Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Development ; 149(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35029658

RESUMEN

Worldwide prevalence of obesity is associated with the increase of lifestyle-related diseases. The accumulation of intermuscular adipose tissue (IMAT) is considered a major problem whereby obesity leads to sarcopenia and metabolic disorders and thus is a promising target for treating these pathological conditions. However, whereas obesity-associated IMAT is suggested to originate from PDGFRα+ mesenchymal progenitors, the processes underlying this adipogenesis remain largely unexplored. Here, we comprehensively investigated intra- and extracellular changes associated with these processes using single-cell RNA sequencing and mass spectrometry. Our single-cell RNA sequencing analysis identified a small PDGFRα+ cell population in obese mice directed strongly toward adipogenesis. Proteomic analysis showed that the appearance of this cell population is accompanied by an increase in galectin-3 in interstitial environments, which was found to activate adipogenic PPARγ signals in PDGFRα+ cells. Moreover, IMAT formation during muscle regeneration was significantly suppressed in galectin-3 knockout mice. Our findings, together with these multi-omics datasets, could unravel microenvironmental networks during muscle regeneration highlighting possible therapeutic targets against IMAT formation in obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Galectina 3/metabolismo , Músculo Esquelético/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Actinas/genética , Actinas/metabolismo , Adipogénesis , Tejido Adiposo/citología , Animales , Cardiotoxinas/farmacología , Diferenciación Celular , Senescencia Celular/genética , Dieta Alta en Grasa , Femenino , Galectina 3/deficiencia , Galectina 3/genética , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Obesidad/metabolismo , Obesidad/patología , PPAR gamma/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/deficiencia , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Regeneración , Transducción de Señal/genética
2.
Stem Cells ; 41(6): 552-559, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-36943314

RESUMEN

Skeletal muscle is mainly composed of multinucleated cells called myofibers and has excellent regenerative and adaptive abilities. These abilities are granted by muscle satellite cells (MuSCs), which are anatomically defined cells located between myofibers and basal lamina. In addition to myofibers and MuSCs, skeletal muscle contains several types of cells located in interstitial areas, such as mesenchymal progenitors. These cells are positive for platelet-derived growth factor receptor alpha and are called fibro/adipogenic progenitors (FAPs) or mesenchymal stromal cells. Although mesenchymal progenitors were originally identified as the causative cells of ectopic fat accumulation in skeletal muscles, recent studies have shed light on their beneficial roles in homeostasis, regeneration, and hypertrophy. Furthermore, the heterogeneity of mesenchymal progenitors is of great interest in understanding skeletal muscle development, homeostasis, regeneration, aging, and diseases. In this concise review, we summarize recent findings on the physiological roles of mesenchymal progenitors and their heterogeneity and discuss the remaining critical concerns.


Asunto(s)
Adipogénesis , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/metabolismo , Músculo Esquelético/metabolismo , Hipertrofia/metabolismo , Homeostasis , Diferenciación Celular/fisiología
3.
Development ; 146(4)2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30745427

RESUMEN

The undifferentiated state of muscle stem (satellite) cells (MuSCs) is maintained by the canonical Notch pathway. Although three bHLH transcriptional factors, Hey1, HeyL and Hes1, are considered to be potential effectors of the Notch pathway exerting anti-myogenic effects, neither HeyL nor Hes1 inhibits myogenic differentiation of myogenic cell lines. Furthermore, whether these factors work redundantly or cooperatively is unknown. Here, we showed cell-autonomous functions of Hey1 and HeyL in MuSCs using conditional and genetic null mice. Analysis of cultured MuSCs revealed anti-myogenic activity of both HeyL and Hes1. We found that HeyL forms heterodimeric complexes with Hes1 in living cells. Moreover, our ChIP-seq experiments demonstrated that, compared with HeyL alone, the HeyL-Hes1 heterodimer binds with high affinity to specific sites in the chromatin, including the binding sites of Hey1. Finally, analyses of myogenin promoter activity showed that HeyL and Hes1 act synergistically to suppress myogenic differentiation. Collectively, these results suggest that HeyL and Hey1 function redundantly in MuSCs, and that HeyL requires Hes1 for effective DNA binding and biological activity.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Regulación de la Expresión Génica , Células Satélite del Músculo Esquelético/citología , Factor de Transcripción HES-1/metabolismo , Alelos , Animales , Sitios de Unión , Separación Celular , Cromatina/química , ADN/química , Citometría de Flujo , Ratones , Ratones Noqueados , Ratones Transgénicos , Regiones Promotoras Genéticas , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Receptores Notch/metabolismo , Transducción de Señal
4.
Stem Cells ; 39(3): 306-317, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33295098

RESUMEN

Muscle stem cells, also called muscle satellite cells (MuSCs), are responsible for skeletal muscle regeneration and are sustained in an undifferentiated and quiescent state under steady conditions. The calcitonin receptor (CalcR)-protein kinase A (PKA)-Yes-associated protein 1 (Yap1) axis is one pathway that maintains quiescence in MuSCs. Although CalcR signaling in MuSCs has been identified, the critical CalcR signaling targets are incompletely understood. Here, we show the relevance between the ectopic expression of delta-like non-canonical Notch ligand 1 (Dlk1) and the impaired quiescent state in CalcR-conditional knockout (cKO) MuSCs. Dlk1 expression was rarely detected in both quiescent and proliferating MuSCs in control mice, whereas Dlk1 expression was remarkably increased in CalcR-cKO MuSCs at both the mRNA and protein levels. It is noteworthy that all Ki67+ non-quiescent CalcR-cKO MuSCs express Dlk1, and non-quiescent CalcR-cKO MuSCs are enriched in the Dlk1+ fraction by cell sorting. Using mutant mice, we demonstrated that PKA-activation or Yap1-depletion suppressed Dlk1 expression in CalcR-cKO MuSCs, which suggests that the CalcR-PKA-Yap1 axis inhibits the expression of Dlk1 in quiescent MuSCs. Moreover, the loss of Dlk1 rescued the quiescent state in CalcR-cKO MuSCs, which indicates that the ectopic expression of Dlk1 disturbs quiescence in CalcR-cKO. Collectively, our results suggest that ectopically expressed Dlk1 is responsible for the impaired quiescence in CalcR-cKO MuSCs.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Músculo Esquelético/metabolismo , Receptores de Calcitonina/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Proliferación Celular/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre/metabolismo
5.
EMBO Rep ; 20(3)2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30622218

RESUMEN

Promoter-associated long non-coding RNAs (lncRNAs) regulate the expression of adjacent genes; however, precise roles of these lncRNAs in skeletal muscle remain largely unknown. Here, we characterize a promoter-associated lncRNA, Myoparr, in myogenic differentiation and muscle disorders. Myoparr is expressed from the promoter region of the mouse and human myogenin gene, one of the key myogenic transcription factors. We show that Myoparr is essential both for the specification of myoblasts by activating neighboring myogenin expression and for myoblast cell cycle withdrawal by activating myogenic microRNA expression. Mechanistically, Myoparr interacts with Ddx17, a transcriptional coactivator of MyoD, and regulates the association between Ddx17 and the histone acetyltransferase PCAF Myoparr also promotes skeletal muscle atrophy caused by denervation, and knockdown of Myoparr rescues muscle wasting in mice. Our findings demonstrate that Myoparr is a novel key regulator of muscle development and suggest that Myoparr is a potential therapeutic target for neurogenic atrophy in humans.


Asunto(s)
Diferenciación Celular/genética , Desarrollo de Músculos/genética , Miogenina/genética , Regiones Promotoras Genéticas , ARN Largo no Codificante/metabolismo , Animales , Ciclo Celular , Línea Celular , ARN Helicasas DEAD-box/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , Modelos Biológicos , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/patología , Proteína MioD/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Unión Proteica , ARN Largo no Codificante/genética , Factor de Crecimiento Transformador beta/metabolismo , Factores de Transcripción p300-CBP/metabolismo
6.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638584

RESUMEN

Skeletal muscle is a vital organ for a healthy life, but its mass and function decline with aging, resulting in a condition termed sarcopenia. The etiology of sarcopenia remains unclear. We recently demonstrated that interstitial mesenchymal progenitors are essential for homeostatic muscle maintenance, and a diminished expression of the mesenchymal-specific gene Bmp3b is associated with sarcopenia. Here, we assessed the protective function of Bmp3b against sarcopenia by generating conditional transgenic (Tg) mice that enable a forced expression of Bmp3b specifically in mesenchymal progenitors. The mice were grown until they reached the geriatric stage, and the age-related muscle phenotypes were examined. The Tg mice had significantly heavier muscles compared to control mice, and the type IIB myofiber cross-sectional areas were preserved in Tg mice. The composition of the myofiber types did not differ between the genotypes. The Tg mice showed a decreasing trend of fibrosis, but the degree of fat infiltration was as low as that in the control mice. Finally, we observed the preservation of innervated neuromuscular junctions (NMJs) in the Tg muscle in contrast to the control muscle, where the NMJ degeneration was conspicuous. Thus, our results indicate that the transgenic expression of Bmp3b in mesenchymal progenitors alleviates age-related muscle deterioration. Collectively, this study strengthens the beneficial role of mesenchymal Bmp3b against sarcopenia and suggests that preserving the youthfulness of mesenchymal progenitors may be an effective means of combating sarcopenia.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Unión Neuromuscular/metabolismo , Envejecimiento/metabolismo , Animales , Factor 10 de Diferenciación de Crecimiento/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/metabolismo , Sarcopenia/metabolismo
7.
Int J Mol Sci ; 22(22)2021 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-34830237

RESUMEN

The main function of skeletal muscles is to generate force. The force developed by myofiber contraction is transmitted to the tendon. There are two pathways of force transmission from myofibers to tendons: longitudinal transmission that depends on tension elicited via the myotendinous junction and lateral transmission that depends on shear elicited via the interface between the myofiber surface and surrounding connective tissue. Experiments using animal muscle and mathematical models indicated that lateral transmission is the dominant pathway in muscle force transmission. Studies using rat muscle showed that the efficiency of lateral force transmission declines with age. Here, the lateral transmission of force was measured using the extensor digitorum longus muscle from young and old mice. Dependence on longitudinal transmission increased in the old muscle, and there was a trend for lower efficiency of lateral force transmission in the old muscle compared to the young muscle. There was a noticeable increase in the connective tissue volume in the old muscle; however, there was no significant change in the expression of dystrophin, a critical molecule for the link between the myofiber cytoskeleton and extracellular matrix. This study demonstrates the measurement of lateral force transmission in mouse muscles and that alteration in force transmission property may underlie age-related muscle weakness.


Asunto(s)
Envejecimiento/fisiología , Contracción Muscular/fisiología , Músculo Esquelético/fisiología , Sarcopenia/fisiopatología , Tendones/fisiología , Factores de Edad , Animales , Fenómenos Biomecánicos , Colágeno Tipo III/genética , Colágeno Tipo III/metabolismo , Distrofina/genética , Distrofina/metabolismo , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Miotomía/métodos , Sarcopenia/genética , Sarcopenia/metabolismo , Tenotomía/métodos
8.
J Cell Physiol ; 234(2): 1689-1698, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30070693

RESUMEN

Nuclear factor erythroid 2-related factor 2 (Nrf2) is a master regulator for the induction of antioxidative genes and plays roles in diverse cellular functions. The roles of Nrf2 in muscle regeneration have been investigated, and both important and unimportant roles of Nrf2 for muscle regeneration have been reported. Here, using aged Nrf2-null and Nrf2-dystrophic double-null mice, we showed nonsignificant phenotypes in the muscle regeneration ability of Nrf2-null mice. In contrast with these results, strikingly, almost all Nrf2-null muscle stem cells (MuSCs) isolated by fluorescence-activated cell sorting died in vitro of apoptosis and were not rescued by antioxidative reagents. Although their proliferation was still impaired, the Nrf2-null MuSCs attached to myofibers activated and divided normally, at least in the first round. To elucidate these discrepancies of MuSCs behaviors, we focused on the basal lamina, because both in vivo and single myofiber culture allow MuSCs within the basal lamina to become activated. In a basal lamina-disrupted model, Nrf2-null mice exhibited remarkable regeneration defects without increased levels of reactive oxidative species in MuSCs, suggesting that the existence of the basal lamina affects the survival of Nrf2-null MuSCs. Taken together, these results suggest that the basal lamina compensates for the loss of Nrf2, independent of the antioxidative roles of Nrf2. In addition, experimental conditions might explain the discrepant results of Nrf2-null regenerative ability.


Asunto(s)
Membrana Basal/metabolismo , Desarrollo de Músculos , Músculo Esquelético/metabolismo , Factor 2 Relacionado con NF-E2/deficiencia , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Células Satélite del Músculo Esquelético/metabolismo , Animales , Apoptosis , Técnicas de Cultivo de Célula , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Colágeno/metabolismo , Combinación de Medicamentos , Laminina/metabolismo , Ratones Endogámicos mdx , Ratones Noqueados , Músculo Esquelético/patología , Factor 2 Relacionado con NF-E2/genética , Proteoglicanos/metabolismo , Regeneración , Células Satélite del Músculo Esquelético/patología , Transducción de Señal
9.
Int J Mol Sci ; 20(13)2019 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-31277245

RESUMEN

Delta like non-canonical Notch ligand 1 (Dlk1) is a paternally expressed gene which is also known as preadipocyte factor 1 (Pref-1). The accumulation of adipocytes and expression of Dlk1 in regenerating muscle suggests a correlation between fat accumulation and Dlk1 expression in the muscle. Additionally, mice overexpressing Dlk1 show increased muscle weight, while Dlk1-null mice exhibit decreased body weight and muscle mass, indicating that Dlk1 is a critical factor in regulating skeletal muscle mass during development. The muscle regeneration process shares some features with muscle development. However, the role of Dlk1 in regeneration processes remains controversial. Here, we show that mesenchymal progenitors also known as adipocyte progenitors exclusively express Dlk1 during muscle regeneration. Eliminating developmental effects, we used conditional depletion models to examine the specific roles of Dlk1 in muscle stem cells or mesenchymal progenitors. Unexpectedly, deletion of Dlk1 in neither the muscle stem cells nor the mesenchymal progenitors affected the regenerative ability of skeletal muscle. In addition, fat accumulation was not increased by the loss of Dlk1. Collectively, Dlk1 plays essential roles in muscle development, but does not greatly impact regeneration processes and adipogenic differentiation in adult skeletal muscle regeneration.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Músculo Esquelético/fisiología , Regeneración , Células Madre/metabolismo , Animales , Proteínas de Unión al Calcio/fisiología , Ratones , Ratones Noqueados , Desarrollo de Músculos , Células Madre/fisiología
10.
Development ; 142(1): 51-61, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25480916

RESUMEN

Muscle satellite cells are indispensable for muscle regeneration, but the functional diversity of their daughter cells is unknown. Here, we show that many Pax7(+)MyoD(-) cells locate both beneath and outside the basal lamina during myofiber maturation. A large majority of these Pax7(+)MyoD(-) cells are not self-renewed satellite cells, but have different potentials for both proliferation and differentiation from Pax7(+)MyoD(+) myoblasts (classical daughter cells), and are specifically marked by expression of the doublecortin (Dcx) gene. Transplantation and lineage-tracing experiments demonstrated that Dcx-expressing cells originate from quiescent satellite cells and that the microenvironment induces Dcx in myoblasts. Expression of Dcx seems to be necessary for myofiber maturation because Dcx-deficient mice exhibited impaired myofiber maturation resulting from a decrease in the number of myonuclei. Furthermore, in vitro and in vivo studies suggest that one function of Dcx in myogenic cells is acceleration of cell motility. These results indicate that Dcx is a new marker for the Pax7(+)MyoD(-) subpopulation, which contributes to myofiber maturation during muscle regeneration.


Asunto(s)
Diferenciación Celular , Proteínas Asociadas a Microtúbulos/metabolismo , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/fisiología , Neuropéptidos/metabolismo , Regeneración/fisiología , Células Madre/citología , Animales , Cardiotoxinas/administración & dosificación , Movimiento Celular , Microambiente Celular , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Proteína MioD/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Neuropéptidos/deficiencia , Factor de Transcripción PAX7/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Madre/metabolismo
11.
Am J Pathol ; 187(12): 2627-2634, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28919111

RESUMEN

Fatty degeneration of skeletal muscle leads to muscle weakness and loss of function. Preventing fatty degeneration in skeletal muscle is important, but no drug has been used clinically. In this study, we performed drug repositioning using human platelet-derived growth factor receptor α (PDGFRα)-positive mesenchymal progenitors that have been proved to be an origin of ectopic adipocytes in skeletal muscle. We found that promethazine hydrochloride (PH) inhibits adipogenesis in a dose-dependent manner without cell toxicity. PH inhibited expression of adipogenic markers and also suppressed phosphorylation of cAMP response-element binding protein, which was reported to be a primary regulator of adipogenesis. We established a mouse model of tendon rupture with intramuscular fat deposition and confirmed that emerged ectopic adipocytes are derived from PDGFRα+ cells using lineage tracing mice. When these injured mice were treated with PH, formation of ectopic adipocytes was suppressed significantly. Our results show that PH inhibits PDGFRα+ mesenchymal progenitor-dependent ectopic adipogenesis in skeletal muscle and suggest that treatment with PH can be a promising approach to prevent fatty degeneration of skeletal muscle.


Asunto(s)
Adipocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Antagonistas de los Receptores Histamínicos H1/farmacología , Músculo Esquelético/patología , Prometazina/farmacología , Adipocitos/patología , Adipogénesis/efectos de los fármacos , Animales , Reposicionamiento de Medicamentos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Derivado de Plaquetas/metabolismo
12.
Am J Pathol ; 187(12): 2674-2685, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28919112

RESUMEN

Muscle dysfunction is the most important modifiable mediating factor in primary osteoarthritis (OA) because properly contracting muscles are a key absorber of forces acting on a joint. However, the pathological features of disuse muscle atrophy in OA patients have been rarely studied. Vastus medialis muscles of 14 female patients with OA (age range, 69 to 86 years), largely immobile for 1 or more years, were obtained during arthroplastic surgery and analyzed histologically. These were compared with female patients without arthritis, two with patellar fracture and two with patellar subluxation. Areas occupied by myofibers and adipose tissue were quantified. Large numbers of myofibers were lost in the vastus medialis of OA patients. The loss of myofibers was a possible cause of the reduction in muscle strength of the operated on knee. These changes were significantly correlated with an increase in intramuscular ectopic adipose tissue, and not observed in knees of nonarthritic patients. Resident platelet-derived growth factor receptor α-positive mesenchymal progenitor cells contributed to ectopic adipogenesis in vastus medialis muscles of OA patients. The present study suggests that significant loss of myofibers and ectopic adipogenesis in vastus medialis muscles are common pathological features of advanced knee OA patients with long-term loss of mobility. These changes may be related to the loss of joint function in patients with knee OA.


Asunto(s)
Tejido Adiposo , Coristoma/patología , Trastornos Musculares Atróficos/patología , Osteoartritis/complicaciones , Músculo Cuádriceps/patología , Adipogénesis/fisiología , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Trastornos Musculares Atróficos/etiología
13.
Stem Cells ; 33(8): 2456-68, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25917344

RESUMEN

Sarcopenia, age-related muscle weakness, increases the frequency of falls and fractures in elderly people, which can trigger severe muscle injury. Rapid and successful recovery from muscle injury is essential not to cause further frailty and loss of independence. In fact, we showed insufficient muscle regeneration in aged mice. Although the number of satellite cells, muscle stem cells, decreases with age, the remaining satellite cells maintain the myogenic capacity equivalent to young mice. Transplantation of young green fluorescent protein (GFP)-Tg mice-derived satellite cells into young and aged mice revealed that age-related deterioration of the muscle environment contributes to the decline in regenerative capacity of satellite cells. Thus, extrinsic changes rather than intrinsic changes in satellite cells appear to be a major determinant of inefficient muscle regeneration with age. Comprehensive protein expression analysis identified a decrease in insulin-like growth factor-II (IGF-II) level in regenerating muscle of aged mice. We found that pro- and big-IGF-II but not mature IGF-II specifically express during muscle regeneration and the expressions are not only delayed but also decreased in absolute quantity with age. Supplementation of pro-IGF-II in aged mice ameliorated the inefficient regenerative response by promoting proliferation of satellite cells, angiogenesis, and suppressing adipogenic differentiation of platelet derived growth factor receptor (PDGFR)α(+) mesenchymal progenitors. We further revealed that pro-IGF-II but not mature IGF-II specifically inhibits the pathological adipogenesis of PDGFRα(+) cells. Together, these results uncovered a distinctive pro-IGF-II-mediated self-reinforcement mechanism of muscle regeneration and suggest that supplementation of pro-IGF-II could be one of the most effective therapeutic approaches for muscle injury in elderly people.


Asunto(s)
Envejecimiento/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Músculo Esquelético/fisiología , Precursores de Proteínas/metabolismo , Regeneración/fisiología , Células Satélite del Músculo Esquelético/metabolismo , Envejecimiento/genética , Animales , Factor II del Crecimiento Similar a la Insulina/genética , Ratones , Ratones Noqueados , Precursores de Proteínas/genética
14.
Nihon Rinsho ; 74(9): 1554-1559, 2016 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-30557492

RESUMEN

Aging is inevitable for all living things. The average life span of Japanese women is over 86 years, and that of Japanese men exceeds 80 years. However, many people cannot live without help and care from other people for nearly ten years. Musculoskeletal system such as muscles, bones and joints is also affected by aging. Atrophy of skeletal muscle by aging impairs physical activity. Sarcopenia means muscle loss and is a condition characterized by low skeletal muscle mass and low muscle strength in older people. Physical activity is also affected. Sarcopenia is related with physical disability, poor quality of life and lethality. Sar- copenia is becoming one of major problems not only in Japan but also in many developed countries.


Asunto(s)
Envejecimiento , Sarcopenia , Anciano , Anciano de 80 o más Años , Envejecimiento/fisiología , Femenino , Humanos , Japón , Masculino , Fuerza Muscular , Músculo Esquelético , Calidad de Vida , Sarcopenia/complicaciones
15.
Development ; 138(21): 4609-19, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21989910

RESUMEN

Satellite cells, which are skeletal muscle stem cells, divide to provide new myonuclei to growing muscle fibers during postnatal development, and then are maintained in an undifferentiated quiescent state in adult skeletal muscle. This state is considered to be essential for the maintenance of satellite cells, but their molecular regulation is unknown. We show that Hesr1 (Hey1) and Hesr3 (Heyl) (which are known Notch target genes) are expressed simultaneously in skeletal muscle only in satellite cells. In Hesr1 and Hesr3 single-knockout mice, no obvious abnormalities of satellite cells or muscle regenerative potentials are observed. However, the generation of undifferentiated quiescent satellite cells is impaired during postnatal development in Hesr1/3 double-knockout mice. As a result, myogenic (MyoD and myogenin) and proliferative (Ki67) proteins are expressed in adult satellite cells. Consistent with the in vivo results, Hesr1/3-null myoblasts generate very few Pax7(+) MyoD(-) undifferentiated cells in vitro. Furthermore, the satellite cell number gradually decreases in Hesr1/3 double-knockout mice even after it has stabilized in control mice, and an age-dependent regeneration defect is observed. In vivo results suggest that premature differentiation, but not cell death, is the reason for the reduced number of satellite cells in Hesr1/3 double-knockout mice. These results indicate that Hesr1 and Hesr3 are essential for the generation of adult satellite cells and for the maintenance of skeletal muscle homeostasis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/fisiología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biomarcadores/metabolismo , Peso Corporal , Recuento de Células , Proteínas de Ciclo Celular/genética , Proliferación Celular , Células Cultivadas , Ratones , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Tamaño de los Órganos , Fenotipo
16.
Cell Rep ; 43(4): 114052, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38573860

RESUMEN

Skeletal muscles exert remarkable regenerative or adaptive capacities in response to injuries or mechanical loads. However, the cellular networks underlying muscle adaptation are poorly understood compared to those underlying muscle regeneration. We employed single-cell RNA sequencing to investigate the gene expression patterns and cellular networks activated in overloaded muscles and compared these results with those observed in regenerating muscles. The cellular composition of the 4-day overloaded muscle, when macrophage infiltration peaked, closely resembled that of the 10-day regenerating muscle. In addition to the mesenchymal progenitor-muscle satellite cell (MuSC) axis, interactome analyses or targeted depletion experiments revealed communications between mesenchymal progenitors-macrophages and macrophages-MuSCs. Furthermore, granulin, a macrophage-derived factor, inhibited MuSC differentiation, and Granulin-knockout mice exhibited blunted muscle hypertrophy due to the premature differentiation of overloaded MuSCs. These findings reveal the critical role of granulin through the relayed communications of mesenchymal progenitors, macrophages, and MuSCs in facilitating efficient muscle hypertrophy.


Asunto(s)
Diferenciación Celular , Hipertrofia , Macrófagos , Células Madre Mesenquimatosas , Ratones Noqueados , Células Satélite del Músculo Esquelético , Animales , Células Satélite del Músculo Esquelético/metabolismo , Células Satélite del Músculo Esquelético/patología , Macrófagos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Granulinas , Comunicación Celular , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Masculino , Regeneración
17.
J Cachexia Sarcopenia Muscle ; 15(3): 907-918, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38533539

RESUMEN

BACKGROUND: Recent studies have indicated the importance of muscle quality in addition to muscle quantity in sarcopenia pathophysiology. Intramuscular adipose tissue (IMAT), which originates from mesenchymal progenitors (MPs) in adult skeletal muscle, is a key factor affecting muscle quality in older adults, suggesting that controlling IMAT formation is a promising therapeutic strategy for sarcopenia. However, the molecular mechanism underlying IMAT formation in older adults has not been clarified. We recently found that the vitamin D receptor (VDR) is highly expressed in MPs in comparison to myotubes (P = 0.028, N = 3), indicating a potential role of vitamin D signalling in MPs. In this study, we aimed to clarify the role of vitamin D signalling in MP kinetics, with a focus on adipogenesis. METHODS: MPs isolated from mouse skeletal muscles were subjected to adipogenic differentiation conditions with or without vitamin D (1α,25(OH)2D3, 100 nM) for 7 days, and adipogenicity was evaluated based on adipogenic marker expression. For in vivo analysis, tamoxifen-inducible MP-specific VDR-deficient (VdrMPcKO) mice were newly developed to investigate whether lack of vitamin D signalling in MPs is involved in IMAT formation. To induce muscle atrophy, VdrMPcKO male mice were subjected to tenotomy of the gastrocnemius muscle, and then muscle weight, myofibre cross-sectional area, adipogenic marker expression, and fatty infiltration into the muscle were evaluated at 3 weeks after operation (N = 3-4). In addition, a vitamin D-deficient diet was provided to wild-type male mice (3 and 20 months of age, N = 5) for 3 months to investigate whether vitamin D deficiency causes IMAT formation. RESULTS: Vitamin D treatment nearly completely inhibited adipogenesis of MPs through Runx1-mediated transcriptional modifications of early adipogenic factors such as PPARγ (P = 0.0031) and C/EBPα (P = 0.0027), whereas VDR-deficient MPs derived from VdrMPcKO mice differentiated into adipocytes even in the presence of vitamin D (P = 0.0044, Oil-Red O+ area). In consistency with in-vitro findings, VdrMPcKO mice and mice fed a vitamin D-deficient diet exhibited fat deposition in atrophied (P = 0.0311) and aged (P = 0.0216) skeletal muscle, respectively. CONCLUSIONS: Vitamin D signalling is important to prevent fate decision of MPs towards the adipogenic lineage. As vitamin D levels decline with age, our data indicate that decreased vitamin D levels may be one of the causes of IMAT formation in older adults, and vitamin D signalling may be a novel therapeutic target for sarcopenia.


Asunto(s)
Células Madre Mesenquimatosas , Músculo Esquelético , Receptores de Calcitriol , Transducción de Señal , Vitamina D , Animales , Ratones , Vitamina D/metabolismo , Vitamina D/farmacología , Células Madre Mesenquimatosas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Masculino , Receptores de Calcitriol/metabolismo , Tejido Adiposo/metabolismo , Adipogénesis , Modelos Animales de Enfermedad , Diferenciación Celular
18.
J Cell Sci ; 124(Pt 21): 3654-64, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045730

RESUMEN

Accumulation of adipocytes and collagen type-I-producing cells (fibrosis) is observed in muscular dystrophies. The origin of these cells had been largely unknown, but recently we identified mesenchymal progenitors positive for platelet-derived growth factor receptor alpha (PDGFRα) as the origin of adipocytes in skeletal muscle. However, the origin of muscle fibrosis remains largely unknown. In this study, clonal analyses show that PDGFRα(+) cells also differentiate into collagen type-I-producing cells. In fact, PDGFRα(+) cells accumulated in fibrotic areas of the diaphragm in the mdx mouse, a model of Duchenne muscular dystrophy. Furthermore, mRNA of fibrosis markers was expressed exclusively in the PDGFRα(+) cell fraction in the mdx diaphragm. Importantly, TGF-ß isoforms, known as potent profibrotic cytokines, induced expression of markers of fibrosis in PDGFRα(+) cells but not in myogenic cells. Transplantation studies revealed that fibrogenic PDGFRα(+) cells mainly derived from pre-existing PDGFRα(+) cells and that the contribution of PDGFRα(-) cells and circulating cells was limited. These results indicate that mesenchymal progenitors are the main origin of not only fat accumulation but also fibrosis in skeletal muscle.


Asunto(s)
Adipogénesis , Fibrosis/fisiopatología , Células Madre Mesenquimatosas/citología , Músculo Esquelético/citología , Adipocitos/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Fibrosis/genética , Fibrosis/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
20.
Methods Mol Biol ; 2640: 117-127, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36995591

RESUMEN

Mesenchymal progenitors, which are resident progenitor populations residing in skeletal muscle interstitial space, contribute to pathogeneses such as fat infiltration, fibrosis, and heterotopic ossification. In addition to their pathological roles, mesenchymal progenitors have also been shown to play important roles for successful muscle regeneration and homeostatic muscle maintenance. Therefore, detailed and accurate analyses of these progenitors are essential for the research on muscle diseases and health. Here, we describe a method for purification of mesenchymal progenitors based on the expression of PDGFRα, which is a specific and well-established marker for mesenchymal progenitors, using fluorescence-activated cell sorting (FACS). Purified cells can be used in several downstream experiments including cell culture, cell transplantation, and gene expression analysis. We also describe the method for whole-mount 3-dimensional imaging of mesenchymal progenitors by utilizing tissue clearing. The methods described herein provide a powerful platform for studying mesenchymal progenitors in skeletal muscle.


Asunto(s)
Músculo Esquelético , Osificación Heterotópica , Humanos , Citometría de Flujo/métodos , Separación Celular/métodos , Diferenciación Celular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA