Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cells ; 11(18)2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-36139418

RESUMEN

For many years optimal treatment for dysfunctional skeletal muscle characterized, for example, by impaired or limited regeneration, has been searched. Among the crucial factors enabling its development is finding the appropriate source of cells, which could participate in tissue reconstruction or serve as an immunomodulating agent (limiting immune response as well as fibrosis, that is, connective tissue formation), after transplantation to regenerating muscles. MSCs, including those derived from bone marrow, are considered for such applications in terms of their immunomodulatory properties, as their naive myogenic potential is rather limited. Injection of autologous (syngeneic) or allogeneic BMSCs has been or is currently being tested and compared in many potential clinical treatments. In the present study, we verified which approach, that is, the transplantation of either syngeneic or allogeneic BMSCs or the injection of BMSC-conditioned medium, would be the most beneficial for skeletal muscle regeneration. To properly assess the influence of the tested treatments on the inflammation, the experiments were carried out using immunocompetent mice, which allowed us to observe immune response. Combined analysis of muscle histology, immune cell infiltration, and levels of selected chemokines, cytokines, and growth factors important for muscle regeneration, showed that muscle injection with BMSC-conditioned medium is the most beneficial strategy, as it resulted in reduced inflammation and fibrosis development, together with enhanced new fiber formation, which may be related to, i.e., elevated level of IGF-1. In contrast, transplantation of allogeneic BMSCs to injured muscles resulted in a visible increase in the immune response, which hindered regeneration by promoting connective tissue formation. In comparison, syngeneic BMSC injection, although not detrimental to muscle regeneration, did not result in such significant improvement as CM injection.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Mesenquimatosas , Animales , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Fibrosis , Inflamación/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Músculo Esquelético
2.
Cells ; 10(9)2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34571854

RESUMEN

PAX7 transcription factor plays a crucial role in embryonic myogenesis and in adult muscles in which it secures proper function of satellite cells, including regulation of their self renewal. PAX7 downregulation is necessary for the myogenic differentiation of satellite cells induced after muscle damage, what is prerequisite step for regeneration. Using differentiating pluripotent stem cells we documented that the absence of functional PAX7 facilitates proliferation. Such action is executed by the modulation of the expression of two proteins involved in the DNA methylation, i.e., Dnmt3b and Apobec2. Increase in Dnmt3b expression led to the downregulation of the CDK inhibitors and facilitated cell cycle progression. Changes in Apobec2 expression, on the other hand, differently impacted proliferation/differentiation balance, depending on the experimental model used.


Asunto(s)
Desaminasas APOBEC/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteínas Musculares/metabolismo , Factor de Transcripción PAX7/metabolismo , Desaminasas APOBEC/genética , Animales , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Proliferación Celular/genética , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Femenino , Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Desarrollo de Músculos/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/fisiología , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/fisiología , Células Satélite del Músculo Esquelético/metabolismo , ADN Metiltransferasa 3B
3.
Stem Cell Res Ther ; 12(1): 448, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34372911

RESUMEN

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.


Asunto(s)
Células Madre Mesenquimatosas , Animales , Médula Ósea , Células de la Médula Ósea , Diferenciación Celular , Células Cultivadas , Hipoxia , Ratones , Músculo Esquelético , Mioblastos , Células Madre , Porcinos
4.
Cells ; 9(6)2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32560483

RESUMEN

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.


Asunto(s)
Quimiocina CXCL12/farmacología , Interleucina-4/farmacología , Mioblastos/citología , Células del Estroma/efectos de los fármacos , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Humanos , Ratones , Regeneración/efectos de los fármacos , Trasplante de Células Madre/métodos , Células Madre/citología , Células Madre/efectos de los fármacos
5.
Stem Cell Res Ther ; 11(1): 238, 2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32552916

RESUMEN

BACKGROUND: Pluripotent stem cells present the ability to self-renew and undergo differentiation into any cell type building an organism. Importantly, a lot of evidence on embryonic stem cell (ESC) differentiation comes from in vitro studies. However, ESCs cultured in vitro do not necessarily behave as cells differentiating in vivo. For this reason, we used teratomas to study early and advanced stages of in vivo ESC myogenic differentiation and the role of Pax7 in this process. Pax7 transcription factor plays a crucial role in the formation and differentiation of skeletal muscle precursor cells during embryonic development. It controls the expression of other myogenic regulators and also acts as an anti-apoptotic factor. It is also involved in the formation and maintenance of satellite cell population. METHODS: In vivo approach we used involved generation and analysis of pluripotent stem cell-derived teratomas. Such model allows to analyze early and also terminal stages of tissue differentiation, for example, terminal stages of myogenesis, including the formation of innervated and vascularized mature myofibers. RESULTS: We determined how the lack of Pax7 function affects the generation of different myofiber types. In Pax7-/- teratomas, the skeletal muscle tissue occupied significantly smaller area, as compared to Pax7+/+ ones. The proportion of myofibers expressing Myh3 and Myh2b did not differ between Pax7+/+ and Pax7-/- teratomas. However, the area of Myh7 and Myh2a myofibers was significantly lower in Pax7-/- ones. Molecular characteristic of skeletal muscles revealed that the levels of mRNAs coding Myh isoforms were significantly lower in Pax7-/- teratomas. The level of mRNAs encoding Pax3 was significantly higher, while the expression of Nfix, Eno3, Mck, Mef2a, and Itga7 was significantly lower in Pax7-/- teratomas, as compared to Pax7+/+ ones. We proved that the number of satellite cells in Pax7-/- teratomas was significantly reduced. Finally, analysis of neuromuscular junction localization in samples prepared with the iDISCO method confirmed that the organization of neuromuscular junctions in Pax7-/- teratomas was impaired. CONCLUSIONS: Pax7-/- ESCs differentiate in vivo to embryonic myoblasts more readily than Pax7+/+ cells. In the absence of functional Pax7, initiation of myogenic differentiation is facilitated, and as a result, the expression of mesoderm embryonic myoblast markers is upregulated. However, in the absence of functional Pax7 neuromuscular junctions, formation is abnormal, what results in lower differentiation potential of Pax7-/- ESCs during advanced stages of myogenesis.


Asunto(s)
Células Satélite del Músculo Esquelético , Teratoma , Animales , Diferenciación Celular , Ratones , Células Madre Embrionarias de Ratones , Desarrollo de Músculos/genética , Músculo Esquelético , Factores de Transcripción NFI , Factor de Transcripción PAX7/genética , Teratoma/genética
6.
Cells ; 9(6)2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32466405

RESUMEN

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.


Asunto(s)
Músculo Esquelético/patología , Osificación Heterotópica/patología , Animales , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Osificación Heterotópica/diagnóstico , Osificación Heterotópica/terapia , Osteogénesis , Transducción de Señal
7.
Int J Mol Sci ; 21(9)2020 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-32392778

RESUMEN

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.


Asunto(s)
Tejido Adiposo/citología , Quimiocina CXCL12/farmacología , Interleucina-4/farmacología , Músculo Esquelético/lesiones , Regeneración , Células del Estroma/trasplante , Tejido Adiposo/efectos de los fármacos , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Ratones , Músculo Esquelético/fisiología , Ratas , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Cicatrización de Heridas
8.
Stem Cell Res Ther ; 10(1): 343, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31753006

RESUMEN

BACKGROUND: Satellite cells, a population of unipotent stem cells attached to muscle fibers, determine the excellent regenerative capability of injured skeletal muscles. Myogenic potential is also exhibited by other cell populations, which exist in the skeletal muscles or come from other niches. Mesenchymal stromal/stem cells inhabiting the bone marrow do not spontaneously differentiate into muscle cells, but there is some evidence that they are capable to follow the myogenic program and/or fuse with myoblasts. METHODS: In the present study we analyzed whether IGF-1, IL-4, IL-6, and SDF-1 could impact human and porcine bone marrow-derived mesenchymal stromal/stem cells (hBM-MSCs and pBM-MSCs) and induce expression of myogenic regulatory factors, skeletal muscle-specific structural, and adhesion proteins. Moreover, we investigated whether these factors could induce both types of BM-MSCs to fuse with myoblasts. IGF-1, IL-4, IL-6, and SDF-1 were selected on the basis of their role in embryonic myogenesis as well as skeletal muscle regeneration. RESULTS: We found that hBM-MSCs and pBM-MSCs cultured in vitro in the presence of IGF-1, IL-4, IL-6, or SDF-1 did not upregulate myogenic regulatory factors. Consequently, we confirmed the lack of their naïve myogenic potential. However, we noticed that IL-4 and IL-6 impacted proliferation and IL-4, IL-6, and SDF-1 improved migration of hBM-MSCs. IL-4 treatment resulted in the significant increase in the level of mRNA encoding CD9, NCAM, VCAM, and m-cadherin, i.e., proteins engaged in cell fusion during myotube formation. Additionally, the CD9 expression level was also driven by IGF-1 treatment. Furthermore, the pre-treatment of hBM-MSCs either with IGF-1, IL-4, or SDF-1 and treatment of pBM-MSCs either with IGF-1 or IL-4 increased the efficacy of hybrid myotube formation between these cells and C2C12 myoblasts. CONCLUSIONS: To conclude, our study revealed that treatment with IGF-1, IL-4, IL-6, or SDF-1 affects BM-MSC interaction with myoblasts; however, it does not directly promote myogenic differentiation of these cells.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Madre Mesenquimatosas/metabolismo , Fibras Musculares Esqueléticas/fisiología , Mioblastos/metabolismo , Regeneración , Animales , Células de la Médula Ósea/citología , Fusión Celular , Línea Celular , Humanos , Células Madre Mesenquimatosas/citología , Fibras Musculares Esqueléticas/citología , Mioblastos/citología , Porcinos
9.
Int J Mol Sci ; 20(16)2019 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-31412558

RESUMEN

Pluripotent stem cells convert into skeletal muscle tissue during teratoma formation or chimeric animal development. Thus, they are characterized by naive myogenic potential. Numerous attempts have been made to develop protocols enabling efficient and safe conversion of pluripotent stem cells into functional myogenic cells in vitro. Despite significant progress in the field, generation of myogenic cells from pluripotent stem cells is still challenging-i.e., currently available methods require genetic modifications, animal-derived reagents, or are long lasting-and, therefore, should be further improved. In the current study, we investigated the influence of interleukin 4, a factor regulating inter alia migration and fusion of myogenic cells and necessary for proper skeletal muscle development and maintenance, on pluripotent stem cells. We assessed the impact of interleukin 4 on proliferation, selected gene expression, and ability to fuse in case of both undifferentiated and differentiating mouse embryonic stem cells. Our results revealed that interleukin 4 slightly improves fusion of pluripotent stem cells with myoblasts leading to the formation of hybrid myotubes. Moreover, it increases the level of early myogenic genes such as Mesogenin1, Pax3, and Pax7 in differentiating embryonic stem cells. Thus, interleukin 4 moderately enhances competence of mouse pluripotent stem cells for myogenic conversion.


Asunto(s)
Interleucina-4/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Autorrenovación de las Células/genética , Técnicas de Cocultivo , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Interleucina-4/genética , Interleucina-4/farmacología , Ratones , Desarrollo de Músculos , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citología , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Células Madre Pluripotentes/efectos de los fármacos
10.
Int J Mol Sci ; 20(13)2019 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-31284492

RESUMEN

In case of large injuries of skeletal muscles the pool of endogenous stem cells, i.e., satellite cells, might be not sufficient to secure proper regeneration. Such failure in reconstruction is often associated with loss of muscle mass and excessive formation of connective tissue. Therapies aiming to improve skeletal muscle regeneration and prevent fibrosis may rely on the transplantation of different types of stem cell. Among such cells are adipose tissue-derived stromal cells (ADSCs) which are relatively easy to isolate, culture, and manipulate. Our study aimed to verify applicability of ADSCs in the therapies of severely injured skeletal muscles. We tested whether 3D structures obtained from Matrigel populated with ADSCs and transplanted to regenerating mouse gastrocnemius muscles could improve the regeneration. In addition, ADSCs used in this study were pretreated with myoblasts-conditioned medium or anti-TGFß antibody, i.e., the factors modifying their ability to proliferate, migrate, or differentiate. Analyses performed one week after injury allowed us to show the impact of 3D cultured control and pretreated ADSCs at muscle mass and structure, as well as fibrosis development immune response of the injured muscle.


Asunto(s)
Tejido Adiposo/citología , Colágeno/farmacología , Laminina/farmacología , Músculo Esquelético/patología , Proteoglicanos/farmacología , Regeneración/efectos de los fármacos , Animales , Anticuerpos/farmacología , Forma de la Célula/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Combinación de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/genética , Inflamación/patología , Masculino , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Mioblastos/citología , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/trasplante , Factor de Crecimiento Transformador beta/metabolismo
11.
Stem Cell Res Ther ; 9(1): 258, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30261919

RESUMEN

BACKGROUND: Skeletal muscle regeneration is possible thanks to unipotent stem cells, which are satellite cells connected to the myofibers. Populations of stem cells other than muscle-specific satellite cells are considered as sources of cells able to support skeletal muscle reconstruction. Among these are bone marrow-derived mesenchymal stem cells (BM-MSCs), which are multipotent, self-renewing stem cells present in the bone marrow stroma. Available data documenting the ability of BM-MSCs to undergo myogenic differentiation are not definitive. In the current work, we aimed to check if the satellite cell niche could impact the ability of bone marrow-derived cells to follow a myogenic program. METHODS: We established a new in-vitro method for the coculture of bone marrow-derived cells (BMCs) that express CXCR4 (CXCR4+BMCs; the stromal-derived factor-1 (Sdf-1) receptor) with myofibers. Using various tests, we analyzed the myogenic identity of BMCs and their ability to fuse with myoblasts in vitro and in vivo. RESULTS: We showed that Sdf-1 treatment increased the number of CXCR4+BMCs able to bind the myofiber and occupy the satellite cell niche. Moreover, interaction with myofibers induced the expression of myogenic regulatory factors (MRFs) in CXCR4+BMCs. CXCR4+BMCs, pretreated by the coculture with myofibers and Sdf-1, participated in myotube formation in vitro and also myofiber reconstruction in vivo. We also showed that Sdf-1 overexpression in vivo (in injured and regenerating muscles) supported the participation of CXCR4+BMCs in new myofiber formation. CONCLUSION: We showed that CXCR4+BMC interaction with myofibers (that is, within the satellite cell niche) induced CXCR4+BMC myogenic commitment. CXCR4+BMCs, pretreated using such a method of culture, were able to participate in skeletal muscle regeneration.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , Regeneración/genética , Células Satélite del Músculo Esquelético/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Técnicas de Cocultivo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Masculino , Células Madre Mesenquimatosas/citología , Ratones , Ratones Transgénicos , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/lesiones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Cultivo Primario de Células , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Células Satélite del Músculo Esquelético/citología , Transducción de Señal
12.
Int J Dev Biol ; 61(1-2): 1-3, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28287240

RESUMEN

Professor Andrzej Krzysztof Tarkowski passed away last September (2016) at the age of 83. His findings, have become indispensable tools for immunological, genetic, and oncological studies, as well as for generating transgenic animals which are instrumental for studying gene function in living animals. His work and discoveries provided a tremendous input to the contemporary developmental biology of mammals.


Asunto(s)
Biología Evolutiva/historia , Historia del Siglo XX , Historia del Siglo XXI , Polonia
13.
Eur J Cell Biol ; 96(1): 47-60, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28017376

RESUMEN

Regeneration of skeletal muscle relies on the presence of satellite cells. Satellite cells deficiency accompanying some degenerative diseases is the reason for the search for the "replacement cells" that can be used in the muscle therapies. Due to their unique properties embryonic stem cells (ESCs), as well as myogenic cells derived from them, are considered as a promising source of therapeutic cells. Among the factors crucial for the specification of myogenic precursor cells is Pax7 that sustains proper function of satellite cells. In our previous studies we showed that ESCs lacking functional Pax7 are able to form myoblasts in vitro when differentiated within embryoid bodies and their outgrowths. In the current study we showed that ESCs lacking functional Pax7, cultured in vitro in monolayer in the medium supplemented with horse serum and 5azaC, expressed higher levels of factors associated with myogenesis, such as Pdgfra, Pax3, Myf5, and MyoD. Importantly, skeletal myosin immunolocalization confirmed that myogenic differentiation of ESCs was more effective in case of cells lacking Pax7. Our in vivo studies showed that ESCs transplanted into regenerating skeletal muscles were detectable at day 7 of regeneration and the number of Pax7-/- ESCs detected was significantly higher than of control cells. Our results support the concept that lack of functional Pax7 promotes proliferation of differentiating ESCs and for this reason more of them can turn into myogenic lineage.


Asunto(s)
Azacitidina/farmacología , Células Madre Embrionarias de Ratones , Músculo Esquelético/fisiología , Factor de Transcripción PAX7/deficiencia , Regeneración/efectos de los fármacos , Trasplante de Células Madre , Animales , Femenino , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Embrionarias de Ratones/trasplante , Regeneración/genética
14.
J Cachexia Sarcopenia Muscle ; 7(4): 483-96, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27239402

RESUMEN

BACKGROUND: The skeletal muscle has the ability to regenerate after injury. This process is mediated mainly by the muscle specific stem cells, that is, satellite cells. In case of extensive damage or under pathological conditions, such as muscular dystrophy, the process of muscle reconstruction does not occur properly. The aim of our study was to test whether mobilized stem cells, other than satellite cells, could participate in skeletal muscle reconstruction. METHODS: Experiments were performed on wild-type mice and mice lacking the functional Pax7 gene, that is, characterized by the very limited satellite cell population. Gastrocnemius mice muscles were injured by cardiotoxin injection, and then the animals were treated by stromal derived factor-1 (Sdf-1) with or without granulocyte-colony stimulating factor (G-CSF) for 4 days. The muscles were subjected to thorough assessment of the tissue regeneration process using histological and in vitro methods, as well as evaluation of myogenic factors' expression at the transcript and protein levels. RESULTS: Stromal derived factor-1 alone and Sdf-1 in combination with G-CSF significantly improved the regeneration of Pax7-/- skeletal muscles. The Sdf-1 and G-CSF treatment caused an increase in the number of mononucleated cells associated with muscle fibres. Further analysis showed that Sdf-1 and G-CSF treatment led to the rise in the number of CD34+ and Cxcr4+ cells and expression of Cxcr7. CONCLUSIONS: Stromal derived factor-1 and G-CSF stimulated regeneration of the skeletal muscles deficient in satellite cells. We suggest that mobilized CD34+, Cxcr4+, and Cxcr7+ cells can efficiently participate in the skeletal muscle reconstruction and compensate for the lack of satellite cells.

15.
J Muscle Res Cell Motil ; 36(6): 363-75, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26715014

RESUMEN

Pluripotent stem cells (PSCs), such as embryonic stem cells or induced pluripotent stem cells are a promising source of cells for regenerative medicine as they can differentiate into all cell types building a mammalian body. However, protocols leading to efficient and safe in vitro generation of desired cell types must be perfected before PSCs can be used in cell therapies or tissue engineering. In vivo, i.e. in developing mouse embryo or teratoma, PSCs can differentiate into skeletal muscle, but in vitro their spontaneous differentiation into myogenic cells is inefficient. Numerous attempts have been undertaken to enhance this process. Many of them involved mimicking the interactions occurring during embryonic myogenesis. The key regulators of embryonic myogenesis, such as Wnts proteins, fibroblast growth factor 2, and retinoic acid, have been tested to improve the frequency of in vitro myogenic differentiation of PSCs. This review summarizes the current state of the art, comparing spontaneous and directed myogenic differentiation of PSCs as well as the protocols developed this far to facilitate this process.


Asunto(s)
Desarrollo de Músculos/fisiología , Células Madre Pluripotentes/fisiología , Animales , Diferenciación Celular/fisiología , Humanos , Músculo Esquelético/fisiología
16.
J Muscle Res Cell Motil ; 36(6): 395-404, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26613733

RESUMEN

The skeletal muscle injury triggers the inflammatory response which is crucial for damaged muscle fiber degradation and satellite cell activation. Immunodeficient mice are often used as a model to study the myogenic potential of transplanted human stem cells. Therefore, it is crucial to elucidate whether such model truly reflects processes occurring under physiological conditions. To answer this question we compared skeletal muscle regeneration of BALB/c, i.e. animals producing all types of inflammatory cells, and SCID mice. Results of our study documented that initial stages of muscles regeneration in both strains of mice were comparable. However, lower number of mononucleated cells was noticed in regenerating SCID mouse muscles. Significant differences in the number of CD14-/CD45+ and CD14+/CD45+ cells between BALB/c and SCID muscles were also observed. In addition, we found important differences in M1 and M2 macrophage levels of BALB/c and SCID mouse muscles identified by CD68 and CD163 markers. Thus, our data show that differences in inflammatory response during muscle regeneration, were not translated into significant modifications in muscle regeneration.


Asunto(s)
Inflamación/patología , Músculo Esquelético/patología , Regeneración/fisiología , Animales , Biomarcadores/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Inflamación/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología
17.
Crit Rev Eukaryot Gene Expr ; 25(1): 1-11, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25955813

RESUMEN

Duchenne muscular dystrophy (DMD), the most common and most severe form of all muscular dystrophies, leads to progressive muscle fiber necrosis, fibroblast proliferation, and growth of fibrous tissue and fat. The most common cause of death in DMD patients is cardiac and respiratory failure. Current pharmacological and other treatment methods do not lead to full recovery. For this reason, new alternatives for skeletal muscle regeneration are being investigated. Transplantation of myoblasts from healthy donors is one studied approach to muscle treatment in DMD patients. However, the results of intramuscular injection of in vitro cultured myoblasts are still not satisfactory. The use of autologous stem cells is also proposed. Despite many ongoing studies, this therapy is still in preliminary testing and requires more experiments.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Distrofia Muscular de Duchenne/terapia , Mioblastos/trasplante , Trasplante de Células Madre , Adolescente , Niño , Preescolar , Humanos , Metaanálisis como Asunto , Distrofia Muscular de Duchenne/patología , Regeneración
18.
Stem Cell Res Ther ; 6: 46, 2015 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-25890097

RESUMEN

INTRODUCTION: Understanding the mechanism of stem cell mobilization into injured skeletal muscles is a prerequisite step for the development of muscle disease therapies. Many of the currently studied stem cell types present myogenic potential; however, when introduced either into the blood stream or directly into the tissue, they are not able to efficiently engraft injured muscle. For this reason their use in therapy is still limited. Previously, we have shown that stromal-derived factor-1 (Sdf-1) caused the mobilization of endogenous (not transplanted) stem cells into injured skeletal muscle improving regeneration. Here, we demonstrate that the beneficial effect of Sdf-1 relies on the upregulation of the tetraspanin CD9 expression in stem cells. METHODS: The expression pattern of adhesion proteins, including CD9, was analysed after Sdf-1 treatment during regeneration of rat skeletal muscles and mouse Pax7-/- skeletal muscles, that are characterized by the decreased number of satellite cells. Next, we examined the changes in CD9 level in satellite cells-derived myoblasts, bone marrow-derived mesenchymal stem cells, and embryonic stem cells after Sdf-1 treatment or silencing expression of CXCR4 and CXCR7. Finally, we examined the potential of stem cells to fuse with myoblasts after Sdf-1 treatment. RESULTS: In vivo analyses of Pax7-/- mice strongly suggest that Sdf-1-mediates increase in CD9 levels also in mobilized stem cells. In the absence of CXCR4 receptor the effect of Sdf-1 on CD9 expression is blocked. Next, in vitro studies show that Sdf-1 increases the level of CD9 not only in satellite cell-derived myoblasts but also in bone marrow derived mesenchymal stem cells, as well as embryonic stem cells. Importantly, the Sdf-1 treated cells migrate and fuse with myoblasts more effectively. CONCLUSIONS: We suggest that Sdf-1 binding CXCR4 receptor improves skeletal muscle regeneration by upregulating expression of CD9 and thus, impacting at stem cells mobilization to the injured muscles.


Asunto(s)
Quimiocina CXCL12/farmacología , Músculo Esquelético/fisiología , Regeneración , Células Madre/efectos de los fármacos , Tetraspanina 29/metabolismo , Animales , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/citología , Músculo Esquelético/lesiones , Mioblastos/citología , Mioblastos/metabolismo , Factor de Transcripción PAX7/deficiencia , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Receptores CXCR/antagonistas & inhibidores , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Madre/citología , Células Madre/metabolismo
19.
Stem Cells Dev ; 23(20): 2455-68, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24940624

RESUMEN

Pluripotent stem cells are a potential source of various cell types for use in regenerative medicine. Despite accumulating knowledge, there is currently no efficient and reproducible protocol that does not require genetic manipulation for generation of myogenic cells from pluripotent stem cells. Here, we examined whether mouse embryonic stem (ES) cells are able to undergo myogenic differentiation and fusion in response to signals released by differentiating myoblasts. Using ES cells expressing the histone 2B-green fluorescent fusion protein, we were able to detect hybrid myotubes formed by ES cells and differentiating myoblasts. ES cells that fused with myoblasts downregulated the expression of pluripotency markers and induced the expression of myogenic markers, while unfused ES cells did not exhibit this expression pattern. Thus, the signals released by myoblasts were not sufficient to induce myogenic differentiation of ES cells. Although ES cells synthesize many proteins involved in myoblast adhesion and fusion, we did not observe any myotubes formed exclusively by ES cells. We found that ES cells lacked M-cadherin and vascular cell adhesion molecule-1, which may account for the low frequency of hybrid myotube formation in ES cell-myoblast co-cultures and the inability of ES cells alone to form myotubes.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/metabolismo , Desarrollo de Músculos , Mioblastos/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Fusión Celular , Línea Celular , Células Madre Embrionarias/citología , Ratones , Mioblastos/citología
20.
Curr Stem Cell Res Ther ; 8(1): 82-90, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23270632

RESUMEN

Different sources of stem cells are considered as a potential source of precursor cells that could improve skeletal muscle regeneration. Under physiological conditions muscle regeneration is based on the satellite cells, i.e. adult muscle precursor cells that are localized between muscle fiber and surrounding basal lamina. These cells remain quiescent but after skeletal muscle injury activate, proliferate, differentiate, and fuse either to form new muscle fibers or reconstruct the damaged ones. As it was shown in many studies few populations of stem cells other than satellite cells are able to support skeletal muscle regeneration. Among them are mesenchymal stem cells (MSCs) that are present in many niches within adult organism and also in fetal tissues, such as human umbilical cord blood (HUCB) or umbilical cord connective tissue, i.e. Wharton's jelly. Thus, MSCs are intensively tested to prove that they are able to differentiate into various cell types, including skeletal myoblasts, and therefore could be useful in regenerative medicine. In our previous study we showed that MSCs isolated from Wharton's jelly expressed pluripotency as well as myogenic markers and were able to undergo myogenic differentiation both in vitro and in vivo. We also analyzed the potential of HUCB cells population which contains not only MSCs but also hematopoietic precursors. Our analyses of whole population of HUCB cells showed that these cells express myogenic regulatory factors, i.e. MyoD, and are able to contribute to skeletal muscle regeneration. In the present study we document that adherent fraction of HUCB cells, i.e. the cells that constitute the subpopulation enriched in MSCs, expresses pluripotency and myogenic markers, and have a positive impact at the regeneration of injured mouse skeletal muscle.


Asunto(s)
Sangre Fetal/citología , Células Madre Mesenquimatosas/citología , Desarrollo de Músculos/fisiología , Músculo Esquelético/fisiología , Medicina Regenerativa , Adulto , Animales , Diferenciación Celular , Células Cultivadas , Femenino , Sangre Fetal/fisiología , Humanos , Técnicas para Inmunoenzimas , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Músculo Esquelético/citología , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Embarazo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA