Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 373
Filtrar
1.
Curr Top Dev Biol ; 158: 53-82, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38670716

RESUMEN

Myocyte fusion is a pivotal process in the development and regeneration of skeletal muscle. Failure during fusion can lead to a range of developmental as well as pathological consequences. This review aims to comprehensively explore the intricate processes underlying myocyte fusion, from the molecular to tissue scale. We shed light on key players, such as the muscle-specific fusogens - Myomaker and Myomixer, in addition to some lesser studied molecules contributing to myocyte fusion. Conserved across vertebrates, Myomaker and Myomixer play a crucial role in driving the merger of plasma membranes of fusing myocytes, ensuring the formation of functional muscle syncytia. Our multiscale approach also delves into broader cell and tissue dynamics that orchestrate the timing and positioning of fusion events. In addition, we explore the relevance of muscle fusogens to human health and disease. Mutations in fusogen genes have been linked to congenital myopathies, providing unique insights into the molecular basis of muscle diseases. We conclude with a discussion on potential therapeutic avenues that may emerge from manipulating the myocyte fusion process to remediate skeletal muscle disorders.


Asunto(s)
Fusión Celular , Humanos , Animales , Músculo Esquelético/metabolismo , Músculo Esquelético/citología , Células Musculares/metabolismo , Células Musculares/citología , Proteínas Musculares/metabolismo , Proteínas Musculares/genética
2.
Int J Mol Sci ; 23(3)2022 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-35163664

RESUMEN

Cardiac patch implantation helps maximize the paracrine function of grafted cells and serves as a reservoir of soluble proangiogenic factors required for the neovascularization of infarcted hearts. We have previously fabricated a cardiac patch, EF-HAM, composed of a human amniotic membrane (HAM) coated with aligned PLGA electrospun fibers (EF). In this study, we aimed to evaluate the biocompatibility and angiogenic effects of EF-HAM scaffolds with varying fiber thicknesses on the paracrine behavior of skeletal muscle cells (SkM). Conditioned media (CM) obtained from SkM-seeded HAM and EF-HAM scaffolds were subjected to multiplex analysis of angiogenic factors and tested on HUVECs for endothelial cell viability, migration, and tube formation analyses. All three different groups of EF-HAM scaffolds demonstrated excellent biocompatibility with SkM. CM derived from SkM-seeded EF-HAM 7 min scaffolds contained significantly elevated levels of proangiogenic factors, including angiopoietin-1, IL-8, and VEGF-C compared to plain CM, which was obtained from SkM cultured on the plain surface. CM obtained from all SkM-seeded EF-HAM scaffolds significantly increased the viability of HUVECs compared to plain CM after five days of culture. However, only EF-HAM 7 min CM induced a higher migration capacity in HUVECs and formed a longer and more elaborate capillary-like network on Matrigel compared with plain CM. Surface roughness and wettability of EF-HAM 7 min scaffolds might have influenced the proportion of skeletal myoblasts and fibroblasts growing on the scaffolds and subsequently potentiated the angiogenic paracrine function of SkM. This study demonstrated the angioinductive properties of EF-HAM composite scaffold and its potential applications in the repair and regeneration of ischemic tissues.


Asunto(s)
Isquemia/terapia , Neovascularización Fisiológica , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Regeneración/fisiología , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Amnios , Angiopoyetina 1/metabolismo , Materiales Biocompatibles/química , Movimiento Celular , Supervivencia Celular , Medios de Cultivo Condicionados/farmacología , Fibroblastos/citología , Fibroblastos/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interleucina-8/metabolismo , Isquemia/patología , Células Musculares/citología , Células Musculares/metabolismo , Células Musculares/ultraestructura , Músculo Esquelético/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Int J Mol Sci ; 22(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34575979

RESUMEN

MicroRNAs (miRNAs) play an essential role in the regulation of a number of physiological functions. miR-133a and other muscular miRs (myomiRs) play a key role in muscle cell growth and in some type of cancers. Here, we show that miR133a is upregulated in individuals that undertake physical exercise. We used a skeletal muscle differentiation model to dissect miR-133a's role and to identify new targets, identifying Tropomyosin-4 (TPM4). This protein is expressed during muscle differentiation, but importantly it is an essential component of microfilament cytoskeleton and stress fibres formation. The microfilament scaffold remodelling is an essential step in cell transformation and tumour progression. Using the muscle system, we obtained valuable information about the microfilament proteins, and the knowledge on these molecular players can be transferred to the cytoskeleton rearrangement observed in cancer cells. Further investigations showed a role of TPM4 in cancer physiology, specifically, we found that miR-133a downregulation leads to TPM4 upregulation in colon carcinoma (CRC), and this correlates with a lower patient survival. At molecular level, we demonstrated in myocyte differentiation that TPM4 is positively regulated by the TA isoform of the p63 transcription factor. In muscles, miR-133a generates a myogenic stimulus, reducing the differentiation by downregulating TPM4. In this system, miR-133a counteracts the differentiative TAp63 activity. Interestingly, in CRC cell lines and in patient biopsies, miR-133a is able to regulate TPM4 activity, while TAp63 is not active. The downregulation of the miR leads to TPM4 overexpression, this modifies the architecture of the cell cytoskeleton contributing to increase the invasiveness of the tumour and associating with a poor prognosis. These results add data to the interesting question about the link between physical activity, muscle physiology and protection against colorectal cancer. The two phenomena have in common the cytoskeleton remodelling, due to the TPM4 activity, that is involved in stress fibres formation.


Asunto(s)
Diferenciación Celular/genética , Neoplasias del Colon/genética , MicroARNs/genética , Factores de Transcripción/genética , Tropomiosina/genética , Proteínas Supresoras de Tumor/genética , Citoesqueleto de Actina/genética , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias del Colon/patología , Citoesqueleto/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Células Musculares/citología , Desarrollo de Músculos/genética , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Fibras de Estrés/genética
4.
Genes Cells ; 26(7): 495-512, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33960573

RESUMEN

Mesenchymal stem cells (MSCs) are the likely precursors of multiple lines of mesenchymal cells. The existence of bona fide MSCs with self-renewal capacity and differentiation potential into all mesenchymal lineages, however, has been unclear because of the lack of MSC-specific marker(s) that are not expressed by the terminally differentiated progeny. Meflin, a glycosylphosphatidylinositol-anchored protein, is an MSC marker candidate that is specifically expressed in rare stromal cells in all tissues. Our previous report showed that Meflin expression becomes down-regulated in bone marrow-derived MSCs cultured on plastic, making it difficult to examine the self-renewal and differentiation of Meflin-positive cells at the single-cell level. Here, we traced the lineage of Meflin-positive cells in postnatal and adult mice, showing that those cells differentiated into white and brown adipocytes, osteocytes, chondrocytes and skeletal myocytes. Interestingly, cells derived from Meflin-positive cells formed clusters of differentiated cells, implying the in situ proliferation of Meflin-positive cells or their lineage-committed progenitors. These results, taken together with previous findings that Meflin expression in cultured MSCs was lost upon their multilineage differentiation, suggest that Meflin is a useful potential marker to localize MSCs and/or their immature progenitors in multiple tissues.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Inmunoglobulinas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Animales , Condrocitos/citología , Condrocitos/metabolismo , Inmunoglobulinas/genética , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Células Musculares/citología , Células Musculares/metabolismo , Osteocitos/citología , Osteocitos/metabolismo
5.
J Vet Med Sci ; 83(7): 1022-1030, 2021 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-33967186

RESUMEN

This study aimed to investigate the function of estrogen receptors (ERs) in myoregeneration and intermuscular adipogenesis. Ovariectomized (OVX) ERα knockout (KO) mice and ERß KO mice were used to assess the effect of estrogen on the myoregenerative process. Tibialis anterior muscle was collected on days 7, 10, and 14 after cardiotoxin injection to assess myotube morphology and adipogenesis area. Regenerated myotubes from OVX-ERß KO mice were consistently smaller in diameter than those from OVX-ERα KO and OVX-wild-type mice, whereas the adipogenesis area of OVX-ERß KO mice was consistently greater than that of the other types. Therefore, ERß may be an influential factor in promoting myoregeneration and adipogenesis inhibition compared to ERα.


Asunto(s)
Adipogénesis , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Células Musculares/citología , Regeneración , Animales , Estradiol , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Estrógenos , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovariectomía/veterinaria
6.
J Biol Chem ; 296: 100606, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33789162

RESUMEN

In addition to maintaining cellular ER Ca2+ stores, store-operated Ca2+ entry (SOCE) regulates several Ca2+-sensitive cellular enzymes, including certain adenylyl cyclases (ADCYs), enzymes that synthesize the secondary messenger cyclic AMP (cAMP). Ca2+, acting with calmodulin, can also increase the activity of PDE1-family phosphodiesterases (PDEs), which cleave the phosphodiester bond of cAMP. Surprisingly, SOCE-regulated cAMP signaling has not been studied in cells expressing both Ca2+-sensitive enzymes. Here, we report that depletion of ER Ca2+ activates PDE1C in human arterial smooth muscle cells (HASMCs). Inhibiting the activation of PDE1C reduced the magnitude of both SOCE and subsequent Ca2+/calmodulin-mediated activation of ADCY8 in these cells. Because inhibiting or silencing Ca2+-insensitive PDEs had no such effects, these data identify PDE1C-mediated hydrolysis of cAMP as a novel and important link between SOCE and its activation of ADCY8. Functionally, we showed that PDE1C regulated the formation of leading-edge protrusions in HASMCs, a critical early event in cell migration. Indeed, we found that PDE1C populated the tips of newly forming leading-edge protrusions in polarized HASMCs, and co-localized with ADCY8, the Ca2+ release activated Ca2+ channel subunit, Orai1, the cAMP-effector, protein kinase A, and an A-kinase anchoring protein, AKAP79. Because this polarization could allow PDE1C to control cAMP signaling in a hyper-localized manner, we suggest that PDE1C-selective therapeutic agents could offer increased spatial specificity in HASMCs over agents that regulate cAMP globally in cells. Similarly, such agents could also prove useful in regulating crosstalk between Ca2+/cAMP signaling in other cells in which dysregulated migration contributes to human pathology, including certain cancers.


Asunto(s)
Arterias/citología , Calcio/metabolismo , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Células Musculares/citología , Transducción de Señal , Transporte Biológico , Movimiento Celular , Regulación Enzimológica de la Expresión Génica , Humanos , Cinética
7.
Nat Chem Biol ; 17(3): 326-334, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33199915

RESUMEN

Secreted polypeptides are a fundamental axis of intercellular and endocrine communication. However, a global understanding of the composition and dynamics of cellular secretomes in intact mammalian organisms has been lacking. Here, we introduce a proximity biotinylation strategy that enables labeling, detection and enrichment of secreted polypeptides in a cell type-selective manner in mice. We generate a proteomic atlas of hepatocyte, myocyte, pericyte and myeloid cell secretomes by direct purification of biotinylated secreted proteins from blood plasma. Our secretome dataset validates known cell type-protein pairs, reveals secreted polypeptides that distinguish between cell types and identifies new cellular sources for classical plasma proteins. Lastly, we uncover a dynamic and previously undescribed nutrient-dependent reprogramming of the hepatocyte secretome characterized by the increased unconventional secretion of the cytosolic enzyme betaine-homocysteine S-methyltransferase (BHMT). This secretome profiling strategy enables dynamic and cell type-specific dissection of the plasma proteome and the secreted polypeptides that mediate intercellular signaling.


Asunto(s)
Betaína-Homocisteína S-Metiltransferasa/genética , Biotina/química , Proteínas Sanguíneas/genética , Hepatocitos/metabolismo , Proteoma/genética , Coloración y Etiquetado/métodos , Animales , Betaína-Homocisteína S-Metiltransferasa/metabolismo , Biotina/administración & dosificación , Biotinilación , Proteínas Sanguíneas/metabolismo , Expresión Génica , Células HEK293 , Hepatocitos/citología , Humanos , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos C57BL , Células Musculares/citología , Células Musculares/metabolismo , Células Mieloides/citología , Células Mieloides/metabolismo , Especificidad de Órganos , Pericitos/citología , Pericitos/metabolismo , Proteoma/metabolismo , Proteómica/métodos
8.
Bratisl Lek Listy ; 121(12): 853-857, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33300353

RESUMEN

OBJECTIVES: The objective of this study was to evaluate the effect of IGF1 and FGF2 growth factors on the differentiation of human adipose tissue­derived mesenchymal stem cells (MSCs) into skeletal muscle cells. METHODS: MSCs were divided into four groups. Group I (control group) was incubated only in myogenic medium, group II was incubated by adding 100 ng/ml FGF2 to the myogenic medium, group III was incubated by adding 20 ng/ml IGF1 to the myogenic medium, group IV was incubated by adding 100 ng/ml FGF2 and 20 ng/ml IGF1 to the myogenic medium. Cells dyed with MyoD1 antibodies were analyzed by flow cytometry so as to determine the myogenic differentiation on day 28. RESULTS: It was confirmed that 11.5 % of the control group, 39.2 % of the FGF2 group, 23.1 % of the IGF1 group, and 39.3 % of the IGF1-FGF2 group showed differentiation. CONCLUSION: Our results show that IGF1 and FGF2 have a positive contribution to myogenic differentiation of MSCs. They contribute to the studies related to muscle diseases and their treatment by the fact that growth factors support the feature of regeneration capacity and differentiation (Tab. 2, Fig. 7, Ref. 23).


Asunto(s)
Diferenciación Celular , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Células Madre Mesenquimatosas/citología , Células Musculares/citología , Músculo Esquelético/citología , Células Cultivadas , Humanos
9.
Nat Commun ; 11(1): 5808, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-33199701

RESUMEN

Skeletal muscle promotes metabolic balance by regulating glucose uptake and the stimulation of multiple interorgan crosstalk. We show here that the catalytic activity of Vav2, a Rho GTPase activator, modulates the signaling output of the IGF1- and insulin-stimulated phosphatidylinositol 3-kinase pathway in that tissue. Consistent with this, mice bearing a Vav2 protein with decreased catalytic activity exhibit reduced muscle mass, lack of proper insulin responsiveness and, at much later times, a metabolic syndrome-like condition. Conversely, mice expressing a catalytically hyperactive Vav2 develop muscle hypertrophy and increased insulin responsiveness. Of note, while hypoactive Vav2 predisposes to, hyperactive Vav2 protects against high fat diet-induced metabolic imbalance. These data unveil a regulatory layer affecting the signaling output of insulin family factors in muscle.


Asunto(s)
Biocatálisis , Homeostasis , Metabolismo , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Transducción de Señal , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Biocatálisis/efectos de los fármacos , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Línea Celular , Tamaño de la Célula/efectos de los fármacos , Genotipo , Glucosa/farmacología , Homeostasis/efectos de los fármacos , Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Células Musculares/citología , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Músculo Esquelético/efectos de los fármacos , Tamaño de los Órganos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo
10.
Bull Exp Biol Med ; 169(6): 811-814, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33123916

RESUMEN

The distribution of NO and H2S in the arterial vessels of the anterior abdominal wall after implantation of a polypropylene mesh was studied by immunohistochemical methods at different stages of healing of the surgical wound in mature male Wistar rats. The presence of enzymes of NO and H2S synthesis in the wall of arterial vessels of the soft tissues of the anterior abdominal wall has been established. It has been shown that endothelial NO synthase is localized exclusively in the endothelium of both large and small vessels. Cystathionine γ lyase in small vessels is located only in the endothelial lining, whereas in large arteries and vessels of medium caliber, it is located in the endothelium and in myocytes. Inducible NO synthase appears in the artery wall only in animals with implanted polypropylene mesh by day 5 of the postoperative period, reaching the maximum by day 10. The content and localization of cystathionine γ lyase in the vascular wall of sham-operated and experimental rats did not much differ from the control values.


Asunto(s)
Cistationina gamma-Liasa/genética , Endotelio Vascular/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo II/genética , Polipropilenos/farmacología , Mallas Quirúrgicas , Pared Abdominal/irrigación sanguínea , Pared Abdominal/cirugía , Animales , Vasos Sanguíneos/citología , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/enzimología , Cistationina gamma-Liasa/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Expresión Génica , Sulfuro de Hidrógeno/metabolismo , Implantes Experimentales , Masculino , Células Musculares/citología , Células Musculares/efectos de los fármacos , Células Musculares/enzimología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Ratas Wistar , Cicatrización de Heridas
11.
PLoS Genet ; 16(10): e1009022, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33125370

RESUMEN

Adult skeletal muscles are maintained during homeostasis and regenerated upon injury by muscle stem cells (MuSCs). A heterogeneity in self-renewal, differentiation and regeneration properties has been reported for MuSCs based on their anatomical location. Although MuSCs derived from extraocular muscles (EOM) have a higher regenerative capacity than those derived from limb muscles, the molecular determinants that govern these differences remain undefined. Here we show that EOM and limb MuSCs have distinct DNA methylation signatures associated with enhancers of location-specific genes, and that the EOM transcriptome is reprogrammed following transplantation into a limb muscle environment. Notably, EOM MuSCs expressed host-site specific positional Hox codes after engraftment and self-renewal within the host muscle. However, about 10% of EOM-specific genes showed engraftment-resistant expression, pointing to cell-intrinsic molecular determinants of the higher engraftment potential of EOM MuSCs. Our results underscore the molecular diversity of distinct MuSC populations and molecularly define their plasticity in response to microenvironmental cues. These findings provide insights into strategies designed to improve the functional capacity of MuSCs in the context of regenerative medicine.


Asunto(s)
Plasticidad de la Célula/genética , Epigenoma/genética , Trasplante de Células Madre , Transcriptoma/genética , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Proliferación Celular/genética , Extremidades/crecimiento & desarrollo , Variación Genética/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Células Musculares/citología , Fibras Musculares Esqueléticas , Músculo Esquelético/citología , Mioblastos/citología , Regeneración/genética , Células Madre/citología , Células Madre/metabolismo
12.
Int J Mol Sci ; 21(21)2020 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-33114544

RESUMEN

The 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (statins) exert pleiotropic effects on cardiac cell biology which are not yet fully understood. Here we tested whether statin treatment affects resident endogenous cardiac stem/progenitor cell (CSC) activation in vitro and in vivo after myocardial infarction (MI). Statins (Rosuvastatin, Simvastatin and Pravastatin) significantly increased CSC expansion in vitro as measured by both BrdU incorporation and cell growth curve. Additionally, statins increased CSC clonal expansion and cardiosphere formation. The effects of statins on CSC growth and differentiation depended on Akt phosphorylation. Twenty-eight days after myocardial infarction by permanent coronary ligation in rats, the number of endogenous CSCs in the infarct border zone was significantly increased by Rosuvastatin-treatment as compared to untreated controls. Additionally, commitment of the activated CSCs into the myogenic lineage (c-kitpos/Gata4pos CSCs) was increased by Rosuvastatin administration. Accordingly, Rosuvastatin fostered new cardiomyocyte formation after MI. Finally, Rosuvastatin treatment reversed the cardiomyogenic defects of CSCs in c-kit haploinsufficient mice, increasing new cardiomyocyte formation by endogenous CSCs in these mice after myocardial infarction. In summary, statins, by sustaining Akt activation, foster CSC growth and differentiation in vitro and in vivo. The activation and differentiation of the endogenous CSC pool and consequent new myocyte formation by statins improve myocardial remodeling after coronary occlusion in rodents. Similar effects might contribute to the beneficial effects of statins on human cardiovascular diseases.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Células Musculares/citología , Infarto del Miocardio/tratamiento farmacológico , Miocardio/citología , Células Madre/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ratones , Células Musculares/efectos de los fármacos , Células Musculares/metabolismo , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Fosforilación/efectos de los fármacos , Pravastatina/administración & dosificación , Pravastatina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Rosuvastatina Cálcica/administración & dosificación , Rosuvastatina Cálcica/farmacología , Simvastatina/administración & dosificación , Simvastatina/farmacología , Células Madre/citología , Células Madre/metabolismo
13.
Clin Ther ; 42(10): 1880-1891, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32938532

RESUMEN

PURPOSE: Cardiac pathologies remain a dominant cause of morbidity and mortality within the community. The drive to develop therapies capable of repairing damaged heart tissue to achieve clinically significant restoration of function has motivated the pursuit of novel approaches such as cell therapy. To this end, evidence of therapeutic benefits achieved by using mesenchymal stem cells (MSCs) has captured considerable interest despite a relative lack of information regarding the mechanisms involved. This narrative review synthesizes and interprets the current literature describing mechanisms by which MSCs can elicit cardiac repair, thereby directing attention to avenues of further inquiry. METHODS: OVID versions of MEDLINE and EMBASE were searched for studies describing the role of MSCs in mammalian cardiac repair. Additional studies were sourced from the reference lists of relevant articles and other personal files. FINDINGS: MSCs elicit cardiac repair in a range of in vitro systems and animal models of diseases such as myocardial infarction and heart failure. Important mechanisms include the preservation of myocardial contractility, the promotion of angiogenesis, and the modulation of fibrosis. Exposing in vitro MSCs to a microenvironment reflective of that encountered in the injured heart seems to potentiate these therapeutic mechanisms. IMPLICATIONS: Promising results in animal studies warrant continuation of clinical MSC cardiac therapy studies. Paracrine functions of MSCs seem to be the dominant mechanism of cardiac repair over direct cellular effects. Although integral, the MSC secretome remains poorly defined. In addition, most of the mechanistic data within the literature have been derived from animal MSC research, necessitating more human MSC-based work.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/terapia , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Fibroblastos/citología , Corazón/fisiopatología , Humanos , Células Musculares/citología
14.
Cell Signal ; 74: 109711, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32702440

RESUMEN

The CD34 protein is regarded as a marker of stem cells from multiple origins. Recently a mesenchymal progenitor CD34 positive cell identified from traumatized human skeletal muscle demonstrates differentiation capability into vascular endothelial cells, osteoblasts and adipocytes. Here they were treated with a small inhibitory RNA for CD34, which significantly reduced the cellular level of the CD34 protein. These treated cells had a reduced capacity to proliferate, and migrate. They were both unable to differentiation down multiple pathways and to undergo vascular endothelial differentiation as reflected by a lack of expression of VE cadherin, Tie 2 and CD31. Additionally the cells were unable to form tube-like structures in an endothelial tube assay. These treated cells were also unable to undergo osteogenesis, as revealed by lack of alizarin red and alkaline phosphatase staining and were unable to undergo adipogenesis as revealed by lack of oil red O staining. Finally, when CD34 was expressed in cells lacking this protein, the cells were able to undergo vascular endothelial differentiation as revealed by expression of Tie2, VE-cadherin and CD31. These data indicate that in cells derived from traumatized muscle the CD34 protein is required for enhanced proliferation, migration and differentiation down multiple pathways.


Asunto(s)
Antígenos CD34/fisiología , Células Madre Mesenquimatosas , Músculo Esquelético , Adipocitos/citología , Adipogénesis , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Células Endoteliales/citología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Musculares/citología , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Osteoblastos/citología , Osteogénesis
15.
Biochim Biophys Acta Mol Basis Dis ; 1866(8): 165809, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32353613

RESUMEN

No real therapeutic modality is currently available for Acute kidney injury (AKI) and if any, they are mainly supportive in nature. Therefore, developing a new therapeutic strategy is crucial. Mitochondrial dysfunction proved to be a key contributor to renal tubular cell death during AKI. Thus, replacement or augmentation of damaged mitochondria could be a proper target in AKI treatment. Here, in an animal model of AKI, we auto-transplanted normal mitochondria isolated from healthy muscle cells to injured kidney cells through injection to renal artery. The mitochondria transplantation prevented renal tubular cell death, restored renal function, ameliorated kidney damage, improved regenerative potential of renal tubules, and decreased ischemia/reperfusion-induced apoptosis. Although further studies including clinical trials are required in this regard, our findings suggest a novel therapeutic strategy for treatment of AKI. Improved quality of life of patients suffering from renal failure and decreased morbidity and mortality rates would be the potential advantages of this therapeutic strategy.


Asunto(s)
Lesión Renal Aguda/terapia , Mitocondrias/trasplante , Células Musculares/química , Recuperación de la Función/fisiología , Daño por Reperfusión/prevención & control , Lesión Renal Aguda/patología , Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/cirugía , Animales , Apoptosis , Fraccionamiento Celular/métodos , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Humanos , Pruebas de Función Renal , Túbulos Renales Proximales/patología , Túbulos Renales Proximales/fisiopatología , Túbulos Renales Proximales/cirugía , Masculino , Mitocondrias/fisiología , Células Musculares/citología , Músculo Esquelético/química , Músculo Esquelético/citología , Cultivo Primario de Células , Ratas , Ratas Wistar , Arteria Renal , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Daño por Reperfusión/cirugía
16.
Int J Mol Sci ; 21(9)2020 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-32344794

RESUMEN

Microgravity induces a number of significant physiological changes in the cardiovascular, nervous, immune systems, as well as the bone tissue of astronauts. Changes in cell adhesion properties are one aspect affected during long-term spaceflights in mammalian cells. Cellular adhesion behaviors can be divided into cell-cell and cell-matrix adhesion. These behaviors trigger cell-cell recognition, conjugation, migration, cytoskeletal rearrangement, and signal transduction. Cellular adhesion molecule (CAM) is a general term for macromolecules that mediate the contact and binding between cells or between cells and the extracellular matrix (ECM). In this review, we summarize the four major classes of adhesion molecules that regulate cell adhesion, including integrins, immunoglobulin superfamily (Ig-SF), cadherins, and selectin. Moreover, we discuss the effects of spaceflight and simulated microgravity on the adhesion of endothelial cells, immune cells, tumor cells, stem cells, osteoblasts, muscle cells, and other types of cells. Further studies on the effects of microgravity on cell adhesion and the corresponding physiological behaviors may help increase the safety and improve the health of astronauts in space.


Asunto(s)
Adhesión Celular , Vuelo Espacial , Simulación de Ingravidez , Animales , Astronautas , Moléculas de Adhesión Celular/clasificación , Moléculas de Adhesión Celular/fisiología , Citoesqueleto/ultraestructura , Células Endoteliales/citología , Matriz Extracelular/metabolismo , Humanos , Sistema Inmunológico/citología , Ratones , Células Musculares/citología , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/citología , Especificidad de Órganos , Osteoblastos/citología , Ratas , Células Madre/citología
17.
Cell Physiol Biochem ; 54(2): 271-286, 2020 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-32233339

RESUMEN

BACKGROUND/AIMS: Pericytes (PCs) are multipotent vascular precursors that play a critical physiological role in the development and maintenance of blood vessel integrity. In this study, we aim to characterize PCs isolated from human abdominal adipose tissue and develop an integration-free induced pluripotent stem cells (iPSCs) using episomal vectors. METHODS: The ultrastructure of adipose tissue-derived PCs was determined using scanning and transmission electron microscopy. The expression of mesenchymal stem cells (MSCs) and pericyte markers were examined using flow cytometry and PCR analysis. PCs were induced to adipogenic, osteogenic and myogenic lineages, and their angiogenic potential was determined using tube formation assay. We further established pericyte reprogramming protocol using episomal vectors. RESULTS: Our data showed that human adipose tissue-derived PCs uniformly expressed MSCs, CD105 and CD73, and PCs markers, desmin, and alpha smooth muscle actin (α-SMA), while lacked the expression of HLA-DR and the hematopoietic markers CD34, CD11b and CD45. Ultrastructure analysis showed typical internal structure for the PCs with a characteristic prominent eccentric nuclei and cytoplasmic invaginations forming a caveolar system. Functional analysis showed efficient differentiation into adipocytes, osteocytes, and myocyte-like cells. Adipose tissue-derived PCs showed angiogenic potential using tube-forming assay. To determine further application of these cells for personalized therapy, we reprogrammed PCs into induced pluripotent stem cells (iPSCs) using episomal vectors. Reprogrammed cells gradually lost their fusiform shape, acquired the epithelial cell morphology and formed colonies. Furthermore, reprogrammed cells successfully expressed the pluripotency markers OCT4, Nanog, SSEA-4, and ß-catenin, an early reprogramming marker. CONCLUSION: The accessibility and abundance of human fat supports the application of adipose derived PCs as a novel and promising source of cell therapy and regenerative medicine.


Asunto(s)
Tejido Adiposo/citología , Técnicas de Reprogramación Celular/métodos , Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Pericitos/citología , 5'-Nucleotidasa/metabolismo , Actinas/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Tejido Adiposo/ultraestructura , Linaje de la Célula , Células Cultivadas , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Desmina/metabolismo , Endoglina/metabolismo , Citometría de Flujo , Proteínas Ligadas a GPI/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/ultraestructura , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Células Musculares/citología , Células Musculares/metabolismo , Desarrollo de Músculos/genética , Proteína Homeótica Nanog/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Osteocitos/citología , Osteocitos/metabolismo , Osteogénesis/genética , Pericitos/metabolismo , Pericitos/ultraestructura , Antígenos Embrionarios Específico de Estadio/metabolismo , beta Catenina/metabolismo
18.
Life Sci ; 250: 117546, 2020 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-32184125

RESUMEN

AIM: The enzyme 3-phosphoinositide-dependent protein kinase-1 (PDK1) is associated with cardiac and pathological remodeling and ion channel function regulation. However, whether it regulates hyperpolarization-activated cyclic nucleotide-modulated channels (HCNs) remains unclear. MAIN METHODS: In the atrial myocytes of heart-specific PDK1 "knockout" mouse model and neonatal mice, protein kinase B (AKT)-related inhibitors or agonists as well as knockdown or overexpression plasmids were used to study the relationship between PDK1 and HCNs. KEY FINDINGS: HCN1 expression and AKT phosphorylation at the Thr308 site were significantly decreased in atrial myocytes after PDK1 knockout or inhibition; in contrast, HCN2 and HCN4 levels were significantly increased. Also, a similar trend of HCNs expression has been observed in cultured atrial myocytes after PDK1 inhibition, as further demonstrated via immunofluorescence and patch-clamp experiments. Moreover, these results of PDK1 overexpression indicate an opposite trend compared with the previous experimental results. However, the results of PDK1 inhibition or overexpression could be reversed by activating or inhibiting AKT, respectively. SIGNIFICANCE: These results indicate that the PDK1-AKT signaling pathway is involved in the regulation of HCN mRNA transcription, protein expression, HCN current density, and cell membrane location.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Regulación Enzimológica de la Expresión Génica , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Canales de Potasio/metabolismo , Transducción de Señal , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Células Cultivadas , Femenino , Eliminación de Gen , Atrios Cardíacos/citología , Masculino , Ratones , Ratones Noqueados , Células Musculares/citología , Técnicas de Placa-Clamp , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tirosina/metabolismo
19.
Cell Rep ; 30(10): 3552-3565.e6, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32160556

RESUMEN

Although the factors regulating muscle cell differentiation are well described, we know very little about how differentiating muscle fibers are organized into individual muscle tissue bundles. Disruption of these processes leads to muscle hypoplasia or dysplasia, and replicating these events is vital in tissue engineering approaches. We describe the progressive cellular events that orchestrate the formation of individual limb muscle bundles and directly demonstrate the role of the connective tissue cells that surround muscle precursors in controlling these events. We show how disruption of gene activity within or genetic ablation of connective tissue cells impacts muscle precursors causing disruption of muscle bundle formation and subsequent muscle dysplasia and hypoplasia. We identify several markers of the populations of connective tissue cells that surround muscle precursors and provide a model for how matrix-modifying proteoglycans secreted by these cells may influence muscle bundle formation by effects on the local extracellular matrix (ECM) environment.


Asunto(s)
Células del Tejido Conectivo/citología , Extremidades/fisiología , Desarrollo de Músculos , Músculo Esquelético/fisiología , Animales , Tipificación del Cuerpo , Agregación Celular , Eliminación de Gen , Integrasas/metabolismo , Ratones Transgénicos , Morfogénesis , Células Musculares/citología , Fibras Musculares Esqueléticas/citología , Proteínas de Dominio T Box/metabolismo , Tendones/citología , Factores de Transcripción/metabolismo
20.
Geriatr Gerontol Int ; 20(1): 78-84, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31758637

RESUMEN

AIM: We examined the changes in oxidative stress, mitochondrial function and muscle atrophy during aging in mice. METHODS: We used 6-, 12- and 24-month (6 M, 12 M and 24 M)-old C57BL/6J mice. Skeletal muscles were removed from the lower limb and used for quantitative real-time polymerase chain reaction, immunoblotting and histological analyses. RESULTS: The muscle weight and myocyte cross-sectional area were significantly decreased in the 12 M and 24 M mice compared with those of the 6 M mice. The levels of the oxidative stress markers, nicotinamide adenine dinucleotide phosphate oxidase 2, nicotinamide adenine dinucleotide phosphate oxidase 4, mitochondrial 4-hydroxy-2-nonenal and 3-nitrotyrosine, were significantly higher in the 24 M mice compared with those of the 6 M mice. Furthermore, the 24 M mice had lower levels of mitochondrial markers, peroxisome proliferator-activated receptor gamma coactivator 1 (PGC)-α, peroxisome proliferator-activated receptor gamma coactivator-1ß, sirtuin-1, adenosine triphosphate synthase mitochondria F1 complex α subunit 1 and mitochondrial cytochrome c oxidase 1. The ubiquitin-proteasome pathway genes muscle ring finger-1 and atrogin-1 were significantly upregulated in the 12 M and 24 M mice, and protein synthesis markers (phosphorylated-Akt and -p70 ribosomal S6 kinase) were significantly lower in the 24 M mice compared with the 6 M mice (all P < 0.05). CONCLUSIONS: These findings have important implications for the mechanisms that underlie sarcopenia and frailty processes. Geriatr Gerontol Int 2020; 20: 78-84.


Asunto(s)
Envejecimiento/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Estrés Oxidativo/genética , Envejecimiento/genética , Animales , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/fisiología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/enzimología , Mitocondrias/genética , Células Musculares/citología , Células Musculares/metabolismo , Atrofia Muscular/genética , Estrés Oxidativo/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA