Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Sci Food Agric ; 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38551359

RESUMO

BACKGROUND: Leucine (Leu) is an essential amino acid that facilitates skeletal muscle satellite cell differentiation, yet its mechanism remains underexplored. Sestrin2 (SESN2) serves as a Leu sensor, binding directly to Leu, while ribophorin II (RPN2) acts as a signaling factor in multiple pathways. This study aimed to elucidate Leu's impact on mouse C2C12 cell differentiation and skeletal muscle injury repair by modulating RPN2 expression through SESN2, offering a theoretical foundation for clinical skeletal muscle injury prevention and treatment. RESULTS: Leu addition promoted C2C12 cell differentiation compared to the control, enhancing early differentiation via myogenic determinant (MYOD) up-regulation. Sequencing revealed SESN2 binding to and interacting with RPN2. RPN2 overexpression up-regulated MYOD, myogenin and myosin heavy chain 2, concurrently decreased p-GSK3ß and increased nuclear ß-catenin. Conversely, RPN2 knockdown yielded opposite results. Combining RPN2 knockdown with Leu rescued increased p-GSK3ß and decreased nuclear ß-catenin compared to Leu absence. Hematoxylin and eosin staining results showed that Leu addition accelerated mouse muscle damage repair, up-regulating Pax7, MYOD and RPN2 in the cytoplasm, and nuclear ß-catenin, confirming that the role of Leu in muscle injury repair was consistent with the results for C2C12 cells. CONCLUSION: Leu, bound with SESN2, up-regulated RPN2 expression, activated the GSK3ß/ß-catenin pathway, enhanced C2C12 differentiation and expedited skeletal muscle damage repair. © 2024 Society of Chemical Industry.

2.
FASEB J ; 38(5): e23502, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38430223

RESUMO

Podocan, the fifth member of Small Leucine-Rich Proteoglycan (SLRP) family of extracellular matrix components, is poorly known in muscle development. Previous studies have shown that Podocan promotes C2C12 differentiation in mice. In this study, we elucidated the effect of Podocan on skeletal muscle post-injury regeneration and its underlying mechanism. Injection of Podocan protein promoted the process of mice skeletal muscle post-injury regeneration. This effect seemed to be from the acceleration of muscle satellite cell differentiation in vivo. Meanwhile, Podocan promoted myogenic differentiation in vitro by binding with TGF-ß1 to inhibit the activity of the TGF-ß signaling pathway. These results indicated that Podocan had the potential roles to enhance skeletal muscle post-injury regeneration. Its mechanism is likely the regulation of the expression of p-Smad2 and p-Smad4 related to the TGF-ß signaling pathway by interacting with TGF-ß1.


Assuntos
Músculo Esquelético , Proteínas , Regeneração , Fator de Crescimento Transformador beta1 , Animais , Camundongos , Diferenciação Celular , Músculo Esquelético/lesões , Músculo Esquelético/fisiologia , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Proteínas/metabolismo
3.
Food Funct ; 13(24): 12721-12732, 2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36408829

RESUMO

Cyanocobalamin (CNCbl, the compound name of Vitamin B12) is the only mineral vitamin that is essential for growth and development and cannot be produced by animals. Some studies have found that CNCbl can promote the proliferation and migration of C2C12 cells, but the mechanism by which it affects muscle development is still unknown. In this study, we elucidated the effect of CNCbl on muscle development and studied its underlying mechanism. CNCbl could promote the differentiation of C2C12 cells and upregulate Acvr1, p-Smad2 and p-Smad3 in the TGF-ß signaling pathway in vitro. CD320 (the receptor in cell surface for binding with CNCbl transporter transcobalamin II) inhibition could reduce the uptake of CNCbl and significantly downregulate the expression of differentiation marker proteins MyoG and MYH2. Furthermore, the levels of p-Smad2 and p-Smad3 were also reduced with the inhibition of CD320, even though CNCbl was added to the C2C12 culture medium. In addition, the injection of CNCbl could accelerate the process of mouse muscle injury repair, enlarge the diameter of newly formed myofibers and upregulate the expression of MYH2, PAX7, CD320, Acvr1, p-Smad2 and p-Smad3 in vivo. These results suggest that CNCbl can promote muscle development and may play its role by regulating the expression of Acvr1, p-Smad2 and p-Smad3 related to the TGF-ß signaling pathway.


Assuntos
Desenvolvimento Muscular , Fator de Crescimento Transformador beta , Vitamina B 12 , Animais , Camundongos , Diferenciação Celular , Desenvolvimento Muscular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Vitamina B 12/farmacologia , Linhagem Celular
4.
J Anim Sci ; 100(11)2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-35797991

RESUMO

Skeletal muscle is composed of muscle fibers formed from myoblast differentiation. Recently, numerous researchers have demonstrated that microRNAs (miRNAs) play an essential role in modulating the proliferation and differentiation of myoblasts. Our previous study has shown that among the miR-17-92 cluster members, miR-17 and miR-20a together with miR-19b can efficiently promote the differentiation of murine C2C12 and bovine primary myoblasts. However, the role of miR-18 in this process remains elusive. In this study, we revealed that miR-18 inhibited the differentiation of bovine skeletal muscle-derived satellite cells (bMDSCs), whereas an miR-18 inhibitor significantly promoted cell differentiation (p < 0.001). Then, a target gene of miR-18 was found to be myocyte enhancer factor 2D (MEF2D), which is critical for myoblast differentiation. Furthermore, we found that the combination of the miR-18 inhibitor and miR-19 significantly improved the formation of bMDSCs-derived muscle fibers (p < 0.001). This study revealed the role of miR-18 in bovine skeletal muscle differentiation and contributed to the understanding of the regulatory mechanism of mammalian myogenic differentiation.


Beef is a beneficial food source, and improving muscle yield and quality has become a hot topic in the beef industry. Therefore, our study aimed to explore effective methods to improve bovine muscle cell differentiation to increase beef production. The study revealed that microRNA-18 (miR-18) inhibitor could promote the differentiation of bovine skeletal muscle-derived satellite cells (bMDSCs) by increasing the expression of myocyte enhancer factor 2D (MEF2D), a critical gene for myoblast differentiation. Furthermore, we found that combined inhibitors of miR-18 and miR-19 could significantly improve bMDSCs differentiation. Our study demonstrated the role of a new regulatory factor that may enhance beef production level and contributed to elucidating the mechanism of muscle differentiation.


Assuntos
MicroRNAs , Células Satélites de Músculo Esquelético , Animais , Bovinos , Diferenciação Celular , Proliferação de Células/genética , Mamíferos/genética , Mamíferos/metabolismo , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Desenvolvimento Muscular/genética , Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/metabolismo
5.
J Agric Food Chem ; 70(23): 7158-7169, 2022 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-35652451

RESUMO

Previous studies have reported that vitamin C (VC), an essential nutrient, exerts beneficial effects on muscle health. However, the molecular mechanism involved in the VC-mediated regulation of muscle development is still unclear. The roles of VC in muscle development and the underlying molecular mechanisms were examined using cell and molecular biology, transcriptomics, proteomics, and animal experiments in this study. VC upregulated the expression of sodium-dependent vitamin C transporter 2 (SVCT2) and cysteine rich protein 3 (CSRP3). Additionally, VC promoted the differentiation of C2C12 cells and the repair of mouse muscle injury by upregulating the nuclear translocation of CSRP3, which subsequently interacted with MyoD and MyoG. This study provided a theoretical basis for elucidating the mechanism underlying the VC-mediated regulation of muscle development, as well as for developing animal nutritional supplements and therapeutic drugs for muscle diseases.


Assuntos
Ácido Ascórbico , Desenvolvimento Muscular , Animais , Ácido Ascórbico/farmacologia , Diferenciação Celular , Camundongos , Músculos , Vitaminas
6.
Biology (Basel) ; 10(7)2021 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-34356541

RESUMO

Myocilin (MYOC) is a glycoprotein encoded by a gene associated with glaucoma pathology. In addition to the eyes, it also expresses at high transcription levels in the heart and skeletal muscle. MYOC affects the formation of the murine gastrocnemius muscle and is associated with the differentiation of mouse osteoblasts, but its role in the differentiation of C2C12 cells has not yet been reported. Here, MYOC expression was found to increase gradually during the differentiation of C2C12 cells. Overexpression of MYOC resulted in enhanced differentiation of C2C12 cells while its inhibition caused reduced differentiation. Furthermore, immunoprecipitation indicated that MYOC binds to Caveolin-1 (CAV1), a protein that influences the TGF-ß pathway. Laser confocal microscopy also revealed the common sites of action of the two during the differentiation of C2C12 cells. Additionally, CAV1 was upregulated significantly as C2C12 cells differentiated, with CAV1 able to influence the differentiation of the cells. Furthermore, the Western blotting analysis demonstrated that the expression of MYOC affected the TGF-ß pathway. Finally, MYOC was overexpressed while CAV1 was inhibited. The results indicate that reduced CAV1 expression blocked the promotion of C2C12 cell differentiation by MYOC. In conclusion, the results demonstrated that MYOC regulates TGF-ß by influencing CAV1 to promote the differentiation of C2C12 cells.

7.
Cell Adh Migr ; 14(1): 153-164, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32881638

RESUMO

FNDC4 is an anti-inflammatory factor that alters the activation state of macrophages; it is used to treat colitis in mice. However, its role in muscle formation and mechanism of function remains unknown. We found that FNDC4 promotes the bovine MDSCs migration and differentiation. Furthermore, we reported that it interacts with integrin ß1 (ITGß1). FAK, mediated by ITGß1, regulates cell migration. Our results found FNDC4 to influence the expression of p-FAK, p-paxillin, and vinculin. Then, overexpressed or added FNDC4 protein could not influence migration and differentiation any more when the activated form of FAK was reduced. Therefore, we concluded that FNDC4 promotes the differentiation and migration of bovine MDSCs via the FAK, mediated by the ITGß1 receptor.


Assuntos
Diferenciação Celular , Movimento Celular , Domínio de Fibronectina Tipo III , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Proteínas/química , Proteínas/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Animais , Bovinos , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Integrina beta1/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Células Satélites de Músculo Esquelético/efeitos dos fármacos
8.
Animals (Basel) ; 10(8)2020 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-32781616

RESUMO

As an extracellular matrix protein, secreted protein acidic and rich in cysteine (SPARC)-like 1 (SPARCL1) is involved in various cell functions. It was previously implicated in bovine skeletal muscle-derived satellite cell (MDSC) differentiation; however, the underlying mechanism remains unknown. In this study, immunoprecipitation and mass spectrometry revealed that integrin ß1 (ITGB1) combines with SPARCL1. Further, co-immunoprecipitation demonstrated that SPARCL1 interacts with ITGB1. Cell scratch assays explored the influence of SPARCL1 on MDSC migration through ITGB1. In addition, desmin staining for myotube fusion rate and MyoD protein expression results showed that SPARCL1 promotes MDSC early differentiation through ITGB1. Furthermore, Western blotting results demonstrated that SPARCL1 regulates the expression of p-FAK, p-paxillin, vinculin, Cdc42, and Arp2/3 through ITGB1. These findings indicate that SPARCL1 may influence bovine MDSC migration and differentiation through an ITGB1-mediated cell signaling pathway. Herein, we elucidated the mechanism through which SPARCL1 affects MDSC differentiation. Our results provide insight into the molecular mechanism of muscle development and may in the future facilitate skeletal muscle regeneration and treatment.

9.
Mol Cell Biochem ; 470(1-2): 215-227, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32458119

RESUMO

Skeletal muscle is one of the most important tissues of the human body necessary for sporting activities. The differentiation of muscle-derived satellite cells (MDSCs) plays an important role in the development and regeneration of skeletal muscles. Similarly, the Wnt/ß-catenin signalling pathway plays an important role in the process of muscle differentiation. Wnt1-inducible signalling pathway protein-1 (WISP1), a downstream protein of the Wnt/ß-catenin signalling pathway and a member of the CCN family that also plays an important role in the differentiation process, and its expression increase during the differentiation of bovine MDSCs. However, its role in MDSC differentiation is poorly understood. Therefore, we investigated the mechanisms regulating this process via Western blot and immunofluorescence staining. Immunoprecipitation and mass spectrometry detected annexin A1 (ANXA1), a protein that interacts with WISP1. To determine whether WISP1 influences TGF-ß signalling and differentiation independently of ANXA1, the latter was knocked down, while WISP1 was activated. WISP1 expression increased significantly during bovine MDSC differentiation. However, WISP1 did not affect the TGF-ß signalling pathway protein marker when ANXA1 was inhibited. Taken together, WISP1 regulates the TGF-ß signalling pathway through ANXA1 recruitment, thereby promoting bovine MDSC differentiation, suggesting the Wnt/ß-catenin signalling pathway as another target to promote cell differentiation.


Assuntos
Anexina A1/metabolismo , Proteínas de Sinalização Intercelular CCN/metabolismo , Diferenciação Celular , Células Satélites de Músculo Esquelético/citologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Animais Recém-Nascidos , Bovinos , Regulação da Expressão Gênica , Regeneração , Via de Sinalização Wnt
10.
FASEB J ; 34(6): 7759-7772, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32298013

RESUMO

Fibronectin type III domain containing 4 (FNDC4) belongs to the fibronectin type III domain containing protein family. FNDC5, which is highly homologous to FNDC4, can promote the differentiation of cardiac cells. We aimed to investigate the role of FNDC4 in the differentiation of C2C12 mouse skeletal muscle cells. Western blotting and immunofluorescence analysis showed that FNDC4 gradually increased with the differentiation of C2C12. Muscle injury repair experiments indicated that FNDC4 may promote the repair of injured muscles. When FNDC4 was either overexpressed or knocked down, the expression of desmin and myogenin myogenic marker molecules followed that of FNDC4, suggesting that FNDC4 can influence the differentiation of C2C12. In addition, immunoprecipitation results showed that FNDC4 can interact with the Wnt/ß-catenin signaling pathway receptor low-density lipoprotein receptor-related protein 6 (LRP6), and that ß-catenin levels in the nucleus decreased after knocking down FNDC4. Exogenous addition of FNDC4 protein could not restore the blocking of differentiation due to inhibition of both Wnt/ß-catenin signal transduction and LRP6 activity via the ß-catenin inhibitor XAV-939. Overall, our findings indicate that FDNC4 can influence the differentiation of C2C12 by activating Wnt/ß-catenin signal transduction.


Assuntos
Diferenciação Celular/fisiologia , Domínio de Fibronectina Tipo III/fisiologia , Proteínas de Membrana/metabolismo , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo , Animais , Linhagem Celular , Camundongos , Células Musculares/metabolismo , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/metabolismo , Mioblastos/metabolismo
11.
J Cell Physiol ; 235(10): 7183-7193, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32017087

RESUMO

T-complex 11 like 2 (TCP11L2) is a protein containing a serine-rich region in its N-terminal region. However, the function of TCP11L2 is unclear. Here, we showed that TCP11L2 expression gradually increased during muscle-derived satellite cell (MDSC) differentiation in vitro, reaching a peak on Day 3, which is the migration and fusion stage of MDSCs. Using CRISPR/dCas9 gene-editing technology to elevate or repress the expression of TCP11L2, we also showed that TCP11L2 promoted MDSC differentiation. Moreover, wound-healing assays showed that TCP11L2 promoted the migration of MDSCs during differentiation. Additionally, immunofluorescence analyses showed that TCP11L2 was mainly distributed around the microfilament and microtubules. Furthermore, the expression of TCP11L2 affected the expression of actin-related protein 2/3 (ARP2/3) complex. Co-immunoprecipitation assays and immunofluorescence analysis showed that TCP11L2 interacted with formin-like 2 (FMNL2). This protein promoted migration of bovine MDSCs by affecting the expression of ARP2/3. Finally, the activities of TCP11L2 during MDSC differentiation and migration were blocked when FMNL2 was inhibited. Taken together, our data established that TCP11L2 interacted with FMNL2 to promote MDSC migration and differentiation.


Assuntos
Forminas/metabolismo , Proteínas Musculares/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Animais , Sistemas CRISPR-Cas , Bovinos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Movimento Celular/genética , Movimento Celular/fisiologia , Células Cultivadas , Camundongos , Desenvolvimento Muscular/genética , Desenvolvimento Muscular/fisiologia , Proteínas Musculares/química , Proteínas Musculares/genética , Células Satélites de Músculo Esquelético/citologia , Região do Complexo-t do Genoma
12.
Cell Death Dis ; 10(11): 852, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31699966

RESUMO

The extracellular matrix (ECM) is known to regulate tissue development and cell morphology, movement, and differentiation. SPARCL1 is an ECM protein, but its role in mouse cell differentiation has not been widely investigated. The results of western blotting and immunofluorescence showed that SPARCL1 is associated with the repair of muscle damage in mice and that SPARCL1 binds to bone morphogenetic protein 7 (BMP7) by regulating BMP/transforming growth factor (TGF)-ß cell signaling. This pathway promotes the differentiation of C2C12 cells. Using CRISPR/Cas9 technology, we also showed that SPARCL1 activates BMP/TGF-ß to promote the differentiation of C2C12 cells. BMP7 molecules were found to interact with SPARCL1 by immunoprecipitation analysis. Western blotting and immunofluorescence were performed to verify the effect of BMP7 on C2C12 cell differentiation. Furthermore, SPARCL1 was shown to influence the expression of BMP7 and activity of the BMP/TGF-ß signaling pathway. Finally, SPARCL1 activation was accompanied by BMP7 inhibition in C2C12 cells, which confirmed that SPARCL1 affects BMP7 expression and can promote C2C12 cell differentiation through the BMP/TGF-ß pathway. The ECM is essential for muscle regeneration and damage repair. This study intends to improve the understanding of the molecular mechanisms of muscle development and provide new treatment ideas for muscle injury diseases.


Assuntos
Proteína Morfogenética Óssea 7/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica , Músculo Esquelético/citologia , Mioblastos/citologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteína Morfogenética Óssea 7/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas da Matriz Extracelular/genética , Masculino , Camundongos , Camundongos Endogâmicos ICR , Músculo Esquelético/lesões , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/genética
13.
Front Physiol ; 10: 1010, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31447699

RESUMO

BACKGROUND: Small leucine-rich repeat proteins (SLRPs) are highly effective and selective modulators of cell proliferation and differentiation. Podocan is a newly discovered member of the SLRP family. Its potential roles in the differentiation of bovine muscle-derived satellite cells (MDSCs) and its underlying functional mechanism remain unclear. Our study aimed to characterize the function of the podocan gene in the differentiation of bovine MDSCs and to clarify the molecular mechanism by which podocan functions in order to contribute to a better understanding of the molecular mechanism by which extracellular matrix promotes bovine MDSC differentiation and provide a theoretical basis for the improvement of beef quality. METHODS: Bovine MDSCs were transfected with vectors to overexpress or inhibit podocan, and podocan protein was added to differentiation culture medium. qRT-PCR, western blotting, and immunofluorescence were performed to investigate the effects of podocan on MDSC differentiation. Confocal microscopy and western blotting were used to assess the nuclear translocation and expression of ß-catenin. An inhibitor and activator of ß-catenin were used to assess the effects of the Wnt/ß-catenin signaling pathway on MDSC differentiation. We inhibited ß-catenin while overexpressing podocan in MDSCs. Then, we performed mass spectrometry to identify which proteins interact with podocan to regulate the Wnt/ß-catenin signaling pathway. Finally, we confirmed the relationship between podocan and Wnt4 by co-immunoprecipitation and western blotting. RESULTS: Podocan protein expression increased significantly during bovine MDSC differentiation. Differentiation of bovine MDSC was promoted and suppressed by podocan overexpression or inhibition, respectively. Podocan was also shown to modulate the Wnt/ß-catenin signaling pathway. Treatment of bovine MDSCs with ß-catenin inhibitor and activator showed that the Wnt/ß-catenin pathway is involved in bovine MDSC differentiation. Furthermore, the effect of podocan on bovine MDSC differentiation was suppressed when this pathway was inhibited. We also found that podocan interacts with Wnt4. When Wnt4 was inhibited, podocan-induced promotion of bovine MDSC differentiation was attenuated through Wnt/ß-catenin signaling. CONCLUSION: Podocan regulates Wnt/ß-catenin through Wnt4 to promote bovine MDSC differentiation.

14.
Cell Mol Life Sci ; 76(24): 5041-5054, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31214725

RESUMO

Skeletal myogenesis is a highly coordinated process that involves cell proliferation, differentiation and fusion controlled by a complex gene regulatory network. The microRNA gene cluster miR-17-92 has been shown to be related to this process; however, the exact role of each cluster member remains unclear. Here, we show that miR-17 and miR-20a could effectively promote the differentiation of both C2C12 myoblasts and primary bovine satellite cells. In contrast, miR-18a might play a negative role in C2C12 cell differentiation, while miR-19 and miR-92a had little influence. Transcriptome and target analyses revealed that miR-17 could act on Ccnd2, Jak1 and Rhoc genes that are critical for cell proliferation and/or fusion. Notably, the addition of miR-19 could reverse the lethal effect of miR-17 and could thus facilitate the maturation of myotubes. Furthermore, by co-injecting the lentiviral shRNAs of miR-17 and miR-19 into mouse tibialis anterior muscles, we demonstrated the wound healing abilities of the two miRNAs. Our findings indicate that in combination with miR-19, miR-17 is a potent inducer of skeletal muscle differentiation.


Assuntos
Diferenciação Celular/genética , MicroRNAs/genética , Músculo Esquelético/crescimento & desenvolvimento , Animais , Bovinos , Proliferação de Células/genética , Ciclina D2/genética , Redes Reguladoras de Genes/genética , Janus Quinase 1/genética , Camundongos , Desenvolvimento Muscular/genética , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Proteína de Ligação a GTP rhoC/genética
15.
Cell Adh Migr ; 13(1): 192-202, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31096840

RESUMO

PEAR1 is highly expressed at bovine MDSC differentiation. However, its biological function remains unclear. Western blotting results showed that PEAR1 increased between day 0 and day 2 of cell differentiation and decreased from day 3. Moreover, scratch test showed that wound healing rate increased after PEAR1 overexpression and decreased upon its suppression. Meanwhile, we found that, upon PEAR1 induction, both the expression of the focal adhesion-associated and MyoG, and the myotube fusion rate increased. However, when PEAR1 was suppressed, opposite results were obtained. Immunoprecipitation revealed an association between PEAR1 and ITGB1. Notably, inhibition of FAK and ITGB1 repressed cell differentiation. In conclusion, our study indicated that PEAR1 is involved in the regulation of bovine MDSC migration and differentiation.


Assuntos
Diferenciação Celular , Movimento Celular , Quinase 1 de Adesão Focal/metabolismo , Integrina beta1/metabolismo , Mioblastos/fisiologia , Receptores de Superfície Celular/metabolismo , Células Satélites de Músculo Esquelético/fisiologia , Animais , Bovinos , Células Cultivadas , Quinase 1 de Adesão Focal/genética , Adesões Focais , Integrina beta1/genética , Mioblastos/citologia , Fosforilação , Agregação Plaquetária , Receptores de Superfície Celular/genética , Células Satélites de Músculo Esquelético/citologia , Transdução de Sinais
16.
Cell Biol Int ; 43(7): 799-808, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31050064

RESUMO

Muscle satellite cells are usually at rest, and when externally stimulated or regulated, they can be further differentiated by cell fusion to form new myotubes and muscle fibers. WD repeat domain 13 (WDR13) is highly conserved in vertebrates. Studies have shown that mice lacking the Wdr13 gene develop mild obesity, hyperinsulinemia, and increased islet ß cell proliferation. However, the role of WDR13 in bovine cells is unclear. Here, we investigated the effect of WDR13 on bovine skeletal muscle-derived satellite cells (MDSCs). We found that WDR13 was upregulated in bovine MDSCs using western blotting and immunofluorescence experiments. Moreover, activation and inhibition of WDR13 expression increased and decreased cell differentiation, respectively, suggesting that WDR13 promotes bovine MDSC differentiation. To further understand the mechanism of action of WDR13, we examined changes in the PI3K/AKT signaling pathway following WDR13 activation or inhibition. In addition, cells were treated with a phosphoinositide kinase 3 (PI3K) inhibitor, LY294004, to observe cell differentiation. The results showed that activation of WDR13 inhibited the PI3K/AKT signaling pathway and enhanced cell differentiation. These data suggest that WDR13 can promote the differentiation of bovine MDSCs by affecting the PI3K/AKT signaling pathway.


Assuntos
Diferenciação Celular , Desenvolvimento Muscular , Músculo Esquelético/crescimento & desenvolvimento , Proteínas Nucleares/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Satélites de Músculo Esquelético/citologia , Animais , Bovinos , Proteínas de Ciclo Celular , Proliferação de Células , Células Cultivadas , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Proteínas Nucleares/genética , Células Satélites de Músculo Esquelético/metabolismo , Transdução de Sinais
17.
J Cell Physiol ; 234(11): 21211-21223, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31025379

RESUMO

The glucose-regulated endoplasmic reticulum chaperone protein 94 (GRP94) is required for many biological processes, such as secretion of immune factors and mesoderm induction. Here, we demonstrated that GRP94 promotes muscle differentiation in vitro and in vivo. Moreover, GRP94 inhibited the PI3K/AKT/mTOR signaling pathway. Using both in vitro and in vivo approaches, in myoblasts, we found that this inhibition resulted in reduced proliferation and increased differentiation. To further investigate the mechanism of GRP94-induced muscle differentiation, we used co-immunoprecipitation and proximity ligation assays and found that GRP94 interacted with PI3K-interacting protein 1 (Pik3ip1). The latter protein promoted muscle differentiation by inhibiting the PI3K/AKT/mTOR pathway. Furthermore, GRP94 was found to regulate Pik3ip1 expression. Finally, when Pik3ip1 expression was inhibited, GRP94-induced promotion of muscle differentiation was diminished. Taken together, our data demonstrated that GRP94 promoted muscle differentiation, mediated by Pik3ip1-dependent inhibition of the PI3K/AKT/mTOR signaling pathway.


Assuntos
Glicoproteínas de Membrana/metabolismo , Células Musculares/metabolismo , Mioblastos/metabolismo , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/fisiologia , Camundongos , Células Musculares/citologia , Músculo Esquelético/metabolismo , Mioblastos/citologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo
18.
J Cell Physiol ; 234(7): 10554-10565, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30623413

RESUMO

TCEA3 is a member of the transcription elongation factor family that not only promotes transcription but may also participate in other cytoplasmic processes. However, its mechanisms of action remain unclear. Our previous study indicated that TCEA3 may affect muscle differentiation. In this study, we investigated the expression and localization of TCEA3 in C2C12 cells and examined the role of TCEA3 in differentiation, its interaction with other cell proteins, and mechanisms of action. We found that the expression of TCEA3 increased gradually with an increase in the number of differentiation days and that it is mainly expressed in the cytoplasm of C2C12 cells, of which it promotes differentiation. Coimmunoprecipitation experiments and western blot analysis revealed that TCEA3 interacts with Annexin A1 (ANXA1), which is located in the cytoplasm and also promotes cell differentiation. Collectively, our results indicate that TCEA3 promotes cell differentiation by interacting with ANXA1 and affecting transforming growth factor-ß signaling pathways.


Assuntos
Anexina A1/genética , Transcrição Gênica , Fatores de Elongação da Transcrição/genética , Fator de Crescimento Transformador beta/genética , Animais , Diferenciação Celular/genética , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos , Transdução de Sinais/genética
19.
J Cell Physiol ; 234(7): 11130-11139, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30652305

RESUMO

Podocan, a small leucine-rich repeat protein, is a negative regulator of cell proliferation. In this study, we demonstrated that podocan is involved in the differentiation of C2C12 murine myoblasts. Podocan expression increased with the progression of C2C12 differentiation. As expect, siRNA-mediated podocan knockdown inhibited C2C12 differentiation, as indicated by inhibition of MYOG, MYH2, and desmin expression, as well as reductions in the differentiation and fusion indices. Overexpression of podocan using dCas9 technology promoted C2C12 cell differentiation. In addition, supplementation of culture medium with podocan influenced C2C12 differentiation. Podocan knockdown reduced Wnt/ß-catenin signaling, characterized by a reduction in the nuclear translocation of ß-catenin, whereas podocan overexpression had the opposite effect. Furthermore, treatment with XAV939, an inhibitor of Wnt/ß-catenin, reduced the podocan-mediated promotion of C2C12 differentiation. Induction of muscle injury in mice by bupivacaine administration suggested that podocan may play a role in muscle regeneration. In summary, our results suggest that podocan is required for normal C2C12 differentiation and that its role in myogenesis is mediated by the Wnt/ß-catenin pathway.


Assuntos
Desenvolvimento Muscular/fisiologia , Mioblastos/fisiologia , Proteínas/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Diferenciação Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas/genética , RNA Guia de Cinetoplastídeos , beta Catenina/genética
20.
J Cell Physiol ; 234(1): 632-641, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-30078180

RESUMO

MicroRNAs play an important regulatory role in the proliferation and differentiation of skeletal muscle-derived satellite cells (MDSCs). In particular, miR-139 can inhibit tumor cell proliferation and invasion, and its expression is down-regulated during C2C12 myoblast differentiation. The aim of this study was thus to examine the effect and potential mechanism of miR-139 in bovine MDSCs. The expression of miR-139 was found to be significantly increased during bovine MDSC differentiation by stem-loop reverse transcription-polymerase chain reaction amplification. Statistical analysis of the myotube fusion rate was done through immunofluorescence detection of desmin, and western blotting was used to measure the change in protein expression of the muscle differentiation marker genes MYOG and MYH3. The results showed that the miR-139 mimic could enhance the differentiation of bovine MDSCs, whereas the inhibitor had the opposite effect. By using the dual-luciferase reporter system, miR-139 was found to target the 3'-untranslated region of the dihydrofolate reductase (DHFR) gene and regulate its expression. In addition, the expression of miR-139 was found to be regulated by its host gene phosphodiesterase 2A (PDE2A) via inhibition of the latter by CRISPR interference (CRISPRi). Overall, our findings indicate that miR-139 plays an important role in regulating the differentiation of bovine MDSCs.


Assuntos
MicroRNAs/genética , Músculo Esquelético/crescimento & desenvolvimento , Células Satélites de Músculo Esquelético/metabolismo , Tetra-Hidrofolato Desidrogenase/genética , Animais , Bovinos , Diferenciação Celular/genética , Proliferação de Células/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Desenvolvimento Muscular/genética , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA