S. K. Chan. "Efficient Muscle Regeneration by Human PSC-Derived CD82+ ERBB3+ NGFR+ Skeletal Myogenic Progenitors." Cells 12, no. 3 (January 18, 2023): 362. http://dx.doi.org/10.3390/cells12030362.

Full text
Abstract:
Differentiation of pluripotent stem cells (PSCs) is a promising approach to obtaining large quantities of skeletal myogenic progenitors for disease modeling and cell-based therapy. However, generating skeletal myogenic cells with high regenerative potential is still challenging. We recently reported that skeletal myogenic progenitors generated from mouse PSC-derived teratomas possess robust regenerative potency. We have also found that teratomas derived from human PSCs contain a skeletal myogenic population. Here, we showed that these human PSC-derived skeletal myogenic progenitors had exceptional engraftability. A combination of cell surface markers, CD82, ERBB3, and NGFR enabled efficient purification of skeletal myogenic progenitors. These cells expressed PAX7 and were able to differentiate into MHC+ multinucleated myotubes. We further discovered that these cells are expandable in vitro. Upon transplantation, the expanded cells formed new dystrophin+ fibers that reconstituted almost ¾ of the total muscle volume, and repopulated the muscle stem cell pool. Our study, therefore, demonstrates the possibility of producing large quantities of engraftable skeletal myogenic cells from human PSCs.
APA, Harvard, Vancouver, ISO, and other styles
9

Yoshioka, Kiyoshi, Hiroshi Nagahisa, Fumihito Miura, Hiromitsu Araki, Yasutomi Kamei, Yasuo Kitajima, Daiki Seko, et al. "Hoxa10 mediates positional memory to govern stem cell function in adult skeletal muscle." Science Advances 7, no. 24 (June 2021): eabd7924. http://dx.doi.org/10.1126/sciadv.abd7924.

Full text
Abstract:
Muscle stem cells (satellite cells) are distributed throughout the body and have heterogeneous properties among muscles. However, functional topographical genes in satellite cells of adult muscle remain unidentified. Here, we show that expression of Homeobox-A (Hox-A) cluster genes accompanied with DNA hypermethylation of the Hox-A locus was robustly maintained in both somite-derived muscles and their associated satellite cells in adult mice, which recapitulates their embryonic origin. Somite-derived satellite cells were clearly separated from cells derived from cranial mesoderm in Hoxa10 expression. Hoxa10 inactivation led to genomic instability and mitotic catastrophe in somite-derived satellite cells in mice and human. Satellite cell–specific Hoxa10 ablation in mice resulted in a decline in the regenerative ability of somite-derived muscles, which were unobserved in cranial mesoderm–derived muscles. Thus, our results show that Hox gene expression profiles instill the embryonic history in satellite cells as positional memory, potentially modulating region-specific pathophysiology in adult muscles.
APA, Harvard, Vancouver, ISO, and other styles
10

Byun, Seong-Eun, Changgon Sim, Yoonhui Chung, Hyung Kyung Kim, Sungmoon Park, Do Kyung Kim, Seongmin Cho, and Soonchul Lee. "Skeletal Muscle Regeneration by the Exosomes of Adipose Tissue-Derived Mesenchymal Stem Cells." Current Issues in Molecular Biology 43, no. 3 (October 9, 2021): 1473–88. http://dx.doi.org/10.3390/cimb43030104.

Full text
Abstract:
Profound skeletal muscle loss can lead to severe disability and cosmetic deformities. Mesenchymal stem cell (MSC)-derived exosomes have shown potential as an effective therapeutic tool for tissue regeneration. This study aimed to determine the regenerative capacity of MSC-derived exosomes for skeletal muscle regeneration. Exosomes were isolated from human adipose tissue-derived MSCs (AD-MSCs). The effects of MSC-derived exosomes on satellite cells were investigated using cell viability, relevant genes, and protein analyses. Moreover, NOD-SCID mice were used and randomly assigned to the healthy control (n = 4), muscle defect (n = 6), and muscle defect + exosome (n = 6) groups. Muscle defects were created using a biopsy punch on the quadriceps of the hind limb. Four weeks after the surgery, the quadriceps muscles were harvested, weighed, and histologically analyzed. MSC-derived exosome treatment increased the proliferation and expression of myocyte-related genes, and immunofluorescence analysis for myogenin revealed a similar trend. Histologically, MSC-derived exosome-treated mice showed relatively preserved shapes and sizes of the muscle bundles. Immunohistochemical staining revealed greater expression of myogenin and myoblast determination protein 1 in the MSC-derived exosome-treated group. These results indicate that exosomes extracted from AD-MSCs have the therapeutic potential for skeletal muscle regeneration.
APA, Harvard, Vancouver, ISO, and other styles
11

De Bari, Cosimo, Francesco Dell'Accio, Frank Vandenabeele, Joris R. Vermeesch, Jean-Marc Raymackers, and Frank P. Luyten. "Skeletal muscle repair by adult human mesenchymal stem cells from synovial membrane." Journal of Cell Biology 160, no. 6 (March 10, 2003): 909–18. http://dx.doi.org/10.1083/jcb.200212064.

Full text
Abstract:
We have demonstrated previously that adult human synovial membrane-derived mesenchymal stem cells (hSM-MSCs) have myogenic potential in vitro (De Bari, C., F. Dell'Accio, P. Tylzanowski, and F.P. Luyten. 2001. Arthritis Rheum. 44:1928–1942). In the present study, we have characterized their myogenic differentiation in a nude mouse model of skeletal muscle regeneration and provide proof of principle of their potential use for muscle repair in the mdx mouse model of Duchenne muscular dystrophy. When implanted into regenerating nude mouse muscle, hSM-MSCs contributed to myofibers and to long term persisting functional satellite cells. No nuclear fusion hybrids were observed between donor human cells and host mouse muscle cells. Myogenic differentiation proceeded through a molecular cascade resembling embryonic muscle development. Differentiation was sensitive to environmental cues, since hSM-MSCs injected into the bloodstream engrafted in several tissues, but acquired the muscle phenotype only within skeletal muscle. When administered into dystrophic muscles of immunosuppressed mdx mice, hSM-MSCs restored sarcolemmal expression of dystrophin, reduced central nucleation, and rescued the expression of mouse mechano growth factor.
APA, Harvard, Vancouver, ISO, and other styles
12

Meng, Jinhong, John Counsell, and Jennifer E. Morgan. "Effects of Mini-Dystrophin on Dystrophin-Deficient, Human Skeletal Muscle-Derived Cells." International Journal of Molecular Sciences 21, no. 19 (September 28, 2020): 7168. http://dx.doi.org/10.3390/ijms21197168.

Full text
Abstract:
Background: We are developing a novel therapy for Duchenne muscular dystrophy (DMD), involving the transplantation of autologous, skeletal muscle-derived stem cells that have been genetically corrected to express dystrophin. Dystrophin is normally expressed in activated satellite cells and in differentiated muscle fibres. However, in past preclinical validation studies, dystrophin transgenes have generally been driven by constitutive promoters that would be active at every stage of the myogenic differentiation process, including in proliferating muscle stem cells. It is not known whether artificial dystrophin expression would affect the properties of these cells. Aims: Our aims are to determine if mini-dystrophin expression affects the proliferation or myogenic differentiation of DMD skeletal muscle-derived cells. Methods: Skeletal muscle-derived cells from a DMD patient were transduced with lentivirus coding for mini-dystrophins (R3–R13 spectrin-like repeats (ΔR3R13) or hinge2 to spectrin-like repeats R23 (ΔH2R23)) with EGFP (enhanced green fluorescence protein) fused to the C-terminus, driven by a constitutive promoter, spleen focus-forming virus (SFFV). Transduced cells were purified on the basis of GFP expression. Their proliferation and myogenic differentiation were quantified by ethynyl deoxyuridine (EdU) incorporation and fusion index. Furthermore, dystrophin small interfering ribonucleic acids (siRNAs) were transfected to the cells to reverse the effects of the mini-dystrophin. Finally, a phospho-mitogen-activated protein kinase (MAPK) array assay was performed to investigate signalling pathway changes caused by dystrophin expression. Results: Cell proliferation was not affected in cells transduced with ΔR3R13, but was significantly increased in cells transduced with ΔH2R23. The fusion index of myotubes derived from both ΔR3R13- and ΔH2R23 -expressing cells was significantly compromised in comparison to myotubes derived from non-transduced cells. Dystrophin siRNA transfection restored the differentiation of ΔH2R23-expressing cells. The Erk1/2- signalling pathway is altered in cells transduced with mini-dystrophin constructs. Conclusions: Ectopic expression of dystrophin in cultured human skeletal muscle-derived cells may affect their proliferation and differentiation capacity. Caution should be taken when considering genetic correction of autologous stem cells to express dystrophin driven by a constitutive promoter.
APA, Harvard, Vancouver, ISO, and other styles
13

Shin, Min-Kyoung, Jin Seok Bang, Jeoung Eun Lee, Hoang-Dai Tran, Genehong Park, Dong Ryul Lee, and Junghyun Jo. "Generation of Skeletal Muscle Organoids from Human Pluripotent Stem Cells to Model Myogenesis and Muscle Regeneration." International Journal of Molecular Sciences 23, no. 9 (May 4, 2022): 5108. http://dx.doi.org/10.3390/ijms23095108.

Full text
Abstract:
In vitro organoids derived from human pluripotent stem cells (hPSCs) have been developed as essential tools to study the underlying mechanisms of human development and diseases owing to their structural and physiological similarity to corresponding organs. Despite recent advances, there are a few methodologies for three-dimensional (3D) skeletal muscle differentiation, which focus on the terminal differentiation into myofibers and investigate the potential of modeling neuromuscular disorders and muscular dystrophies. However, these methodologies cannot recapitulate the developmental processes and lack regenerative capacity. In this study, we developed a new method to differentiate hPSCs into a 3D human skeletal muscle organoid (hSkMO). This organoid model could recapitulate the myogenesis process and possesses regenerative capacities of sustainable satellite cells (SCs), which are adult muscle stem/progenitor cells capable of self-renewal and myogenic differentiation. Our 3D model demonstrated myogenesis through the sequential occurrence of multiple myogenic cell types from SCs to myocytes. Notably, we detected quiescent, non-dividing SCs throughout the hSkMO differentiation in long-term culture. They were activated and differentiated to reconstitute muscle tissue upon damage. Thus, hSkMOs can recapitulate human skeletal muscle development and regeneration and may provide a new model for studying human skeletal muscles and related diseases.
APA, Harvard, Vancouver, ISO, and other styles
14

Ruan, Travis, Dylan Harney, Yen Chin Koay, Lipin Loo, Mark Larance, and Leslie Caron. "Anabolic Factors and Myokines Improve Differentiation of Human Embryonic Stem Cell Derived Skeletal Muscle Cells." Cells 11, no. 6 (March 11, 2022): 963. http://dx.doi.org/10.3390/cells11060963.

Full text
Abstract:
Skeletal muscle weakness is linked to many adverse health outcomes. Current research to identify new drugs has often been inconclusive due to lack of adequate cellular models. We previously developed a scalable monolayer system to differentiate human embryonic stem cells (hESCs) into mature skeletal muscle cells (SkMCs) within 26 days without cell sorting or genetic manipulation. Here, building on our previous work, we show that differentiation and fusion of myotubes can be further enhanced using the anabolic factors testosterone (T) and follistatin (F) in combination with a cocktail of myokines (C). Importantly, combined TFC treatment significantly enhanced both the hESC-SkMC fusion index and the expression levels of various skeletal muscle markers, including the motor protein myosin heavy chain (MyHC). Transcriptomic and proteomic analysis revealed oxidative phosphorylation as the most up-regulated pathway, and a significantly higher level of ATP and increased mitochondrial mass were also observed in TFC-treated hESC-SkMCs, suggesting enhanced energy metabolism is coupled with improved muscle differentiation. This cellular model will be a powerful tool for studying in vitro myogenesis and for drug discovery pertaining to further enhancing muscle development or treating muscle diseases.
APA, Harvard, Vancouver, ISO, and other styles
15

Iovino, Salvatore, Alison M. Burkart, Laura Warren, Mary Elizabeth Patti, and C. Ronald Kahn. "Myotubes derived from human-induced pluripotent stem cells mirror in vivo insulin resistance." Proceedings of the National Academy of Sciences 113, no. 7 (February 1, 2016): 1889–94. http://dx.doi.org/10.1073/pnas.1525665113.

Full text
Abstract:
Induced pluripotent stem cells (iPS cells) represent a unique tool for the study of the pathophysiology of human disease, because these cells can be differentiated into multiple cell types in vitro and used to generate patient- and tissue-specific disease models. Given the critical role for skeletal muscle insulin resistance in whole-body glucose metabolism and type 2 diabetes, we have created a novel cellular model of human muscle insulin resistance by differentiating iPS cells from individuals with mutations in the insulin receptor (IR-Mut) into functional myotubes and characterizing their response to insulin in comparison with controls. Morphologically, IR-Mut cells differentiated normally, but had delayed expression of some muscle differentiation-related genes. Most importantly, whereas control iPS-derived myotubes exhibited in vitro responses similar to primary differentiated human myoblasts, IR-Mut myotubes demonstrated severe impairment in insulin signaling and insulin-stimulated 2-deoxyglucose uptake and glycogen synthesis. Transcriptional regulation was also perturbed in IR-Mut myotubes with reduced insulin-stimulated expression of metabolic and early growth response genes. Thus, iPS-derived myotubes from individuals with genetically determined insulin resistance demonstrate many of the defects observed in vivo in insulin-resistant skeletal muscle and provide a new model to analyze the molecular impact of muscle insulin resistance.
APA, Harvard, Vancouver, ISO, and other styles
16

Shi, Ming, Masakazu Ishikawa, Naosuke Kamei, Tomoyuki Nakasa, Nobuo Adachi, Masataka Deie, Takayuki Asahara, and Mitsuo Ochi. "Acceleration of Skeletal Muscle Regeneration in a Rat Skeletal Muscle Injury Model by Local Injection of Human Peripheral Blood-Derived CD133-Positive Cells." STEM CELLS 27, no. 4 (January 15, 2009): 949–60. http://dx.doi.org/10.1002/stem.4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
17

Wang, Lei, Brandon L. Walker, Devang Bhatt, Patrick J. Kennedy, and William T. Tse. "Clonal Dedifferentiation of Human Myogenic Progenitors into Skeletal Muscle Stem Cells Induced by MAPK Inhibition." Blood 110, no. 11 (November 16, 2007): 3701. http://dx.doi.org/10.1182/blood.v110.11.3701.3701.

Full text
Abstract:
Abstract Understanding the biology of skeletal muscle stem cells can facilitate development of effective cellular therapy for muscle diseases such as muscular dystrophy. While studying human bone marrow stromal cells, we identified a stromal cell subclone, WB15-M, which has developed spontaneously into a skeletal muscle cell line. This subclone no longer expressed CD105, CD90 and CD44, cell surface markers typically found on bone marrow stromal cells. Instead they expressed alpha7-integrin, an antigen found on myoblasts and regenerating muscles. WB15-M cells were positive for the myogenic regulatory factors MyoD and Myf5 and, when cultured under a low-serum condition, matured into multinucleated myofibers that expressed sarcomeric alpha-actinin, myosin heavy chain, dystroglycan and dystrophin, indicating that WB15-M cells were committed myogenic progenitors. We asked if the WB15-M cells might contain skeletal muscle stem cells. Immunofluorescent microscopic studies revealed that rare WB15-M cells expressed Pax7, Pax3 and Msx1, nuclear factors found in skeletal muscle stem cells. When WB15-M cells were cultured in SB203580 or PD98059, inhibitors of the p38 and Erk mitogen-activated protein kinases (MAPK), respectively, they markedly enhanced expression of Pax7, Pax3 and Msx1. The increase in the expression of these nuclear factors could be blocked by simultaneous treatment of WB15-M cells with orthovanadate, a protein tyrosine phosphatase. Alternatively, the increase could be induced by chemically inhibiting Mnk1, a common downstream target of the p38 and Erk MAPK signaling cascades. When further stimulated with bone morphogenic protein-2, MAPK inhibitor-treated WB15-M cells acquired the ability to express alkaline phosphatase, an early osteoblast marker, a property also seen in skeletal muscle stem cells. In contrast, untreated WB15-M cells did not exhibit this property. Clonal analysis showed that the biological changes exhibited by WB15-M cells upon MAPK inhibition was not an artifact of cellular heterogeneity but the result of reversion of individual committed myogenic progenitors to stem cell-like precursors that were more primitive in their development. Purified myogenin-expressing cells that have already initiated their myogenic differentiation program could still revert clonally to these stem cell-like precursors upon MAPK inhibition, indicating a bona fide dedifferentiation process and a true reversal of developmental fate. When WB15-M cells treated with MAPK inhibitors were cultured clonally under conditions that promoted both myogenic and osteogenic development, they formed colonies that expressed either myogenin or alkaline phosphatase but not both; untreated WB15-M cells cultured under the same conditions formed only myogenin-expressing colonies. In conclusion, we found that human bone marrow stromal cell-derived myogenic progenitors could be induced by MAPK inhibition to dedifferentiate into precursors that exhibited properties of the skeletal muscle stem cells. This finding should facilitate the development of novel cellular therapy that utilizes skeletal muscle stem cells.
APA, Harvard, Vancouver, ISO, and other styles
18

Chen, Chien-Wen, Mirko Corselli, Bruno Péault, and Johnny Huard. "Human Blood-Vessel-Derived Stem Cells for Tissue Repair and Regeneration." Journal of Biomedicine and Biotechnology 2012 (2012): 1–9. http://dx.doi.org/10.1155/2012/597439.

Full text
Abstract:
Multipotent stem/progenitor cells with similar developmental potentials have been independently identified from diverse human tissue/organ cultures. The increasing recognition of the vascular/perivascular origin of mesenchymal precursors suggested blood vessels being a systemic source of adult stem/progenitor cells. Our group and other laboratories recently isolated multiple stem/progenitor cell subsets from blood vessels of adult human tissues. Each of the three structural layers of blood vessels: intima, media, and adventitia has been found to include at least one precursor population, that is, myogenic endothelial cells (MECs), pericytes, and adventitial cells (ACs), respectively. MECs and pericytes efficiently regenerate myofibers in injured and dystrophic skeletal muscles as well as improve cardiac function after myocardial infarction. The applications of ACs in vascular remodeling and angiogenesis/vasculogenesis have been examined. Our recent finding that MECs and pericytes can be purified from cryogenically banked human primary muscle cell culture further indicates their potential applications in personalized regenerative medicine.
APA, Harvard, Vancouver, ISO, and other styles
19

Hejbøl, Eva K., Mohammad A. Hajjaj, Ole Nielsen, and Henrik D. Schrøder. "Marker Expression of Interstitial Cells in Human Skeletal Muscle: An Immunohistochemical Study." Journal of Histochemistry & Cytochemistry 67, no. 11 (August 14, 2019): 825–44. http://dx.doi.org/10.1369/0022155419871033.

Full text
Abstract:
There is a growing recognition that myogenic stem cells are influenced by their microenvironment during regeneration. Several interstitial cell types have been described as supportive for myoblasts. In this role, both the pericyte as a possible progenitor for mesenchymal stem cells, and interstitial cells in the endomysium have been discussed. We have applied immunohistochemistry on normal and pathological human skeletal muscle using markers for pericytes, or progenitor cells and found a cell type co-expressing CD10, CD34, CD271, and platelet-derived growth factor receptor α omnipresent in the endomysium. The marker profile of these cells changed dynamically in response to muscle damage and atrophy, and they proliferated in response to damage. The cytology and expression profile of the CD10+ cells indicated a capacity to participate in myogenesis. Both morphology and indicated function of these cells matched properties of several previously described interstitial cell types. Our study suggests a limited number of cell types that could embrace many of these described cell types. Our study indicate that the CD10+, CD34+, CD271+, and platelet-derived growth factor receptor α+ cells could have a supportive role in human muscle regeneration, and thus the mechanisms by which they exert their influence could be implemented in stem cell therapy.
APA, Harvard, Vancouver, ISO, and other styles
20

Wei, Yan, Yuan Li, Chao Chen, Katharina Stoelzel, Andreas M. Kaufmann, and Andreas E. Albers. "Human skeletal muscle-derived stem cells retain stem cell properties after expansion in myosphere culture." Experimental Cell Research 317, no. 7 (April 2011): 1016–27. http://dx.doi.org/10.1016/j.yexcr.2011.01.019.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Yamakawa, Hiroyuki, Dai Kusumoto, Hisayuki Hashimoto, and Shinsuke Yuasa. "Stem Cell Aging in Skeletal Muscle Regeneration and Disease." International Journal of Molecular Sciences 21, no. 5 (March 6, 2020): 1830. http://dx.doi.org/10.3390/ijms21051830.

Full text
Abstract:
Skeletal muscle comprises 30–40% of the weight of a healthy human body and is required for voluntary movements in humans. Mature skeletal muscle is formed by multinuclear cells, which are called myofibers. Formation of myofibers depends on the proliferation, differentiation, and fusion of muscle progenitor cells during development and after injury. Muscle progenitor cells are derived from muscle satellite (stem) cells (MuSCs), which reside on the surface of the myofiber but beneath the basement membrane. MuSCs play a central role in postnatal maintenance, growth, repair, and regeneration of skeletal muscle. In sedentary adult muscle, MuSCs are mitotically quiescent, but are promptly activated in response to muscle injury. Physiological and chronological aging induces MuSC aging, leading to an impaired regenerative capability. Importantly, in pathological situations, repetitive muscle injury induces early impairment of MuSCs due to stem cell aging and leads to early impairment of regeneration ability. In this review, we discuss (1) the role of MuSCs in muscle regeneration, (2) stem cell aging under physiological and pathological conditions, and (3) prospects related to clinical applications of controlling MuSCs.
APA, Harvard, Vancouver, ISO, and other styles
22

Nowaczyk, Magdalena, Agnieszka Malcher, Agnieszka Zimna, Natalia Rozwadowska, and Maciej Kurpisz. "Effect of miR-195 inhibition on human skeletal muscle-derived stem/progenitor cells." Kardiologia Polska 80, no. 7-8 (August 31, 2022): 813–24. http://dx.doi.org/10.33963/kp.a2022.0127.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Choi, Alee, Sang Eon Park, Jang Bin Jeong, Suk-joo Choi, Soo-young Oh, Gyu Ha Ryu, Jeehun Lee, Hong Bae Jeon, and Jong Wook Chang. "Anti-Fibrotic Effect of Human Wharton’s Jelly-Derived Mesenchymal Stem Cells on Skeletal Muscle Cells, Mediated by Secretion of MMP-1." International Journal of Molecular Sciences 21, no. 17 (August 29, 2020): 6269. http://dx.doi.org/10.3390/ijms21176269.

Full text
Abstract:
Extracellular matrix (ECM) components play an important role in maintaining skeletal muscle function, but excessive accumulation of ECM components interferes with skeletal muscle regeneration after injury, eventually inducing fibrosis. Increased oxidative stress level caused by dystrophin deficiency is a key factor in fibrosis in Duchenne muscular dystrophy (DMD) patients. Mesenchymal stem cells (MSCs) are considered a promising therapeutic agent for various diseases involving fibrosis. In particular, the paracrine factors secreted by MSCs play an important role in the therapeutic effects of MSCs. In this study, we investigated the effects of MSCs on skeletal muscle fibrosis. In 2–5-month-old mdx mice intravenously injected with 1 × 105 Wharton’s jelly (WJ)-derived MSCs (WJ-MSCs), fibrosis intensity and accumulation of calcium/necrotic fibers were significantly decreased. To elucidate the mechanism of this effect, we verified the effect of WJ-MSCs in a hydrogen peroxide-induced fibrosis myotubes model. In addition, we demonstrated that matrix metalloproteinase-1 (MMP-1), a paracrine factor, is critical for this anti-fibrotic effect of WJ-MSCs. These findings demonstrate that WJ-MSCs exert anti-fibrotic effects against skeletal muscle fibrosis, primarily via MMP-1, indicating a novel target for the treatment of muscle diseases, such as DMD.
APA, Harvard, Vancouver, ISO, and other styles
24

Yamanaka, Yukito, Nana Takenaka, Hidetoshi Sakurai, Morio Ueno, Shigeru Kinoshita, Chie Sotozono, and Takahiko Sato. "Human Skeletal Muscle Cells Derived from the Orbicularis Oculi Have Regenerative Capacity for Duchenne Muscular Dystrophy." International Journal of Molecular Sciences 20, no. 14 (July 14, 2019): 3456. http://dx.doi.org/10.3390/ijms20143456.

Full text
Abstract:
Skeletal muscle stem cells (MuSCs) have been proposed as suitable candidates for cell therapy in muscular disorders since they exhibit good capacity for myogenic regeneration. However, for better therapeutic outcomes, it is necessary to isolate human MuSCs from a suitable tissue source with high myogenic differentiation. In this context, we isolated CD56+CD82+ cells from the extra eyelid tissue of young and aged patients, and tested in vitro myogenic differentiation potential. In the current study, myogenic cells derived from extra eyelid tissue were characterized and compared with immortalized human myogenic cells. We found that myogenic cells derived from extra eyelid tissue proliferated and differentiated myofibers in vitro, and restored DYSTROPHIN or PAX7 expression after transplantation with these cells in mice with Duchenne muscular dystrophy. Thus, human myogenic cells derived from extra eyelid tissue including the orbicularis oculi might be good candidates for stem cell-based therapies for treating muscular diseases.
APA, Harvard, Vancouver, ISO, and other styles
25

Bechshøft, Cecilie J. L., Simon M. Jensen, Peter Schjerling, Jesper L. Andersen, Rene B. Svensson, Christian S. Eriksen, Nonhlanhla S. Mkumbuzi, Michael Kjaer, and Abigail L. Mackey. "Age and prior exercise in vivo determine the subsequent in vitro molecular profile of myoblasts and nonmyogenic cells derived from human skeletal muscle." American Journal of Physiology-Cell Physiology 316, no. 6 (June 1, 2019): C898—C912. http://dx.doi.org/10.1152/ajpcell.00049.2019.

Full text
Abstract:
The decline in skeletal muscle regenerative capacity with age is partly attributed to muscle stem cell (satellite cell) dysfunction. Recent evidence has pointed to a strong interaction between myoblasts and fibroblasts, but the influence of age on this interaction is unknown. Additionally, while the native tissue environment is known to determine the properties of myogenic cells in vitro, how the aging process alters this cell memory has not been established at the molecular level. We recruited 12 young and 12 elderly women, who performed a single bout of heavy resistance exercise with the knee extensor muscles of one leg. Five days later, muscle biopsies were collected from both legs, and myogenic cells and nonmyogenic cells were isolated for in vitro experiments with mixed or separated cells and analyzed by immunostaining and RT-PCR. A lower myogenic fusion index was detected in the cells from the old versus young women, in association with differences in gene expression levels of key myogenic regulatory factors and senescence, which were further altered by performing exercise before tissue sampling. Coculture with nonmyogenic cells from the elderly led to a higher myogenic differentiation index compared with nonmyogenic cells from the young. These findings show that the in vitro phenotype and molecular profile of human skeletal muscle myoblasts and fibroblasts is determined by the age and exercise state of the original in vivo environment and help explain how exercise can enhance muscle stem cell function in old age.
APA, Harvard, Vancouver, ISO, and other styles
26

Vieira, Natássia M., Vanessa Brandalise, Eder Zucconi, Tatiana Jazedje, Mariane Secco, Viviane A. Nunes, Bryan E. Strauss, Mariz Vainzof, and Mayana Zatz. "Human multipotent adipose-derived stem cells restore dystrophin expression of Duchenne skeletal-muscle cells in vitro." Biology of the Cell 100, no. 4 (April 2008): 231–41. http://dx.doi.org/10.1042/bc20070102.

Full text
APA, Harvard, Vancouver, ISO, and other styles
27

Dell'Agnola, Chiara, Zejing Wang, Rainer Storb, Stephen J. Tapscott, Christian S. Kuhr, Stephen D. Hauschka, Richard S. Lee, et al. "Hematopoietic stem cell transplantation does not restore dystrophin expression in Duchenne muscular dystrophy dogs." Blood 104, no. 13 (December 15, 2004): 4311–18. http://dx.doi.org/10.1182/blood-2004-06-2247.

Full text
Abstract:
Abstract Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene on the X-chromosome that result in skeletal and cardiac muscle damage and premature death. Studies in mice, including the mdx mouse model of DMD, have demonstrated that circulating bone marrow–derived cells can participate in skeletal muscle regeneration, but the potential clinical utility of treating human DMD by allogeneic marrow transplantation from a healthy donor remains unknown. To assess whether allogeneic hematopoietic cell transplantation (HCT) provides clinically relevant levels of donor muscle cell contribution in dogs with canine X-linked muscular dystrophy (c-xmd), 7 xmd dogs were given hematopoietic cell (HC) transplants from nonaffected littermates. Compared with the pretransplantation baseline, the number of dystrophin-positive fibers and the amount of wild-type dystrophin RNA did not increase after HCT, with observation periods ranging from 28 to 417 days. Similar results were obtained when the recipient dogs were given granulocyte colony-stimulating factor (G-CSF) after their initial transplantation to mobilize the cells. Despite successful allogeneic HCT and a permissive environment for donor muscle engraftment, there was no detectable contribution of bone marrow–derived cells to either skeletal muscle or muscle precursor cells assayed by clonal analyses at a level of sensitivity that should detect as little as 0.1% donor contribution.
APA, Harvard, Vancouver, ISO, and other styles
28

Mikšiūnas, Rokas, Siegfried Labeit, and Daiva Bironaitė. "The Effect of Heat Shock on Myogenic Differentiation of Human Skeletal-Muscle-Derived Mesenchymal Stem/Stromal Cells." Cells 11, no. 20 (October 13, 2022): 3209. http://dx.doi.org/10.3390/cells11203209.

Full text
Abstract:
Muscle injuries, degenerative diseases and other lesions negatively affect functioning of human skeletomuscular system and thus quality of life. Therefore, the investigation of molecular mechanisms, stimulating myogenic differentiation of primary skeletal-muscle-derived mesenchymal stem/stromal cells (SM-MSCs), is actual and needed. The aim of the present study was to investigate the myogenic differentiation of CD56 (neural cell adhesion molecule, NCAM)-positive and -negative SM-MSCs and their response to the non-cytotoxic heat stimulus. The SM-MSCs were isolated from the post operation muscle tissue, sorted by flow cytometer according to the CD56 biomarker and morphology, surface profile, proliferation and myogenic differentiation has been investigated. Data show that CD56(+) cells were smaller in size, better proliferated and had significantly higher levels of CD146 (MCAM) and CD318 (CDCP1) compared with the CD56(−) cells. At control level, CD56(+) cells significantly more expressed myogenic differentiation markers MYOD1 and myogenin (MYOG) and better differentiated to the myogenic direction. The non-cytotoxic heat stimulus significantly stronger stimulated expression of myogenic markers in CD56(+) than in CD56(−) cells that correlated with the multinucleated cell formation. Data show that regenerative properties of CD56(+) SM-MSCs can be stimulated by an extracellular stimulus and be used as a promising skeletal muscle regenerating tool in vivo.
APA, Harvard, Vancouver, ISO, and other styles
29

Barbon, Silvia, Elena Stocco, Martina Contran, Federico Facchin, Rafael Boscolo-Berto, Silvia Todros, Deborah Sandrin, et al. "Preclinical Development of Bioengineered Allografts Derived from Decellularized Human Diaphragm." Biomedicines 10, no. 4 (March 22, 2022): 739. http://dx.doi.org/10.3390/biomedicines10040739.

Full text
Abstract:
Volumetric muscle loss (VML) is the traumatic/surgical loss of skeletal muscle, causing aesthetic damage and functional impairment. Suboptimal current surgical treatments are driving research towards the development of optimised regenerative therapies. The grafting of bioengineered scaffolds derived from decellularized skeletal muscle may be a valid option to promote structural and functional healing. In this work, a cellular human diaphragm was considered as a scaffold material for VML treatment. Decellularization occurred through four detergent-enzymatic protocols involving (1) sodium dodecyl sulfate (SDS), (2) SDS + TergitolTM, (3) sodium deoxycholate, and (4) TergitolTM. After decellularization, cells, DNA (≤50 ng/mg of tissue), and muscle fibres were efficiently removed, with the preservation of collagen/elastin and 60%–70% of the glycosaminoglycan component. The detergent-enzymatic treatments did not affect the expression of specific extracellular matrix markers (Collagen I and IV, Laminin), while causing the loss of HLA-DR expression to produce non-immunogenic grafts. Adipose-derived stem cells grown by indirect co-culture with decellularized samples maintained 80%–90% viability, demonstrating the biosafety of the scaffolds. Overall, the tested protocols were quite equivalent, with the patches treated by SDS + TergitolTM showing better collagen preservation. After subcutaneous implant in Balb/c mice, these acellular diaphragmatic grafts did not elicit a severe immune reaction, integrating with the host tissue.
APA, Harvard, Vancouver, ISO, and other styles
30

Farrugia, Georgiana, and Rena Balzan. "Stem Cell Repair for Cardiac Muscle Regeneration: A Review of the Literature." International Journal of Medical Students 4, no. 1 (April 30, 2016): 19–25. http://dx.doi.org/10.5195/ijms.2016.145.

Full text
Abstract:
The notion that the human adult heart is a quiescent organ incapable of self-regeneration has been successfully challenged. It is now evident that the heart possesses a significant ability for repair and regeneration. Stem cells of endogenous cardiac origin are currently considered to possess the greatest ability to differentiate into cardiomyocytes. The major types of cardiac stem cells that show a promising potential to replace damaged cardiomyocytes include C-KIT positive (C-KIT+) cardiac progenitor cells, cardiosphere-derived progenitor cells, islet-1 (Isl1+) cardiac progenitor cells, side-population cardiac progenitor cells, epicardium-derived progenitor cells and stem cell antigen-1 (SCA1+) cardiac progenitor cells. Moreover, stem cells of extra-cardiac origin are also thought to restore contractility and vascularization of the myocardium. These include skeletal myoblasts, bone marrow mononuclear cells, mesenchymal stem cells, endothelial progenitor cells as well as embryonic stem cells. The need for further investigation on cardiac stem cell therapeutic strategies still remains.
APA, Harvard, Vancouver, ISO, and other styles
31

Frank, Natasha Y., Alvin T. Kho, Tobias Schatton, George F. Murphy, Michael J. Molloy, Qian Zhan, Marco F. Ramoni, Markus H. Frank, Isaac S. Kohane, and Emanuela Gussoni. "Regulation of myogenic progenitor proliferation in human fetal skeletal muscle by BMP4 and its antagonist Gremlin." Journal of Cell Biology 175, no. 1 (October 2, 2006): 99–110. http://dx.doi.org/10.1083/jcb.200511036.

Full text
Abstract:
Skeletal muscle side population (SP) cells are thought to be “stem”-like cells. Despite reports confirming the ability of muscle SP cells to give rise to differentiated progeny in vitro and in vivo, the molecular mechanisms defining their phenotype remain unclear. In this study, gene expression analyses of human fetal skeletal muscle demonstrate that bone morphogenetic protein 4 (BMP4) is highly expressed in SP cells but not in main population (MP) mononuclear muscle-derived cells. Functional studies revealed that BMP4 specifically induces proliferation of BMP receptor 1a–positive MP cells but has no effect on SP cells, which are BMPR1a-negative. In contrast, the BMP4 antagonist Gremlin, specifically up-regulated in MP cells, counteracts the stimulatory effects of BMP4 and inhibits proliferation of BMPR1a-positive muscle cells. In vivo, BMP4-positive cells can be found in the proximity of BMPR1a-positive cells in the interstitial spaces between myofibers. Gremlin is expressed by mature myofibers and interstitial cells, which are separate from BMP4-expressing cells. Together, these studies propose that BMP4 and Gremlin, which are highly expressed by human fetal skeletal muscle SP and MP cells, respectively, are regulators of myogenic progenitor proliferation.
APA, Harvard, Vancouver, ISO, and other styles
32

Aboalola, Doaa, and Victor K. M. Han. "Insulin-Like Growth Factor Binding Protein-6 Alters Skeletal Muscle Differentiation of Human Mesenchymal Stem Cells." Stem Cells International 2017 (2017): 1–17. http://dx.doi.org/10.1155/2017/2348485.

Full text
Abstract:
Insulin-like growth factor binding protein-6 (IGFBP-6), the main regulator of insulin-like growth factor-2 (IGF-2), is a component of the stem cell niche in developing muscle cells. However, its role in muscle development has not been clearly defined. In this study, we investigated the role of IGFBP-6 in muscle commitment and differentiation of human mesenchymal stem cells derived from the placenta. We showed that placental mesenchymal stem cells (PMSCs) have the ability to differentiate into muscle cells when exposed to a specific culture medium by expressing muscle markers Pax3/7, MyoD, myogenin, and myosin heavy chain in a stage-dependent manner with the ultimate formation of multinucleated fibers and losing pluripotency-associated markers, OCT4 and SOX2. The addition of IGFBP-6 significantly increased pluripotency-associated markers as well as muscle differentiation markers at earlier time points, but the latter decreased with time. On the other hand, silencing IGFBP-6 decreased both pluripotent and differentiation markers at early time points. The levels of these markers increased as IGFBP-6 levels were restored. These findings indicate that IGFBP-6 influences MSC pluripotency and myogenic differentiation, with more prominent effects observed at the beginning of the differentiation process before muscle commitment.
APA, Harvard, Vancouver, ISO, and other styles
33

Bruge, Celine, Marine Geoffroy, Manon Benabides, Emilie Pellier, Evelyne Gicquel, Jamila Dhiab, Lucile Hoch, Isabelle Richard, and Xavier Nissan. "Skeletal Muscle Cells Derived from Induced Pluripotent Stem Cells: A Platform for Limb Girdle Muscular Dystrophies." Biomedicines 10, no. 6 (June 16, 2022): 1428. http://dx.doi.org/10.3390/biomedicines10061428.

Full text
Abstract:
Limb girdle muscular dystrophies (LGMD), caused by mutations in 29 different genes, are the fourth most prevalent group of genetic muscle diseases. Although the link between LGMD and its genetic origins has been determined, LGMD still represent an unmet medical need. Here, we describe a platform for modeling LGMD based on the use of human induced pluripotent stem cells (hiPSC). Thanks to the self-renewing and pluripotency properties of hiPSC, this platform provides a renewable and an alternative source of skeletal muscle cells (skMC) to primary, immortalized, or overexpressing cells. We report that skMC derived from hiPSC express the majority of the genes and proteins that cause LGMD. As a proof of concept, we demonstrate the importance of this cellular model for studying LGMDR9 by evaluating disease-specific phenotypes in skMC derived from hiPSC obtained from four patients.
APA, Harvard, Vancouver, ISO, and other styles
34

Morris, Elizabeth, Alex Scibetta, Aiping Lu, Xueqin Gao, and Johnny Huard. "449. IGF-1 Treatment Enhances the Myogenic Potential of Human Skeletal Muscle-Derived Stem Cells." Molecular Therapy 24 (May 2016): S178—S179. http://dx.doi.org/10.1016/s1525-0016(16)33258-0.

Full text
APA, Harvard, Vancouver, ISO, and other styles
35

Bansal, Varun, Debojyoti De, Jieun An, Tong Mook Kang, Hyeon-Ju Jeong, Jong-Sun Kang, and Kyeong Kyu Kim. "Chemical induced conversion of mouse fibroblasts and human adipose-derived stem cells into skeletal muscle-like cells." Biomaterials 193 (February 2019): 30–46. http://dx.doi.org/10.1016/j.biomaterials.2018.11.037.

Full text
APA, Harvard, Vancouver, ISO, and other styles
36

Park, S., Y. Choi, N. Jung, J. Kim, B. Choi, and S. Jung. "Therapeutic Potential of Human Tonsil-Derived Stem Cell for Skeletal Muscle Regeneration." Cytotherapy 18, no. 6 (June 2016): S23—S24. http://dx.doi.org/10.1016/j.jcyt.2016.03.067.

Full text
APA, Harvard, Vancouver, ISO, and other styles
37

Al Tanoury, Ziad, John F. Zimmerman, Jyoti Rao, Daniel Sieiro, Harold M. McNamara, Thomas Cherrier, Alejandra Rodríguez-delaRosa, et al. "Prednisolone rescues Duchenne muscular dystrophy phenotypes in human pluripotent stem cell–derived skeletal muscle in vitro." Proceedings of the National Academy of Sciences 118, no. 28 (July 6, 2021): e2022960118. http://dx.doi.org/10.1073/pnas.2022960118.

Full text
Abstract:
Duchenne muscular dystrophy (DMD) is a devastating genetic disease leading to degeneration of skeletal muscles and premature death. How dystrophin absence leads to muscle wasting remains unclear. Here, we describe an optimized protocol to differentiate human induced pluripotent stem cells (iPSC) to a late myogenic stage. This allows us to recapitulate classical DMD phenotypes (mislocalization of proteins of the dystrophin-associated glycoprotein complex, increased fusion, myofiber branching, force contraction defects, and calcium hyperactivation) in isogenic DMD-mutant iPSC lines in vitro. Treatment of the myogenic cultures with prednisolone (the standard of care for DMD) can dramatically rescue force contraction, fusion, and branching defects in DMD iPSC lines. This argues that prednisolone acts directly on myofibers, challenging the largely prevalent view that its beneficial effects are caused by antiinflammatory properties. Our work introduces a human in vitro model to study the onset of DMD pathology and test novel therapeutic approaches.
APA, Harvard, Vancouver, ISO, and other styles
38

Abati, Elena, Emanuele Sclarandi, Giacomo Pietro Comi, Valeria Parente, and Stefania Corti. "Perspectives on hiPSC-Derived Muscle Cells as Drug Discovery Models for Muscular Dystrophies." International Journal of Molecular Sciences 22, no. 17 (September 6, 2021): 9630. http://dx.doi.org/10.3390/ijms22179630.

Full text
Abstract:
Muscular dystrophies are a heterogeneous group of inherited diseases characterized by the progressive degeneration and weakness of skeletal muscles, leading to disability and, often, premature death. To date, no effective therapies are available to halt or reverse the pathogenic process, and meaningful treatments are urgently needed. From this perspective, it is particularly important to establish reliable in vitro models of human muscle that allow the recapitulation of disease features as well as the screening of genetic and pharmacological therapies. We herein review and discuss advances in the development of in vitro muscle models obtained from human induced pluripotent stem cells, which appear to be capable of reproducing the lack of myofiber proteins as well as other specific pathological hallmarks, such as inflammation, fibrosis, and reduced muscle regenerative potential. In addition, these platforms have been used to assess genetic correction strategies such as gene silencing, gene transfer and genome editing with clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9), as well as to evaluate novel small molecules aimed at ameliorating muscle degeneration. Furthermore, we discuss the challenges related to in vitro drug testing and provide a critical view of potential therapeutic developments to foster the future clinical translation of preclinical muscular dystrophy studies.
APA, Harvard, Vancouver, ISO, and other styles
39

Mollard, R., BJ Conley, and AO Trounson. "Human embryonic stem cells: prototypical pluripotential progenitors." Reproductive Medicine Review 10, no. 3 (October 2002): 187–99. http://dx.doi.org/10.1017/s0962279902000340.

Full text
Abstract:
Embryonic stem (ES) cells are a primitive cell type derived from the inner cell mass (ICM) of the developing embryo. When cultured for extended periods, ES cells maintain a high telomerase activity, normal karyotype and the pluripotential developmental capacity of their ICM derivatives. Such capacity is best demonstrated by mouse ES cells which can contribute to all tissues of the developing embryo following either their injection into host blastocysts or tetraploid embryo complimentation (for a review see Robertson). For both practical and ethical reasons it is not possible to inject human ES cells into blastocysts for the development of a term fetus. However, when injected beneath the testicular capsule of severe combined immunodeficient (SCID) mice, human ES cells form teratomas comprising tissue representatives of all three embryonic germ layers (ectoderm, mesoderm and endoderm) thus attesting to their pluripotency. Based upon morphological criteria, neuronal, cardiac, bone, squamous epithelium, skeletal muscle, gut and respiratory epithelia are readily identifiable within the human ES-cell-derived teratomas. With the demonstrated capability to isolate and maintain pluripotent human ES cells in vitro, their ability to give rise to tissue representatives of all three embryonic germ layers and the technical advances made possible by research on mouse ES cells, a rapid increase in human ES cell research aimed at drug discovery and human cell and gene therapies has occurred. Indeed in the mouse, dissociated embryoid bodies (EBs) have already been demonstrated capable of repopulating the haematopoietic system of recipient animals (for a review see Keller) and mouse ES cells are currently being used in attempts to repair mouse neural degenerative lesions.
APA, Harvard, Vancouver, ISO, and other styles
40

Nakajima, Nobuyuki, Tetsuro Tamaki, Maki Hirata, Shuichi Soeda, Masahiro Nitta, Akio Hoshi, and Toshiro Terachi. "Purified Human Skeletal Muscle-Derived Stem Cells Enhance the Repair and Regeneration in the Damaged Urethra." Transplantation 101, no. 10 (October 2017): 2312–20. http://dx.doi.org/10.1097/tp.0000000000001613.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

Vieira, N., M. Mitne, E. Zucconi, T. Jazedje, V. Nunes, B. Strauss, M. Vainzof, and M. Zatz. "G.P.16.14 Human multipotent adipose derived stem cells restore dystrophin expression of Duchenne skeletal muscle cells in vitro." Neuromuscular Disorders 17, no. 9-10 (October 2007): 878. http://dx.doi.org/10.1016/j.nmd.2007.06.390.

Full text
APA, Harvard, Vancouver, ISO, and other styles
42

Skuk, Daniel. "Cell Transplantation and “Stem Cell Therapy” in the Treatment of Myopathies: Many Promises in Mice, Few Realities in Humans." ISRN Transplantation 2013 (October 21, 2013): 1–25. http://dx.doi.org/10.5402/2013/582689.

Full text
Abstract:
Myopathies produce deficits in skeletal muscle function and, in some cases, progressive and irreversible loss of skeletal muscles. The transplantation of myogenic cells, that is, cells able to differentiate into myofibers, is an experimental strategy for the potential treatment of some of these diseases. The objectives pursued by the transplantation of these cells are essentially three: (a) the fusion with the patient’s myofibers to obtain the expression of therapeutic proteins into them, (b) the neoformation of new functional myofibers in skeletal muscles that were too degenerated by the progressive degeneration, and (c) the formation of a new pool of healthy donor-derived satellite cells. Although the repertoire of myogenic cells appears to have expanded in recent years, myoblasts are the only cells that have been demonstrated to engraft in humans. The present work aims to make a comprehensive review of the subject, from its beginnings to recent advances, including the preclinical experience in different animal models and recent clinical findings.
APA, Harvard, Vancouver, ISO, and other styles
43

Su, Wen-Hong, Ching-Jen Wang, Hung-Chun Fu, Chien-Ming Sheng, Ching-Chin Tsai, Jai-Hong Cheng, and Pei-Chin Chuang. "Human Umbilical Cord Mesenchymal Stem Cells Extricate Bupivacaine-Impaired Skeletal Muscle Function via Mitigating Neutrophil-Mediated Acute Inflammation and Protecting against Fibrosis." International Journal of Molecular Sciences 20, no. 17 (September 3, 2019): 4312. http://dx.doi.org/10.3390/ijms20174312.

Full text
Abstract:
Skeletal muscle injury presents a challenging traumatological dilemma, and current therapeutic options remain mediocre. This study was designed to delineate if engraftment of mesenchymal stem cells derived from umbilical cord Wharton’s jelly (uMSCs) could aid in skeletal muscle healing and persuasive molecular mechanisms. We established a skeletal muscle injury model by injection of myotoxin bupivacaine (BPVC) into quadriceps muscles of C57BL/6 mice. Post BPVC injection, neutrophils, the first host defensive line, rapidly invaded injured muscle and induced acute inflammation. Engrafted uMSCs effectively abolished neutrophil infiltration and activation, and diminished neutrophil chemotaxis, including Complement component 5a (C5a), Keratinocyte chemoattractant (KC), Macrophage inflammatory protein (MIP)-2, LPS-induced CXC chemokine (LIX), Fractalkine, Leukotriene B4 (LTB4), and Interferon-γ, as determined using a Quantibody Mouse Cytokine Array assay. Subsequently, uMSCs noticeably prevented BPVC-accelerated collagen deposition and fibrosis, measured by Masson’s trichrome staining. Remarkably, uMSCs attenuated BPVC-induced Transforming growth factor (TGF)-β1 expression, a master regulator of fibrosis. Engrafted uMSCs attenuated TGF-β1 transmitting through interrupting the canonical Sma- And Mad-Related Protein (Smad)2/3 dependent pathway and noncanonical Smad-independent Transforming growth factor beta-activated kinase (TAK)-1/p38 mitogen-activated protein kinases signaling. The uMSCs abrogated TGF-β1-induced fibrosis by reducing extracellular matrix components including fibronectin-1, collagen (COL) 1A1, and COL10A1. Most importantly, uMSCs modestly extricated BPVC-impaired gait functions, determined using CatWalk™ XT gait analysis. This work provides several innovative insights into and molecular bases for employing uMSCs to execute therapeutic potential through the elimination of neutrophil-mediated acute inflammation toward protecting against fibrosis, thereby rescuing functional impairments post injury.
APA, Harvard, Vancouver, ISO, and other styles
44

Massenet, Jimmy, Cyril Gitiaux, Mélanie Magnan, Sylvain Cuvellier, Arnaud Hubas, Patrick Nusbaum, F. Jeffrey Dilworth, Isabelle Desguerre, and Bénédicte Chazaud. "Derivation and Characterization of Immortalized Human Muscle Satellite Cell Clones from Muscular Dystrophy Patients and Healthy Individuals." Cells 9, no. 8 (July 26, 2020): 1780. http://dx.doi.org/10.3390/cells9081780.

Full text
Abstract:
In Duchenne muscular dystrophy (DMD) patients, absence of dystrophin causes muscle wasting by impacting both the myofiber integrity and the properties of muscle stem cells (MuSCs). Investigation of DMD encompasses the use of MuSCs issued from human skeletal muscle. However, DMD-derived MuSC usage is restricted by the limited number of divisions that human MuSCs can undertake in vitro before losing their myogenic characteristics and by the scarcity of human material available from DMD muscle. To overcome these limitations, immortalization of MuSCs appears as a strategy. Here, we used CDK4/hTERT expression in primary MuSCs and we derived MuSC clones from a series of clinically and genetically characterized patients, including eight DMD patients with various mutations, four congenital muscular dystrophies and three age-matched control muscles. Immortalized cultures were sorted into single cells and expanded as clones into homogeneous populations. Myogenic characteristics and differentiation potential were tested for each clone. Finally, we screened various promoters to identify the preferred gene regulatory unit that should be used to ensure stable expression in the human MuSC clones. The 38 clonal immortalized myogenic cell clones provide a large collection of controls and DMD clones with various genetic defects and are available to the academic community.
APA, Harvard, Vancouver, ISO, and other styles
45

Yang, Xiaoping, Jingjing Gu, Tingting Yang, Rui Zhou, and Bo Zheng. "Biological Characteristics of Human Skeletal Muscle-Derived Pericytes/Perivascular Cells and Their Supporting Effects on Hematopoietic Stem Cell in Vitro." Blood 134, Supplement_1 (November 13, 2019): 5006. http://dx.doi.org/10.1182/blood-2019-130416.

Full text
Abstract:
Mesenchymal stem/stromal cells (MSCs) as precursor cells of bone marrow stromal cells, in addition to the formation of the bone marrow microenvironment, secrete a variety of blood-related factors, promote hematopoietic recovery and reconstruction. However, mesenchymal stem/stromal cells are extremely rare within human bone marrow, which limit their experimental and clinical applications. Our previous study demonstrated that MSCs were derived from pericytes/perivascular cells (PCs), and PCs were the precursor of MSCs. PCs are widely distributed, especially in skeletal muscle tissue, which are rich in content, easy to be harvested with minimal damage, and rapid cell proliferation. However, very few researches have been done on whether human skeletal muscle-derived pericytes/perivascular cells (hMD-PCs) are involved in HSPCs expansion/proliferation, differentiation and possible hematopoietic regulation mechanisms. Based on the above research background and research ideas, in current study, we intend to isolate, purify and characterize the CD146+ hMD-PCs and further investigate their supporting effect on human umbilical cord blood CD34+ cells in vitro. Methods 1. hMD-PCs with phenotype CD146+CD56-CD34-CD144-CD45- were sorted from human skeletal muscle by enzymatic digestion and multiparameter fluorescence-activated cell sorting (MP-FACS), and their biological characteristic were conducted by detecting the surface marker of MSCs and inducing multilineage differentiation. 2. Human UCB CD34+cells were sorted by immunomagnetic beads, and then established the in vitro expansion culture system of UCB CD34+ cells co-culture with CD146+ hMD-PCs (experimental group), human bone marrow MSCs for feeder layer (positive control) and UCB CD34+ cells alone culture system (blank control). After 1 week, 2 weeks and 4 weeks of co-culture, the number of cells, the colony formation ability and immunophenotype were analysed. Result 1.CD146+ hMD-PCs were sorted by MP-FACS and the purity was (91.5±1.85)% (n=5); CD146+ hMD-PCs expressed mesenchymal surface markers CD73, CD90, CD105, CD44, and did not express hematopoietic cell and endothelial cell marker CD45, CD34, CD31. After induced culture, CD146+ hMD-PCs can differentiate into osteoblasts, chondrogenesis, adipocytes and myoblasts; 2.UCB CD34+ cells were sorted by magnetic beads which number was (9.18±3.50)×105, and then co-cultured with CD146+ hMD-PCs and human BM-MSCs at a density of 5×104/well, respectively. After 1, 2 and 4 weeks of co-culture, there were no statistically significant differences in cell number, colony formation ability and immunophenotype (CD45+cells, CD34+CD33-cells, CD14+cells, CD10+/CD19+cells) between CD146+ hMD-PCs for feeder layer culture system and BM-MSCs for feeder layer culture system (P>0.05, n=6). However, after 1 week, 2 weeks of co-culture, the both cells number of CD146+ hMD-PCs and human BM-MSCs culture system were significantly different from the blank control group, (P<0.01) and (P<0.001) respectively. The colony culture could not be performed due to low cell number of the blank control group. Conclusion CD146+ hMD-PCs, like human BM-MSCs, have hematopoietic support capacity in vitro; therefore CD146+ hMD-PCs can be used as another source of stromal cells for the expansion of hematopoietic stem cells. Disclosures No relevant conflicts of interest to declare.
APA, Harvard, Vancouver, ISO, and other styles
46

Langenau, David M., Yun Wei, Qian Qin, and Luca Pinello. "Abstract IA014: Stem cell and developmental hierarchies in rhabdomyosarcoma." Clinical Cancer Research 28, no. 18_Supplement (September 15, 2022): IA014. http://dx.doi.org/10.1158/1557-3265.sarcomas22-ia014.

Full text
Abstract:
Abstract Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma of childhood and is comprised of at least two major molecular subtypes. Despite sharing features with skeletal muscle, the conservation of underlying cellular hierarchy with human muscle development and the identification of molecularly-defined tumor-propagating cells have not been reported. Using single-cell RNA sequencing of patient-derived RMS, DNA-barcode cell fate mapping, antibody enrichment and functional stem cell assays, and mouse xenograft modeling, we have uncovered tumor cell hierarchies in Fusion-negative (FN-) RMS that are shared with normal human muscle development. We also identified common developmental stages at which tumor cells become arrested. FN-RMS resemble early muscle found in embryonic and larval development, while fusion-positive (FP-) RMS express a highly specific developmental gene program found in muscle cells transiting from embryonic to fetal development at 7-7.75 weeks of age. FP-RMS also have neural-pathway enriched cell states, suggesting less-rigid adherence to muscle development hierarchies in this disease. Finally, we identify a new molecularly-defined tumor-propagating cell in FN-RMS that shares remarkable similarity to the newly described bi-potent, muscle mesenchyme stem/progenitor cell that makes both muscle and osteogenic cells. Citation Format: David M. Langenau, Yun Wei, Qian Qin, Luca Pinello. Stem cell and developmental hierarchies in rhabdomyosarcoma [abstract]. In: Proceedings of the AACR Special Conference: Sarcomas; 2022 May 9-12; Montreal, QC, Canada. Philadelphia (PA): AACR; Clin Cancer Res 2022;28(18_Suppl):Abstract nr IA014.
APA, Harvard, Vancouver, ISO, and other styles
47

Nowaczyk, Magdalena, Agnieszka Malcher, Agnieszka Zimna, Wojciech Łabędź, Łukasz Kubaszewski, Katarzyna Fiedorowicz, Kamil Wierzbiński, Natalia Rozwadowska, and Maciej Kurpisz. "Transient and Stable Overexpression of Extracellular Superoxide Dismutase is Positively Associated with the Myogenic Function of Human Skeletal Muscle-Derived Stem/Progenitor Cells." Antioxidants 9, no. 9 (September 2, 2020): 817. http://dx.doi.org/10.3390/antiox9090817.

Full text
Abstract:
In the present study, the genetic modification of human skeletal muscle-derived stem/progenitor cells (SkMDS/PCs) was investigated to identify the optimal protocol for myogenic cell preparation for use in post-infarction heart therapy. We used two types of modifications: GFP-transfection (using electroporation) and SOD3 transduction (using a lentiviral vector). SkMDS/PCs were cultured under different in vitro conditions, including standard (21% oxygen) and hypoxic (3% oxygen), the latter of which corresponded to the prevailing conditions in the post-infarction heart. Transfection/transduction efficacy, skeletal myogenic cell marker expression (CD56), cellular senescence, and apoptosis, as well as the expression of antioxidant (SOD1, SOD2, and SOD3), anti-aging (SIRT1 and FOXO), anti-apoptotic (BCL2), and myogenic (MyoD and MyoG) genes, were evaluated. The percentage of GFP-positive SkMDS/PCs was determined as an indicator of the efficacy of transfection, which reached 55%, while transduction showed better efficiency, reaching approximately 85% as estimated by fluorescence microscopy. The CD56-positive SkMDS/PCs were present in approximately 77% of the tested cells after transient transfection and approximately 96% after transduction. Under standard in vitro culture conditions, the ability of the differentiated, transfected SkMDS/PCs to form myotubes was greater than that of the wild type (WT) cell population (p < 0.001), while the cells transduced with the SOD3 gene exhibited an increase in cell fusion under both standard (p < 0.05) and hypoxic conditions (p < 0.001). In transduced SkMDS/PCs, we observed a positive influence of SOD3 overexpression on cell ageing and apoptosis. We observed an increase in the percentage of young cells under standard (p < 0.05) and hypoxic (p < 0.001) in vitro culture conditions, with a notable decrease in the percentage of senescent and advanced senescent cells in the SOD3-overexpressing cell population detected compared to that observed for the untransduced muscle-derived cells. A lower percentage of apoptotic cells was observed for transduced SkMDS/PCs than that for WT cells under hypoxic in vitro culture conditions. In transiently transfected SkMDS/PCs, we observed significantly higher gene expression levels of SOD2 (almost 40-fold) (p < 0.001) and FOXO (p < 0.05) (approximately 3-fold) under both normoxic and hypoxic culture conditions and of BCL2 under hypoxia compared to those observed in untreated cells (WT). In addition, myogenic genes showed a significant increase in MyoD (almost 18-fold) expression under standard culture conditions (p < 0.0001) and decreased MyoG expression (approximately 2-fold) after transfection (p < 0.05) compared with that detected in the WT skeletal muscle-derived cell control. Taken together, these results demonstrate that SOD3-tranduced skeletal muscle-derived cells may have potential for use in the regenerative treatment of the post-infarction heart.
APA, Harvard, Vancouver, ISO, and other styles
48

Hörner, Sarah Janice, Nathalie Couturier, Roman Bruch, Philipp Koch, Mathias Hafner, and Rüdiger Rudolf. "hiPSC-Derived Schwann Cells Influence Myogenic Differentiation in Neuromuscular Cocultures." Cells 10, no. 12 (November 24, 2021): 3292. http://dx.doi.org/10.3390/cells10123292.

Full text
Abstract:
Motoneurons, skeletal muscle fibers, and Schwann cells form synapses, termed neuromuscular junctions (NMJs). These control voluntary body movement and are affected in numerous neuromuscular diseases. Therefore, a variety of NMJ in vitro models have been explored to enable mechanistic and pharmacological studies. So far, selective integration of Schwann cells in these models has been hampered, due to technical limitations. Here we present robust protocols for derivation of Schwann cells from human induced pluripotent stem cells (hiPSC) and their coculture with hiPSC-derived motoneurons and C2C12 muscle cells. Upon differentiation with tuned BMP signaling, Schwann cells expressed marker proteins, S100b, Gap43, vimentin, and myelin protein zero. Furthermore, they displayed typical spindle-shaped morphologies with long processes, which often aligned with motoneuron axons. Inclusion of Schwann cells in coculture experiments with hiPSC-derived motoneurons and C2C12 myoblasts enhanced myotube growth and affected size and number of acetylcholine receptor plaques on myotubes. Altogether, these data argue for the availability of a consistent differentiation protocol for Schwann cells and their amenability for functional integration into neuromuscular in vitro models, fostering future studies of neuromuscular mechanisms and disease.
APA, Harvard, Vancouver, ISO, and other styles
49

Ni, Jianshu, Hongchao Li, Yiwen Zhou, Baojun Gu, Yuemin Xu, Qiang Fu, Xufeng Peng, et al. "Therapeutic Potential of Human Adipose-Derived Stem Cell Exosomes in Stress Urinary Incontinence – An in Vitro and in Vivo Study." Cellular Physiology and Biochemistry 48, no. 4 (2018): 1710–22. http://dx.doi.org/10.1159/000492298.

Full text
Abstract:
Background/Aims: To evaluate whether local injection of exosomes derived from human adipose-derived stem cells (hADSCs) facilitates recovery of stress urinary incontinence (SUI) in a rat model. Methods: For the in vitro study, a Cell Counting Kit-8 (CCK-8) array and proteomic analysis were performed. For the in vivo study, female rats were divided into four groups: sham, SUI, adipose-derived stem cell (ADSC), and exosomes (n = 12 each). The SUI model was generated by pudendal nerve transection and vaginal dilation. Vehicle, hADSCs, or exosomes were injected into the peripheral urethra. After 2, 4, and 8 weeks, the rats underwent cystometrography and leak point pressure (LPP) testing, and tissues were harvested for histochemical analyses. Results: The CCK-8 experiment demonstrated that ADSC-derived exosomes could enhance the growth of skeletal muscle and Schwann cell lines in a dose-dependent manner. Proteomic analysis revealed that ADSC-derived exosomes contained various proteins of different signaling pathways. Some of these proteins are associated with the PI3K-Akt, Jak-STAT, and Wnt pathways, which are related to skeletal muscle and nerve regeneration and proliferation. In vivo experiments illustrated that rats of the exosome group had higher bladder capacity and LPP, and had more striated muscle fibers and peripheral nerve fibers in the urethra than rats of the SUI group. Both urethral function and histology of rats in the exosome group were slightly better than those in the ADSC group. Conclusions: Local injection of hADSC-derived exosomes improved functional and histological recovery after SUI.
APA, Harvard, Vancouver, ISO, and other styles
50

Ikenaka, Akihiro, Yohko Kitagawa, Michiko Yoshida, Chuang-Yu Lin, Akira Niwa, Tatsutoshi Nakahata, and Megumu K. Saito. "SMN promotes mitochondrial metabolic maturation during myogenesis by regulating the MYOD-miRNA axis." Life Science Alliance 6, no. 3 (January 5, 2023): e202201457. http://dx.doi.org/10.26508/lsa.202201457.

Full text
Abstract:
Spinal muscular atrophy (SMA) is a congenital neuromuscular disease caused by the mutation or deletion of thesurvival motor neuron 1 (SMN1)gene. Although the primary cause of progressive muscle atrophy in SMA has classically been considered the degeneration of motor neurons, recent studies have indicated a skeletal muscle–specific pathological phenotype such as impaired mitochondrial function and enhanced cell death. Here, we found that the down-regulation of SMN causes mitochondrial dysfunction and subsequent cell death in in vitro models of skeletal myogenesis with both a murine C2C12 cell line and human induced pluripotent stem cells. During myogenesis, SMN binds to the upstream genomic regions of MYOD1 and microRNA (miR)-1 and miR-206. Accordingly, the loss of SMN down-regulates these miRs, whereas supplementation of the miRs recovers the mitochondrial function, cell survival, and myotube formation of SMN-deficient C2C12, indicating the SMN-miR axis is essential for myogenic metabolic maturation. In addition, the introduction of the miRs into ex vivo muscle stem cells derived from Δ7-SMA mice caused myotube formation and muscle contraction. In conclusion, our data revealed novel transcriptional roles of SMN during myogenesis, providing an alternative muscle-oriented therapeutic strategy for SMA patients.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography