Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Bioorg Med Chem Lett ; 109: 129849, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38876177

RESUMO

Clinical studies have shown that inhibitors of bromodomain and extra-terminal domain (BET) proteins, particularly BRD4, have antitumor activity and efficacy. The BET protein has two domains, BD1 and BD2, and we previously focused on BD1 and reported orally bioavailable BD1-selective inhibitors. In this study, we obtained a BD1 inhibitor, a more potent and highly selective pyrazolopyridone derivative 13a, and confirmed its in vivo efficacy.


Assuntos
Piridonas , Humanos , Administração Oral , Relação Estrutura-Atividade , Animais , Piridonas/química , Piridonas/farmacologia , Piridonas/síntese química , Piridonas/farmacocinética , Pirazóis/química , Pirazóis/farmacologia , Pirazóis/síntese química , Descoberta de Drogas , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Estrutura Molecular , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Camundongos , Domínios Proteicos , Relação Dose-Resposta a Droga , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/síntese química , Ratos , Proteínas que Contêm Bromodomínio
2.
Bioorg Med Chem Lett ; 109: 129848, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38876176

RESUMO

We explored novel immunosuppressive agents with immune tolerance using a phenotypic drug discovery strategy, focusing on costimulatory molecules in T cells, and obtained triazolothienodiazepine derivatives. Their mechanism of action is to inhibit the bromodomain and extra-terminal domain (BET) family, as we have previously reported. Selective inhibition of the first bromodomain (BD1) of the BET family is expected to exert antitumor and immunosuppressive effects, similar to BET inhibitors. This study identified furopyridine derivatives 7 and 8 with high BD1 inhibitory activity and high selectivity over BD2. Compound 7 was found to be orally bioavailable and exhibited anti-inflammatory activity in a lipopolysaccharide-induced model.


Assuntos
Piridinas , Piridinas/química , Piridinas/farmacologia , Piridinas/síntese química , Animais , Humanos , Administração Oral , Relação Estrutura-Atividade , Camundongos , Descoberta de Drogas , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/antagonistas & inibidores , Estrutura Molecular , Ratos , Domínios Proteicos
3.
J Cell Physiol ; 235(3): 2300-2309, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31489965

RESUMO

Niemann-Pick disease type C (NPC) is a genetic disorder in which patient cells have endosomal/lysosomal accumulation of cholesterol and sphingolipids. However, the relationship between sphingolipids and cholesterol accumulation in NPC cells has not been established. Here, we investigated the role of sphingomyelin (SM) on the accumulation of cholesterol in NPC cells. Reduction of SM by inhibition of the ceramide transfer protein CERT decreased the cholesterol accumulation in NPC cells. The accumulation of SM in NPC cells inhibited the transport of cholesterol to the endoplasmic reticulum. Overexpression of Rab9 in NPC cells reduced the cholesterol accumulation, which was recovered by treatment with SM. In NPC cells that overexpressed a Rab9 constitutively active mutant, SM treatment did not lead to the cholesterol accumulation. These results indicate that SM negatively regulates the Rab9-dependent vesicular trafficking of cholesterol, and a reduction in SM levels in NPC cells recovers the Rab9-dependent vesicular trafficking defect.


Assuntos
Transporte Biológico/fisiologia , Colesterol/metabolismo , Doença de Niemann-Pick Tipo C/metabolismo , Esfingomielinas/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Células CHO , Proteínas de Transporte/metabolismo , Linhagem Celular , Cricetulus , Retículo Endoplasmático/metabolismo , Glicoproteínas de Membrana/metabolismo , Transporte Proteico/fisiologia , Esfingolipídeos
4.
PLoS Genet ; 12(7): e1006167, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27415617

RESUMO

Cell differentiation status is defined by the gene expression profile, which is coordinately controlled by epigenetic mechanisms. Cell type-specific DNA methylation patterns are established by chromatin modifiers including de novo DNA methyltransferases, such as Dnmt3a and Dnmt3b. Since the discovery of the myogenic master gene MyoD, myogenic differentiation has been utilized as a model system to study tissue differentiation. Although knowledge about myogenic gene networks is accumulating, there is only a limited understanding of how DNA methylation controls the myogenic gene program. With an aim to elucidate the role of DNA methylation in muscle development and regeneration, we investigate the consequences of mutating Dnmt3a in muscle precursor cells in mice. Pax3 promoter-driven Dnmt3a-conditional knockout (cKO) mice exhibit decreased organ mass in the skeletal muscles, and attenuated regeneration after cardiotoxin-induced muscle injury. In addition, Dnmt3a-null satellite cells (SCs) exhibit a striking loss of proliferation in culture. Transcriptome analysis reveals dysregulated expression of p57Kip2, a member of the Cip/Kip family of cyclin-dependent kinase inhibitors (CDKIs), in the Dnmt3a-KO SCs. Moreover, RNAi-mediated depletion of p57Kip2 replenishes the proliferation activity of the SCs, thus establishing a role for the Dnmt3a-p57Kip2 axis in the regulation of SC proliferation. Consistent with these findings, Dnmt3a-cKO muscles exhibit fewer Pax7+ SCs, which show increased expression of p57Kip2 protein. Thus, Dnmt3a is found to maintain muscle homeostasis by epigenetically regulating the proliferation of SCs through p57Kip2.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Desenvolvimento Muscular , Músculo Esquelético/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Metilação de DNA , DNA Metiltransferase 3A , Epigênese Genética , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Proteína MyoD/metabolismo , Mioblastos/metabolismo , Fator de Transcrição PAX7/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , Regeneração , Tomografia Computadorizada por Raios X
5.
Development ; 142(1): 51-61, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25480916

RESUMO

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.


Assuntos
Diferenciação Celular , Proteínas Associadas aos Microtúbulos/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/fisiologia , Neuropeptídeos/metabolismo , Regeneração/fisiologia , Células-Tronco/citologia , Animais , Cardiotoxinas/administração & dosagem , Movimento Celular , Microambiente Celular , Proteínas do Domínio Duplacortina , Proteína Duplacortina , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/deficiência , Proteína MyoD/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Neuropeptídeos/deficiência , Fator de Transcrição PAX7/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células-Tronco/metabolismo
6.
Am J Pathol ; 187(12): 2674-2685, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28919112

RESUMO

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.


Assuntos
Tecido Adiposo , Coristoma/patologia , Transtornos Musculares Atróficos/patologia , Osteoartrite/complicações , Músculo Quadríceps/patologia , Adipogenia/fisiologia , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Transtornos Musculares Atróficos/etiologia
7.
Nucleic Acids Res ; 44(21): e158, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27566152

RESUMO

Myoblasts are precursor skeletal muscle cells that differentiate into fused, multinucleated myotubes. Current single-cell microfluidic methods are not optimized for capturing very large, multinucleated cells such as myotubes. To circumvent the problem, we performed single-nucleus transcriptome analysis. Using immortalized human myoblasts, we performed RNA-seq analysis of single cells (scRNA-seq) and single nuclei (snRNA-seq) and found them comparable, with a distinct enrichment for long non-coding RNAs (lncRNAs) in snRNA-seq. We then compared snRNA-seq of myoblasts before and after differentiation. We observed the presence of mononucleated cells (MNCs) that remained unfused and analyzed separately from multi-nucleated myotubes. We found that while the transcriptome profiles of myoblast and myotube nuclei are relatively homogeneous, MNC nuclei exhibited significant heterogeneity, with the majority of them adopting a distinct mesenchymal state. Primary transcripts for microRNAs (miRNAs) that participate in skeletal muscle differentiation were among the most differentially expressed lncRNAs, which we validated using NanoString. Our study demonstrates that snRNA-seq provides reliable transcriptome quantification for cells that are otherwise not amenable to current single-cell platforms. Our results further indicate that snRNA-seq has unique advantage in capturing nucleus-enriched lncRNAs and miRNA precursors that are useful in mapping and monitoring differential miRNA expression during cellular differentiation.


Assuntos
Diferenciação Celular/genética , Mioblastos/citologia , Análise de Sequência de RNA/métodos , Linhagem Celular , Núcleo Celular/genética , Regulação da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , MicroRNAs/genética , Fibras Musculares Esqueléticas/citologia , Mioblastos/fisiologia , Fator Regulador Miogênico 5/genética , RNA Longo não Codificante , Análise de Célula Única/métodos
8.
Int J Urol ; 24(6): 461-467, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28332237

RESUMO

OBJECTIVES: To examine the inhibitory effects of tumor necrosis factor-α on myogenic differentiation of human urethral rhabdosphincter cells. METHODS: A rhabdosphincter sample was obtained from a patient who underwent total cystectomy. To expand the lifespan of the primary cultured cells, rhabdosphincter myogenic cells were immortalized with mutated cyclin-dependent kinase 4, cyclin D1 and telomerase. The differential potential of the cells was investigated. The transfected human rhabdosphincter cells were induced for myogenic differentiation with recombinant human tumor necrosis factor-α and/or the tumor necrosis factor-α antagonist etanercept at different concentrations, and activation of signaling pathways was monitored. RESULTS: Human rhabdosphincter cells were selectively cultured for at least 40 passages. Molecular analysis confirmed the expression of myosin heavy chain, which is a specific marker of differentiated muscle cells, significantly increased after differentiation induction. Although tumor necrosis factor-α treatment reduced the myosin heavy chain expression in a concentration-dependent manner, etanercept inhibited this suppression. Tumor necrosis factor-α suppressed phosphorylation of protein kinase B and p38, whereas etanercept pretreatment promoted phosphorylation and myosin heavy chain expression in a concentration-dependent manner. CONCLUSIONS: Tumor necrosis factor-α inhibits differentiation of urethral rhabdosphincter cells in part through the p38 mitogen-activated protein kinase and phosphoinositide 3-kinase pathways. Inhibition of tumor necrosis factor-α might be a useful strategy to treat stress urinary incontinence.


Assuntos
Diferenciação Celular/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Mioblastos Esqueléticos/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cistectomia , Etanercepte/farmacologia , Etanercepte/uso terapêutico , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Desenvolvimento Muscular/efeitos dos fármacos , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/citologia , Mioblastos Esqueléticos/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Cultura Primária de Células , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/isolamento & purificação , Uretra/citologia , Uretra/efeitos dos fármacos , Uretra/fisiologia , Neoplasias da Bexiga Urinária/cirurgia , Incontinência Urinária por Estresse/tratamento farmacológico , Incontinência Urinária por Estresse/fisiopatologia
9.
Stem Cells ; 33(8): 2456-68, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25917344

RESUMO

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.


Assuntos
Envelhecimento/metabolismo , Fator de Crescimento Insulin-Like II/metabolismo , Músculo Esquelético/fisiologia , Precursores de Proteínas/metabolismo , Regeneração/fisiologia , Células Satélites de Músculo Esquelético/metabolismo , Envelhecimento/genética , Animais , Fator de Crescimento Insulin-Like II/genética , Camundongos , Camundongos Knockout , Precursores de Proteínas/genética
10.
Int J Med Sci ; 13(9): 680-5, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27647997

RESUMO

Effects of myostatin (MSTN)-suppression on the regeneration of injured skeletal muscle under unloading condition were investigated by using transgenic mice expressing a dominant-negative form of MSTN (MSTN-DN). Both MSTN-DN and wild-type (WT) mice were subjected to continuous hindlimb suspension (HS) for 6 weeks. Cardiotoxin (CTX) was injected into left soleus muscle under anesthesia 2 weeks after the initiation of HS. Then, the soleus muscles were excised following 6-week HS (4 weeks after CTX-injection). CTX-injection caused to reduce the soleus fiber cross-sectional area (CSA) in WT mice under both unloading and weight-bearing conditions, but not in MSTN-DN mice. Under unloading condition, CTX-injected muscle weight and fiber CSA in MSTN-DN mice were significantly higher than those in WT mice. CTX-injected muscle had many damaged and regenerating fibers having central nuclei in both WT and MSTN-DN mice. Significant increase in the population of Pax7-positive nuclei in CTX-injected muscle was observed in MSTN-DN mice, but not in WT mice. Evidences indicate that the suppression of MSTN cause to increase the regenerative potential of injured soleus muscle via the increase in the population of muscle satellite cells regardless of unloading conditions.


Assuntos
Membro Posterior/crescimento & desenvolvimento , Músculo Esquelético/crescimento & desenvolvimento , Miostatina/biossíntese , Regeneração , Animais , Cardiotoxinas/administração & dosagem , Membro Posterior/efeitos dos fármacos , Membro Posterior/lesões , Membro Posterior/fisiopatologia , Humanos , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Músculo Esquelético/fisiopatologia , Miostatina/antagonistas & inibidores , Células Satélites de Músculo Esquelético/metabolismo , Células Satélites de Músculo Esquelético/patologia , Suporte de Carga
11.
J Cell Biochem ; 116(9): 1898-907, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25716287

RESUMO

We examined the effect of the cellular sphingolipid level on the release of arachidonic acid (AA) and the activity of secretory phospholipase A2 (sPLA2 ) using two Chinese hamster ovary (CHO)-K1 cell mutants, LY-B and LY-A cells, deficient in sphingolipid synthesis. In LY-B cells, deficiency of sphingolipids enhanced the release of AA induced by bee venom sPLA2-III or human sPLA2-V. These alterations were reversed by replenishment of exogenous sphingomyelin (SM). In LY-A cells, deficiency of SM increased the release of AA induced by sPLA2. In CHO-K1 cells, decrease and increase of SM level in the plasma membrane by pharmacological methods increased and inhibited the release of AA, respectively. SM inhibited the activity of sPLA2 in vitro. Niemann-Pick disease type C (NPC) is a lysosomal storage disorder caused by mutation of either the NPC1 or NPC2 gene, and is characterized by accumulation of cholesterol and sphingolipids including SM in late endosomes/lysosomes. Increased levels of AA and sPLA2 activity are involved in various neurodegenerative diseases. In CHO cells lacking NPC1 (A101 cells), SM level was lower in the plasma membrane, while it was higher in late endosomes/lysosomes. The release of AA induced by sPLA2 was increased in A101 cells than that in parental cells (JP17 cells), which was attenuated by adding exogenous SM. In addition, sPLA2 -III-induced cytotoxicity in A101 cells was much higher than that in JP17 cells. These results suggest that SM in the plasma membrane plays important roles in regulating sPLA2 activity and the enzyme-induced cytotoxicity in A101 cells.


Assuntos
Ácido Araquidônico/biossíntese , Membrana Celular/metabolismo , Doença de Niemann-Pick Tipo C/enzimologia , Fosfolipases A2 Secretórias/metabolismo , Esfingomielinas/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Fosfolipases A2 do Grupo III/metabolismo , Fosfolipases A2 do Grupo III/farmacologia , Fosfolipases A2 do Grupo V/metabolismo , Fosfolipases A2 do Grupo V/farmacologia , Humanos , Glicoproteínas de Membrana/deficiência , Modelos Biológicos , Fosfolipases A2 Secretórias/farmacologia , Esfingomielinas/deficiência
12.
Hum Mutat ; 35(8): 998-1010, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24838473

RESUMO

Facioscapulohumeral dystrophy (FSHD) is one of the most prevalent muscular dystrophies. The majority of FSHD cases are linked to a decreased copy number of D4Z4 macrosatellite repeats on chromosome 4q (FSHD1). Less than 5% of FSHD cases have no repeat contraction (FSHD2), most of which are associated with mutations of SMCHD1. FSHD is associated with the transcriptional derepression of DUX4 encoded within the D4Z4 repeat, and SMCHD1 contributes to its regulation. We previously found that the loss of heterochromatin mark (i.e., histone H3 lysine 9 tri-methylation (H3K9me3)) at D4Z4 is a hallmark of both FSHD1 and FSHD2. However, whether this loss contributes to DUX4 expression was unknown. Furthermore, additional D4Z4 homologs exist on multiple chromosomes, but they are largely uncharacterized and their relationship to 4q/10q D4Z4 was undetermined. We found that the suppression of H3K9me3 results in displacement of SMCHD1 at D4Z4 and increases DUX4 expression in myoblasts. The DUX4 open reading frame (ORF) is disrupted in D4Z4 homologs and their heterochromatin is unchanged in FSHD. The results indicate the significance of D4Z4 heterochromatin in DUX4 gene regulation and reveal the genetic and epigenetic distinction between 4q/10q D4Z4 and the non-4q/10q homologs, highlighting the special role of the 4q/10q D4Z4 chromatin and the DUX4 ORF in FSHD.


Assuntos
DNA Satélite , Epigênese Genética , Heterocromatina/metabolismo , Proteínas de Homeodomínio/genética , Distrofia Muscular Facioescapuloumeral/genética , Mutação , Animais , Sequência de Bases , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Cromossomos Humanos Par 10 , Cromossomos Humanos Par 4 , Cricetinae , Expressão Gênica , Histonas/genética , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Distrofia Muscular Facioescapuloumeral/metabolismo , Distrofia Muscular Facioescapuloumeral/patologia , Mioblastos/metabolismo , Mioblastos/patologia , Fases de Leitura Aberta , Cultura Primária de Células , Homologia de Sequência do Ácido Nucleico
13.
BMC Cell Biol ; 14: 37, 2013 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-23978243

RESUMO

BACKGROUND: Previous research indicates that the membrane ruffles and leading edge of lamellipodia of myogenic cells contain presumptive fusion sites. A micrometer-sized lipid raft (microraft) is organized at the presumptive fusion site of mouse myogenic cells in a cell-contact independent way and serves as a platform tethering adhesion proteins that are relevant to cell fusion. However, the mechanisms underlying recruitment of adhesion proteins to lipid rafts and microraft organization remain unknown. RESULTS: Here we show that small G-protein Rac1 was required for microraft organization and subsequent cell fusion. However, Rac1 activity was unnecessary for recruitment of M-cadherin to lipid rafts. We found that p120 catenin (p120) binds to M-cadherin exclusively in lipid rafts of differentiating myogenic cells. The Src kinase inhibitor SU6656 prevented p120 binding to M-cadherin and their recruitment to lipid rafts, then suppressed microraft organization, membrane ruffling, and myogenic cell fusion. Suppression of membrane ruffling in SU6656-treated cells was partially restored by pretreatment with the protein tyrosine phosphatase inhibitor vanadate. The present analyses using an antibody to tyrosine phosphorylated p120 suggest that Src family kinases play a role in binding of p120 to M-cadherin and the recruitment of M-cadherin to lipid rafts through phosphorylation of putative substrates other than p120. CONCLUSIONS: The present study showed that the procedure establishing fusion-competent sites consists of two sequential events: recruitment of adhesion complexes to lipid rafts and organization of microrafts. The recruitment of M-cadherin to lipid rafts depended on interaction with p120 catenin, whereas the organization of microrafts was controlled by a small G protein, Rac1.


Assuntos
Caderinas/genética , Regulação da Expressão Gênica , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/metabolismo , Transdução de Sinais/genética , Animais , Caderinas/metabolismo , Cateninas/genética , Cateninas/metabolismo , Adesão Celular/efeitos dos fármacos , Fusão Celular , Células Cultivadas , Indóis/farmacologia , Microdomínios da Membrana/efeitos dos fármacos , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Fosforilação , Ligação Proteica , Transporte Proteico , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Sulfonamidas/farmacologia , Vanadatos/farmacologia , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo , delta Catenina
15.
Exp Cell Res ; 317(2): 221-33, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20965167

RESUMO

A high concentration of bone morphogenetic proteins (BMPs) stimulates myogenic progenitor cells to undergo heterotopic osteogenic differentiation. However, the physiological role of the Smad signaling pathway during terminal muscle differentiation has not been resolved. We report here that Smad1/5/8 was phosphorylated and activated in undifferentiated growing mouse myogenic progenitor Ric10 cells without exposure to any exogenous BMPs. The amount of phosphorylated Smad1/5/8 was severely reduced during precocious myogenic differentiation under the high cell density culture condition even in growth medium supplemented with a high concentration of serum. Inhibition of the Smad signaling pathway by dorsomorphin, an inhibitor of Smad activation, or noggin, a specific antagonist of BMP, induced precocious terminal differentiation of myogenic progenitor cells in a cell density-dependent fashion even in growth medium. In addition, Smad1/5/8 was transiently activated in proliferating myogenic progenitor cells during muscle regeneration in rats. The present results indicate that the Smad signaling pathway is involved in a critical switch between growth and differentiation of myogenic progenitor cells both in vitro and in vivo. Furthermore, precocious cell density-dependent myogenic differentiation suggests that a community effect triggers the terminal muscle differentiation of myogenic cells by quenching the Smad signaling.


Assuntos
Diferenciação Celular/fisiologia , Mioblastos/metabolismo , Células Satélites de Músculo Esquelético/fisiologia , Transdução de Sinais/fisiologia , Proteínas Smad/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Proteínas Morfogenéticas Ósseas/fisiologia , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Camundongos , Camundongos Endogâmicos ICR , Células Musculares/citologia , Células Musculares/metabolismo , Células Musculares/fisiologia , Fosforilação , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
16.
Exp Cell Res ; 315(17): 3052-63, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19615358

RESUMO

Recent research indicates that the leading edge of lamellipodia of myogenic cells (myoblasts and myotubes) contains presumptive fusion sites, yet the mechanisms that render the plasma membrane fusion-competent remain largely unknown. Here we show that dynamic clustering and dispersion of lipid rafts contribute to both cell adhesion and plasma membrane union during myogenic cell fusion. Adhesion-complex proteins including M-cadherin, beta-catenin, and p120-catenin accumulated at the leading edge of lamellipodia, which contains the presumptive fusion sites of the plasma membrane, in a lipid raft-dependent fashion prior to cell contact. In addition, disruption of lipid rafts by cholesterol depletion directly prevented the membrane union of myogenic cell fusion. Time-lapse recording showed that lipid rafts were laterally dispersed from the center of the lamellipodia prior to membrane fusion. Adhesion proteins that had accumulated at lipid rafts were also removed from the presumptive fusion sites when lipid rafts were laterally dispersed. The resultant lipid raft- and adhesion complex-free area at the leading edge fused with the opposing plasma membrane. These results demonstrate a key role for dynamic clustering/dispersion of lipid rafts in establishing fusion-competent sites of the myogenic cell membrane, providing a novel mechanistic insight into the regulation of myogenic cell fusion.


Assuntos
Microdomínios da Membrana/fisiologia , Fibras Musculares Esqueléticas/fisiologia , Mioblastos/fisiologia , Animais , Anticorpos Monoclonais , Técnicas de Cultura de Células/métodos , Fusão Celular/métodos , Células Clonais , Feminino , Imunofluorescência , Genes Reporter , Proteínas de Fluorescência Verde/genética , Imuno-Histoquímica , Microdomínios da Membrana/ultraestrutura , Camundongos , Camundongos Endogâmicos ICR , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Cadeias Pesadas de Miosina/análise , Cadeias Pesadas de Miosina/imunologia
18.
Commun Biol ; 3(1): 182, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32313117

RESUMO

Understanding the signaling pathways that regulate proliferation and differentiation of muscle progenitors is essential for successful cell transplantation for treatment of Duchenne muscular dystrophy. Here, we report that a γ-secretase inhibitor, DAPT (N-[N-(3,5-difluorophenacetyl-L-alanyl)]-S-phenylglycine tertial butyl ester), which inhibits the release of NICD (Notch intercellular domain), promotes the fusion of human muscle progenitors in vitro and improves their engraftment in the tibialis anterior muscle of immune-deficient mice. Gene expression analysis revealed that DAPT severely down-regulates PTGER2, which encodes prostaglandin (PG) E2 receptor 2 (EP2), in human muscle progenitors in the differentiation condition. Functional analysis suggested that Notch signaling inhibits differentiation and promotes self-renewal of human muscle progenitors via PGE2/EP2 signaling in a cAMP/PKA-independent manner.


Assuntos
Diferenciação Celular , Proliferação de Células , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Receptor Notch3/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Fusão Celular , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Autorrenovação Celular , Sobrevivência Celular , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Dipeptídeos/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos Endogâmicos mdx , Camundongos SCID , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/lesões , Músculo Esquelético/patologia , Músculo Esquelético/cirurgia , Mioblastos Esqueléticos/efeitos dos fármacos , Mioblastos Esqueléticos/transplante , Receptor Notch3/genética , Receptores de Prostaglandina E Subtipo EP2/genética , Transdução de Sinais , Transplante de Células-Tronco
19.
PLoS One ; 12(7): e0181457, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28715470

RESUMO

Alendronate, a nitrogen-containing bisphosphonate, is well established as a treatment for osteoporosis through regulation of osteoclast activity. Previously, the pharmacological effects of bisphosphonates on cells outside the bone environment have been considered irrelevant because bisphosphonates target bone. Here we show that administration of alendronate impairs muscle regeneration in mice after bone fracture. A series of injections of alendronate alone or bone fracture alone did not affect muscle regeneration induced by cold injury. In contrast, alendronate treatment plus bone fracture severely impaired the regeneration of muscle that closely contacts the bone fracture site after cold injury. After cold injury, M-cadherin-positive myogenic cells disappeared in the damaged muscle areas of mice receiving the combination of alendronate treatment and bone fracture. The present results suggest that the muscle regeneration capacity is impaired by bone fracture in mice receiving alendronate treatment. The present research on the pharmacological effects of alendronate on muscle regeneration will aid in understanding of the in vivo action of alendronate on skeletal muscles.


Assuntos
Alendronato/farmacologia , Conservadores da Densidade Óssea/farmacologia , Lesão por Frio/fisiopatologia , Fraturas Ósseas/terapia , Músculo Esquelético/fisiopatologia , Regeneração/fisiologia , Alendronato/efeitos adversos , Animais , Conservadores da Densidade Óssea/efeitos adversos , Lesão por Frio/patologia , Lesão por Frio/terapia , Modelos Animais de Doenças , Fraturas Ósseas/patologia , Fraturas Ósseas/fisiopatologia , Masculino , Camundongos Endogâmicos ICR , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Regeneração/efeitos dos fármacos
20.
PLoS One ; 12(10): e0186584, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29049386

RESUMO

Previous studies have shown that phenotypic modulation of smooth muscle cells (SMCs) plays a pivotal role in human diseases. However, the molecular mechanisms underlying the reversible differentiation of SMCs remain elusive particularly because cultured SMCs that reproducibly exhibit bidirectional phenotypic modulation have not been established. Here we established an immortalized human bladder SMC line designated as hBS11. Under differentiation-inducing conditions, hBS11 cells underwent smooth muscle differentiation accompanied by the robust expression of smooth muscle differentiation markers and isoform-dependent reorganization of actin bundles. The cholinergic receptor agonist carbachol increased intracellular calcium in differentiated hBS11 cells in an acetylcholine muscarinic receptor-dependent manner. Differentiated hBS11 cells displayed contractile properties depending on the elevation in the levels of intracellular calcium. Depolarization of membrane potential triggered inward sodium current in differentiated hBS11 cells. However, differentiated hBS11 cells lost the differentiated phenotype and resumed mitosis when re-fed with growth medium. Our study provides direct evidence pertaining to the human bladder SMCs being able to retain the capacity of reversible differentiation and that the reorganization of actin bundles is involved in the reinstatement of contractility. Moreover, we have established a human SMC line retaining high proliferating potential without compromising differentiation potential.


Assuntos
Actinas/metabolismo , Diferenciação Celular , Músculo Liso/citologia , Bexiga Urinária/citologia , Linhagem Celular Transformada , Humanos , Músculo Liso/metabolismo , Bexiga Urinária/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA