Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
PLoS One ; 14(4): e0214982, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30964931

RESUMO

Disuse induces adaptations in skeletal muscle, which lead to muscle deterioration. Hindlimb-unloading (HU) is a well-established model to investigate cellular mechanisms responsible for disuse-induced skeletal muscle dysfunction. In myosin heavy chain (MHC) type IIB fibers HU induces a reduction in contraction speed (Vo) and a reduction in the relative myosin light chain 3f (MLC3f) protein content compared with myosin light chain 1f (MLC1f) protein. This study tested the hypothesis that increasing the relative MLC3f protein content via rAd-MLC3f vector delivery would attenuate the HU-induced decline in Vo in single MHC type IIB fibers. Fischer-344 rats were randomly assigned to one of three groups: control, HU for 7 days, and HU for 7 days plus rAd-MLC3f. The semimembranosus muscles were injected with rAd-MLC3f (3.75 x 1011-5 x 1011 ifu/ml) at four days after the initiation of HU. In single MHC type IIB fibers the relative MLC3f content decreased by 25% (12.00±0.60% to 9.06±0.66%) and Vo was reduced by 29% (3.22±0.14fl/s vs. 2.27±0.08fl/s) with HU compared to the control group. The rAd-MLC3f injection resulted in an increase in the relative MLC3f content (12.26±1.19%) and a concomitant increase in Vo (2.90±0.15fl/s) of MHC type IIB fibers. A positive relationship was observed between the percent of MLC3f content and Vo. Maximal isometric force and specific tension were reduced with HU by 49% (741.45±44.24µN to 379.09±23.77µN) and 33% (97.58±4.25kN/m2 to 65.05±2.71kN/m2), respectively compared to the control group. The rAd-MLC3f injection did not change the HU-induced decline in force or specific tension. Collectively, these results indicate that rAd-MLC3f injection rescues hindlimb unloading-induced decline in Vo in MHC type IIB single muscle fibers.


Assuntos
Adaptação Fisiológica , Contração Muscular , Fibras Musculares Esqueléticas/metabolismo , Transtornos Musculares Atróficos/prevenção & controle , Cadeias Leves de Miosina/biossíntese , Adenoviridae , Animais , Vetores Genéticos , Elevação dos Membros Posteriores , Masculino , Fibras Musculares Esqueléticas/patologia , Transtornos Musculares Atróficos/genética , Transtornos Musculares Atróficos/metabolismo , Cadeias Leves de Miosina/genética , Ratos , Ratos Endogâmicos F344 , Transdução Genética
2.
Basic Res Cardiol ; 112(1): 1, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27837311

RESUMO

Chronic increased workload of the human heart causes ventricular hypertrophy, re-expression of the atrial essential myosin light chain (hALC-1), and improved contractile function. Although hALC-1 is an important positive inotropic regulator of the human heart, little is known about its regulation. Therefore, we investigated the role of the sex hormone 17ß-estradiol (E2) on hALC-1 gene expression, the underlying molecular mechanisms, and the impact of this regulatory process on cardiac contractile function. We showed that E2 attenuated hALC-1 expression in human atrial tissues of both sexes and in human ventricular AC16 cells. E2 induced the nuclear translocation of estrogen receptor alpha (ERα) and hALC-1 in AC16 cells, where they cooperatively regulate the transcriptional activity of hALC-1 gene promoter. E2-activated ERα required the estrogen response element (ERE) motif within the hALC-1 gene promoter to reduce its transcriptional activity (vehicle: 15.55 ± 4.80 vs. E2: 6.51 ± 3.69; ~2 fold). This inhibitory effect was potentiated in the presence of hALC-1 (vehicle: 11.13 ± 3.66 vs. E2: 2.18 ± 1.10; ~5 fold), and thus, hALC-1 acts as a co-repressor of ERα-mediated transcription. Yeast two-hybrid screening of a human heart cDNA library revealed that ERα interacts physically with hALC-1 in the presence of E2. This interaction was confirmed by Co-Immunoprecipitation and immunofluorescence in human atrium. As a further novel effect, we showed that chronic E2-treatment of adult mouse cardiomyocytes overexpressing hALC-1 resulted in reduced cell-shortening amplitude and twitching kinetics of these cells independent of Ca2+ activation levels. Together, our data showed that the expression of hALC-1 gene is, at least partly, regulated by E2/ERα, while hALC-1 acts as a co-repressor. The inotropic effect of hALC-1 overexpression in cardiomyocytes can be significantly repressed by E2.


Assuntos
Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica/genética , Contração Miocárdica/fisiologia , Cadeias Leves de Miosina/biossíntese , Animais , Western Blotting , Feminino , Imunofluorescência , Humanos , Imunoprecipitação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Miócitos Cardíacos/metabolismo , Cadeias Leves de Miosina/genética , Reação em Cadeia da Polimerase , Técnicas do Sistema de Duplo-Híbrido
3.
Mol Med Rep ; 14(5): 4293-4300, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27748856

RESUMO

The role of myosin light chain II (MLC­II) in cellular differentiation of rat mandibular condylar chondrocytes (MCCs) induced by cyclical uniaxial compressive stress (CUCS) remains unclear. In the current study, a four­point bending system was used to apply CUCS to primary cultured MCCs from rats. It was identified that CUCS stimulated features of cellular differentiation including morphological alterations, cytoskeleton rearrangement and overproduction of proteoglycans. Furthermore, CUCS promoted runt­related transcription factor­2 (RUNX2) expression at mRNA (P<0.01) and protein levels (P<0.05) and elevated alkaline phosphatase (ALP) activity (P<0.01), which are both markers of osteogenic differentiation. Under conditions of stress, western blotting indicated that the ratio of phosphorylated MLC­II to total MLC­II was increased significantly (P<0.05). Silencing MLC­II by RNA interference reduced ALP activity (P<0.01), and eliminated RUNX2 mRNA expression (P<0.01). Addition of the MLC kinase inhibitor, ML­7, reduced the CUCS­associated upregulation of RUNX2 expression (P<0.01) and ALP activity (P<0.01). The data indicated that CUCS promoted cellular differentiation of rat primary MCCs, and this was suggested to be via the phosphorylation of MLC­II.


Assuntos
Miosinas Cardíacas/genética , Condrócitos/citologia , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Côndilo Mandibular/crescimento & desenvolvimento , Cadeias Leves de Miosina/genética , Osteogênese/genética , Fosfatase Alcalina/antagonistas & inibidores , Fosfatase Alcalina/genética , Animais , Azepinas/administração & dosagem , Miosinas Cardíacas/antagonistas & inibidores , Miosinas Cardíacas/biossíntese , Diferenciação Celular/genética , Condrócitos/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Côndilo Mandibular/citologia , Côndilo Mandibular/metabolismo , Cadeias Leves de Miosina/antagonistas & inibidores , Cadeias Leves de Miosina/biossíntese , Naftalenos/administração & dosagem , Fosforilação , Pressão , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos
4.
Biol Trace Elem Res ; 173(1): 116-25, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26779623

RESUMO

Selenium (Se), a nutritionally essential trace element, is associated with health and disease. Selenoprotein T (SelT) was identified as a redoxin protein with a selenocystein, localizing in the endoplasmic reticulum. The myosin light chain kinase (MLCK) and myosin light chain (MLC) play key roles in the contraction process of smooth muscle. The present study was to detect the effect and mechanism of SelT on the contraction process of gastric smooth muscle. The WT rats were fed with different Se concentration diets, and Se and Ca(2+) concentrations were detected in the gastric smooth muscle. Western blot and qPCR were performed to determine SelT, CaM, MLCK, and MLC expressions. MLCK activity was measured by identifying the rates of [γ-32P]ATP incorporated into the MLC. The results showed Se and Ca(2+) concentrations were enhanced with Se intake in gastric smooth muscle tissues. With increasing Se, SelT, CaM, MLCK and MLC expressions increased, and MLCK and MLC activation improved in gastric smooth muscle tissue. The SelT RNA interference experiments showed that Ca(2+) release, MLCK activation, and MLC phosphorylation were regulated by SelT. Se affected the gastric smooth muscle constriction by regulating Ca(2+) release, MLCK activation, and MLC phosphorylation through SelT. Se plays a major role in regulating the contraction processes of gastric smooth muscle with the SelT.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Mucosa Gástrica/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Músculo Liso/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Selênio/farmacologia , Selenoproteínas/biossíntese , Animais , Ativação Enzimática/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Cadeias Leves de Miosina/biossíntese , Ratos , Ratos Wistar
5.
Am J Physiol Heart Circ Physiol ; 309(9): H1509-15, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26386113

RESUMO

With the advent of high-throughput DNA sequencing, the number of identified cardiomyopathy-causing mutations has increased tremendously. As the majority of these mutations affect myofilament proteins, there is a need to understand their functional consequence on contraction. Permeabilized myofilament preparations coupled with protein exchange protocols are a common method for examining into contractile mechanics. However, producing large quantities of myofilament proteins can be time consuming and requires different approaches for each protein of interest. In the present study, we describe a unified automated method to produce troponin C, troponin T, and troponin I as well as myosin light chain 2 fused to a His6-tag followed by a tobacco etch virus (TEV) protease site. TEV protease has the advantage of a relaxed P1' cleavage site specificity, allowing for no residues left after proteolysis and preservation of the native sequence of the protein of interest. After expression in Esherichia coli, cells were lysed by sonication in imidazole-containing buffer. The His6-tagged protein was then purified using a HisTrap nickel metal affinity column, and the His6-tag was removed by His6-TEV protease digestion for 4 h at 30°C. The protease was then removed using a HisTrap column, and complex assembly was performed via column-assisted sequential desalting. This mostly automated method allows for the purification of protein in 1 day and can be adapted to most soluble proteins. It has the advantage of greatly increasing yield while reducing the time and cost of purification. Therefore, production and purification of mutant proteins can be accelerated and functional data collected in a faster, less expensive manner.


Assuntos
Miosinas Cardíacas/biossíntese , Endopeptidases , Histidina , Miofibrilas , Cadeias Leves de Miosina/biossíntese , Oligopeptídeos , Proteínas Recombinantes/biossíntese , Troponina C/biossíntese , Troponina I/biossíntese , Troponina T/biossíntese , Automação , Miosinas Cardíacas/genética , Cromatografia de Afinidade , Dextranos , Escherichia coli/genética , Humanos , Cadeias Leves de Miosina/genética , Níquel , Proteínas Recombinantes/genética , Sefarose , Troponina C/genética , Troponina I/genética , Troponina T/genética
6.
J Biol Chem ; 290(8): 4663-4676, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25538241

RESUMO

Cav1.3 L-type Ca(2+) channel is known to be highly expressed in neurons and neuroendocrine cells. However, we have previously demonstrated that the Cav1.3 channel is also expressed in atria and pacemaking cells in the heart. The significance of the tissue-specific expression of the channel is underpinned by our previous demonstration of atrial fibrillation in a Cav1.3 null mutant mouse model. Indeed, a recent study has confirmed the critical roles of Cav1.3 in the human heart (Baig, S. M., Koschak, A., Lieb, A., Gebhart, M., Dafinger, C., Nürnberg, G., Ali, A., Ahmad, I., Sinnegger-Brauns, M. J., Brandt, N., Engel, J., Mangoni, M. E., Farooq, M., Khan, H. U., Nürnberg, P., Striessnig, J., and Bolz, H. J. (2011) Nat. Neurosci. 14, 77-84). These studies suggest that detailed knowledge of Cav1.3 may have broad therapeutic ramifications in the treatment of cardiac arrhythmias. Here, we tested the hypothesis that there is a functional cross-talk between the Cav1.3 channel and a small conductance Ca(2+)-activated K(+) channel (SK2), which we have documented to be highly expressed in human and mouse atrial myocytes. Specifically, we tested the hypothesis that the C terminus of Cav1.3 may translocate to the nucleus where it functions as a transcriptional factor. Here, we reported for the first time that the C terminus of Cav1.3 translocates to the nucleus where it functions as a transcriptional regulator to modulate the function of Ca(2+)-activated K(+) channels in atrial myocytes. Nuclear translocation of the C-terminal domain of Cav1.3 is directly regulated by intracellular Ca(2+). Utilizing a Cav1.3 null mutant mouse model, we demonstrate that ablation of Cav1.3 results in a decrease in the protein expression of myosin light chain 2, which interacts and increases the membrane localization of SK2 channels.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Núcleo Celular/metabolismo , Regulação da Expressão Gênica/fisiologia , Miócitos Cardíacos/metabolismo , Transcrição Gênica/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Canais de Cálcio Tipo L/genética , Miosinas Cardíacas/biossíntese , Miosinas Cardíacas/genética , Núcleo Celular/genética , Átrios do Coração/citologia , Átrios do Coração/metabolismo , Humanos , Camundongos , Camundongos Knockout , Miócitos Cardíacos/citologia , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Estrutura Terciária de Proteína
7.
Circ Cardiovasc Genet ; 7(5): 667-76, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25077947

RESUMO

BACKGROUND: Nuclear reprogramming inculcates pluripotent capacity by which de novo tissue differentiation is enabled. Yet, introduction of ectopic reprogramming factors may desynchronize natural developmental schedules. This study aims to evaluate the effect of imposed transgene load on the cardiogenic competency of induced pluripotent stem (iPS) cells. METHODS AND RESULTS: Targeted inclusion and exclusion of reprogramming transgenes (c-MYC, KLF4, OCT4, and SOX2) was achieved using a drug-inducible and removable cassette according to the piggyBac transposon/transposase system. Pulsed transgene overexpression, before iPS cell differentiation, hindered cardiogenic outcomes. Delayed in counterparts with maintained integrated transgenes, transgene removal enabled proficient differentiation of iPS cells into functional cardiac tissue. Transgene-free iPS cells generated reproducible beating activity with robust expression of cardiac α-actinin, connexin 43, myosin light chain 2a, α/ß-myosin heavy chain, and troponin I. Although operational excitation-contraction coupling was demonstrable in the presence or absence of transgenes, factor-free derivatives exhibited an expedited maturing phenotype with canonical responsiveness to adrenergic stimulation. CONCLUSIONS: A disproportionate stemness load, caused by integrated transgenes, affects the cardiogenic competency of iPS cells. Offload of transgenes in engineered iPS cells ensures integrity of cardiac developmental programs, underscoring the value of nonintegrative nuclear reprogramming for derivation of competent cardiogenic regenerative biologics.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Transgenes , Actinina/biossíntese , Animais , Miosinas Cardíacas/biossíntese , Diferenciação Celular , Separação Celular , Reprogramação Celular , Conexina 43/biossíntese , Eletrofisiologia , Fibroblastos/metabolismo , Citometria de Fluxo , Técnicas Genéticas , Fator 4 Semelhante a Kruppel , Camundongos , Microscopia Eletrônica , Cadeias Pesadas de Miosina/biossíntese , Cadeias Leves de Miosina/biossíntese , Troponina I/biossíntese
8.
Cell Signal ; 26(11): 2370-80, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25038455

RESUMO

Breast cancer is the leading cause of cancer death in women worldwide which is closely related to metastasis. But the exact molecular mechanism on metastasis is still not fully understood; we now report that both MRTF-A and STAT3 play important role in breast cancer migration of MDA-MB-231 cells. Moreover, MRTF-A and STAT3 synergistically increased MDA-MB-231 cell migration by promoting the expression of migration markers Myl-9 and Cyr-61. Importantly, we identified a detailed molecular mechanism of MDA-MB-231 cell migration controlled via physical interaction between MRTF-A and STAT3, which synergistically promote the transactivity of the migration marker Myl-9 and Cyr-61 by CArG box binding. Interestingly, the two signaling pathways RhoA-MRTF-A and JAK-STAT3 across talk to regulate MDA-MB-231 cell migration. Our data thus provide important and novel insights into the roles of MRTF-A and STAT3 in regulating MDA-MB-231 cell migration.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias da Mama/metabolismo , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Fator de Transcrição STAT3/biossíntese , Transativadores/biossíntese , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proteína Rica em Cisteína 61/biossíntese , Proteína Rica em Cisteína 61/genética , Feminino , Humanos , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Proteínas de Neoplasias/genética , Elementos de Resposta , Fator de Transcrição STAT3/genética , Transativadores/genética , Ativação Transcricional/genética
9.
Neurotox Res ; 23(3): 238-48, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22810835

RESUMO

Rho-associated coil kinase (ROCK) inhibitors reportedly prevent neurodegeneration, and abnormal ROCK activation in the central nervous system induces neurite collapse and retraction. However, it is unclear whether the ROCK inhibitor Y-27632 directly protects hippocampal neurons from excitotoxicity. Here, we determined the effects of Y-27632 on neuroprotection following kainic acid (KA)-induced seizures in mice and during glutamate-induced excitotoxicity in HT22 cells. One day after Y-27632 injection, mice were treated with KA and killed 1-2 days later. Fluoro-Jade B and rapid Golgi staining showed that Y-27632 protected against KA-induced neurodegeneration and neurite dystrophy. Y-27632 inhibited increases in hippocampal RhoA and ROCK2 in KA-treated mice as determined by western blot analysis. Immunohistochemical analysis revealed ROCK2-positive neurons and astrocytes in the KA-treated hippocampus. In HT22 cells, Y-27632 also protected neurons and neurite formation during glutamate-induced excitotoxicity in vitro. These results indicate that ROCK inhibition modulates neurite growth and protects neurons from excitotoxicity-induced cell death.


Assuntos
Amidas/uso terapêutico , Anticonvulsivantes/uso terapêutico , Hipocampo/efeitos dos fármacos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Neurotoxinas/toxicidade , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Convulsões/prevenção & controle , Quinases Associadas a rho/antagonistas & inibidores , Amidas/farmacologia , Animais , Anticonvulsivantes/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/enzimologia , Morte Celular , Células Cultivadas/efeitos dos fármacos , Convulsivantes/antagonistas & inibidores , Convulsivantes/toxicidade , Proteína Glial Fibrilar Ácida , Ácido Glutâmico/toxicidade , Hipocampo/citologia , Hipocampo/enzimologia , Hipocampo/patologia , Ácido Caínico/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos ICR , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neuritos/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/ultraestrutura , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Convulsões/induzido quimicamente , Método Simples-Cego , Proteínas rho de Ligação ao GTP/biossíntese , Proteínas rho de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP
10.
J Smooth Muscle Res ; 48(5-6): 137-47, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23538510

RESUMO

CD3 is a complex of polypeptides which form part of the T cell receptor. Normal human peripheral pan T cells, express not only CD3, but the mRNA for myosin regulatory light chains MYL9, MYL12A, and MYL12B are also significantly expressed. In the Jurkat wild strain, an acute T cell leukemia cell line, CD3 on the surface and MYL9 mRNA are not expressed, while both MYL12A mRNA and MYL12B mRNA are expressed. Jurkat-I, a new clone was established by the transfection of the MYL9 gene into the Jurkat wild strain. As a result, the level of CD3 expressed on the surface of Jurkat-I cells was significantly higher than those in the Jurkat wild strain. Phorbol 12-myristate 13-acetate increased the surface CD3 levels in Jurkat wild strain cells without resulting in MYL9 gene expression, indicating that protein kinase C is partially involved in the expression of CD3 on the surface. These results suggest that surface CD3 expression proceeds through both MYL9-dependent and MYL9-independent pathways (i.e. the protein kinase C- dependent pathway) in Jurkat cells.


Assuntos
Complexo CD3/biossíntese , Regulação da Expressão Gênica/fisiologia , Cadeias Leves de Miosina/biossíntese , Proteína Quinase C/metabolismo , Transdução de Sinais/fisiologia , Complexo CD3/genética , Carcinógenos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células Jurkat , Cadeias Leves de Miosina/genética , Proteína Quinase C/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia
11.
Mol Cell Biol ; 32(3): 633-51, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22144583

RESUMO

MicroRNA-200c (miR-200c) has been shown to suppress epithelial-mesenchymal transition (EMT), which is attributed mainly to targeting of ZEB1/ZEB2, repressors of the cell-cell contact protein E-cadherin. Here we demonstrated that modulation of miR-200c in breast cancer cells regulates cell migration, cell elongation, and transforming growth factor ß (TGF-ß)-induced stress fiber formation by impacting the reorganization of cytoskeleton that is independent of the ZEB/E-cadherin axis. We identified FHOD1 and PPM1F, direct regulators of the actin cytoskeleton, as novel targets of miR-200c. Remarkably, expression levels of FHOD1 and PPM1F were inversely correlated with the level of miR-200c in breast cancer cell lines, breast cancer patient samples, and 58 cancer cell lines of various origins. Furthermore, individual knockdown/overexpression of these target genes phenocopied the effects of miR-200c overexpression/inhibition on cell elongation, stress fiber formation, migration, and invasion. Mechanistically, targeting of FHOD1 by miR-200c resulted in decreased expression and transcriptional activity of serum response factor (SRF), mediated by interference with the translocation of the SRF coactivator mycocardin-related transcription factor A (MRTF-A). This finally led to downregulation of the expression and phosphorylation of the SRF target myosin light chain 2 (MLC2) gene, required for stress fiber formation and contractility. Thus, miR-200c impacts on metastasis by regulating several EMT-related processes, including a novel mechanism involving the direct targeting of actin-regulatory proteins.


Assuntos
Neoplasias da Mama/patologia , Proteínas Fetais/metabolismo , MicroRNAs/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Neoplasias da Mama/metabolismo , Miosinas Cardíacas/biossíntese , Linhagem Celular Tumoral , Movimento Celular , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Feminino , Forminas , Regulação Neoplásica da Expressão Gênica , Humanos , Cadeias Leves de Miosina/biossíntese , Invasividade Neoplásica , Proteínas de Fusão Oncogênica/metabolismo , Fator de Resposta Sérica/biossíntese , Fibras de Estresse/metabolismo , Transativadores , Fator de Crescimento Transformador beta/metabolismo
12.
Cancer Cell ; 20(3): 357-69, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21907926

RESUMO

We show constitutive activation of Rho kinase (ROCK) in cells bearing oncogenic forms of KIT, FLT3, and BCR-ABL, which is dependent on PI3K and Rho GTPase. Genetic or pharmacologic inhibition of ROCK in oncogene-bearing cells impaired their growth as well as the growth of acute myeloid leukemia patient-derived blasts and prolonged the life span of mice bearing myeloproliferative disease. Downstream from ROCK, rapid dephosphorylation or loss of expression of myosin light chain resulted in enhanced apoptosis, reduced growth, and loss of actin polymerization in oncogene-bearing cells leading to significantly prolonged life span of leukemic mice. In summary we describe a pathway involving PI3K/Rho/ROCK/MLC that may contribute to myeloproliferative disease and/or acute myeloid leukemia in humans.


Assuntos
Transformação Celular Neoplásica , Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia/metabolismo , Transtornos Mieloproliferativos/metabolismo , Proteínas Tirosina Quinases/metabolismo , Fator de Células-Tronco/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Quinases Associadas a rho/metabolismo , Actinas/metabolismo , Animais , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Proteínas de Fusão bcr-abl/biossíntese , Proteínas de Fusão bcr-abl/genética , Humanos , Leucemia/mortalidade , Leucemia/patologia , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Transtornos Mieloproliferativos/mortalidade , Transtornos Mieloproliferativos/patologia , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo , Fosfatidilinositol 3-Quinases/biossíntese , Fosforilação , Proteínas Tirosina Quinases/biossíntese , Proteínas Tirosina Quinases/genética , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais/genética , Fator de Células-Tronco/biossíntese , Fator de Células-Tronco/genética , Tirosina Quinase 3 Semelhante a fms/biossíntese , Tirosina Quinase 3 Semelhante a fms/genética , Proteínas rho de Ligação ao GTP/biossíntese , Quinases Associadas a rho/antagonistas & inibidores , Quinases Associadas a rho/genética
13.
Am J Physiol Cell Physiol ; 301(5): C995-C1007, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21832246

RESUMO

Skeletal muscle atrophy commonly occurs in acute and chronic disease. The expression of the muscle-specific E3 ligases atrogin-1 (MAFbx) and muscle RING finger 1 (MuRF1) is induced by atrophy stimuli such as glucocorticoids or absence of IGF-I/insulin and subsequent Akt signaling. We investigated whether glycogen synthase kinase-3ß (GSK-3ß), a downstream molecule in IGF-I/Akt signaling, is required for basal and atrophy stimulus-induced expression of atrogin-1 and MuRF1, and myofibrillar protein loss in C(2)C(12) skeletal myotubes. Abrogation of basal IGF-I signaling, using LY294002, resulted in a prominent induction of atrogin-1 and MuRF1 mRNA and was accompanied by a loss of myosin heavy chain fast (MyHC-f) and myosin light chains 1 (MyLC-1) and -3 (MyLC-3). The synthetic glucocorticoid dexamethasone (Dex) also induced the expression of both atrogenes and likewise resulted in the loss of myosin protein abundance. Genetic ablation of GSK-3ß using small interfering RNA resulted in specific sparing of MyHC-f, MyLC-1, and MyLC-3 protein levels after Dex treatment or impaired IGF-I/Akt signaling. Interestingly, loss of endogenous GSK-3ß suppressed both basal and atrophy stimulus-induced atrogin-1 and MuRF1 expression, whereas pharmacological GSK-3ß inhibition, using CHIR99021 or LiCl, only reduced atrogin-1 mRNA levels in response to LY294002 or Dex. In conclusion, our data reveal that myotube atrophy and myofibrillar protein loss are GSK-3ß dependent, and demonstrate for the first time that basal and atrophy stimulus-induced atrogin-1 mRNA expression requires GSK-3ß enzymatic activity, whereas MuRF1 expression depends solely on the physical presence of GSK-3ß.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Músculo Esquelético/enzimologia , Atrofia Muscular/enzimologia , Mioblastos/enzimologia , Animais , Linhagem Celular , Cromonas/farmacologia , Dexametasona/farmacologia , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Cloreto de Lítio/farmacologia , Camundongos , Morfolinas/farmacologia , Proteínas Musculares/biossíntese , Músculo Esquelético/efeitos dos fármacos , Atrofia Muscular/tratamento farmacológico , Mioblastos/efeitos dos fármacos , Mioblastos/fisiologia , Cadeias Pesadas de Miosina/biossíntese , Cadeias Leves de Miosina/biossíntese , Piridinas/farmacologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/metabolismo , Proteínas Ligases SKP Culina F-Box/biossíntese , Transdução de Sinais/efeitos dos fármacos , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/biossíntese
14.
Cell Res ; 21(4): 579-87, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21102549

RESUMO

Although myocyte cell transplantation studies have suggested a promising therapeutic potential for myocardial infarction, a major obstacle to the development of clinical therapies for myocardial repair is the difficulties associated with obtaining relatively homogeneous ventricular myocytes for transplantation. Human embryonic stem cells (hESCs) are a promising source of cardiomyocytes. Here we report that retinoid signaling regulates the fate specification of atrial versus ventricular myocytes during cardiac differentiation of hESCs. We found that both Noggin and the pan-retinoic acid receptor antagonist BMS-189453 (RAi) significantly increased the cardiac differentiation efficiency of hESCs. To investigate retinoid functions, we compared Noggin+RAi-treated cultures with Noggin+RA-treated cultures. Our results showed that the expression levels of the ventricular-specific gene IRX-4 were radically elevated in Noggin+RAi-treated cultures. MLC-2V, another ventricular-specific marker, was expressed in the majority of the cardiomyocytes in Noggin+RAi-treated cultures, but not in the cardiomyocytes of Noggin+RA-treated cultures. Flow cytometry analysis and electrophysiological studies indicated that with 64.7 ± 0.88% (mean ±s.e.m) cardiac differentiation efficiency, 83% of the cardiomyocytes in Noggin+RAi-treated cultures had embryonic ventricular-like action potentials (APs). With 50.7 ± 1.76% cardiac differentiation efficiency, 94% of the cardiomyocytes in Noggin+RA-treated cultures had embryonic atrial-like APs. These results were further confirmed by imaging studies that assessed the patterns and properties of the Ca(2+) sparks of the cardiomyocytes from the two cultures. These findings demonstrate that retinoid signaling specifies the atrial versus ventricular differentiation of hESCs. This study also shows that relatively homogeneous embryonic atrial- and ventricular-like myocyte populations can be efficiently derived from hESCs by specifically regulating Noggin and retinoid signals.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Átrios do Coração/metabolismo , Ventrículos do Coração/metabolismo , Miócitos Cardíacos/metabolismo , Retinoides/metabolismo , Transdução de Sinais , Potenciais de Ação/efeitos dos fármacos , Sequência de Bases , Western Blotting , Miosinas Cardíacas/biossíntese , Miosinas Cardíacas/genética , Proteínas de Transporte/farmacologia , Linhagem Celular , Células-Tronco Embrionárias/citologia , Citometria de Fluxo , Imunofluorescência , Átrios do Coração/embriologia , Ventrículos do Coração/embriologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Reação em Cadeia da Polimerase , Retinoides/farmacologia
15.
Urology ; 76(6): 1517.e6-11, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20970835

RESUMO

OBJECTIVES: To investigate the effects of estrogen, raloxifene, and levormeloxifene on the expression of Rho-kinase signaling molecules in urethral smooth muscle cells (USMCs). METHODS: USMCs were isolated from female rats. Expression of calponin and estrogen receptors α (ERα) was detected by immunofluorescence staining. Cells were treated with estrogen, raloxifene, or levormeloxifene at 0, 1, 10, and 100 nmol/L for 48 h and then processed for Western blotting with antibodies against RhoA, Rho kinase I and II (Rock-I and Rock-II), myosin light chain (MLC), phosphorylated MLC, and ß-actin. Protein expression was quantitated by densitometry, followed by statistical analysis with ß-actin as control. RESULTS: USMCs expressed calponin and ERα. Treatment of USMCs with estrogen, raloxifene or levormeloxifene resulted in decreased expression of RhoA, Rock-I, Rock-II, and p-MLC in a dosage-dependent manner. CONCLUSIONS: Estrogen, raloxifene, and levormeloxifene may affect urinary continence by inhibiting the expression of Rho-kinase signaling molecules.


Assuntos
Estrogênios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Pirrolidinas/farmacologia , Cloridrato de Raloxifeno/farmacologia , Uretra/efeitos dos fármacos , Quinases Associadas a rho/biossíntese , Animais , Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ligação ao Cálcio/genética , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/enzimologia , Células Cultivadas/metabolismo , Indução Enzimática/efeitos dos fármacos , Receptor alfa de Estrogênio/biossíntese , Receptor alfa de Estrogênio/genética , Feminino , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/metabolismo , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Pirrolidinas/toxicidade , Cloridrato de Raloxifeno/toxicidade , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Uretra/citologia , Incontinência Urinária/fisiopatologia , Quinases Associadas a rho/genética , Proteína rhoA de Ligação ao GTP/biossíntese , Proteína rhoA de Ligação ao GTP/genética , Calponinas
16.
Neurogastroenterol Motil ; 22(10): 1100-e285, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20591105

RESUMO

BACKGROUND: The smooth muscle actin binding proteins Caldesmon and Tropomyosin (Tm) promote thin filament assembly by stabilizing actin polymerization, however, whether filament assembly affects either the stability or activation of these and other smooth muscle regulatory proteins is not known. METHODS: Measurement of smooth muscle regulatory protein levels in wild type zebrafish larvae following antisense knockdown of smooth muscle actin (Acta2) and myosin heavy chain (Myh11) proteins, and in colourless mutants that lack enteric nerves. Comparison of intestinal peristalsis in wild type and colourless larvae. KEY RESULTS: Knockdown of Acta2 led to reduced levels of phospho-Caldesmon and Tm. Total Caldesmon and phospho-myosin light chain (p-Mlc) levels were unaffected. Knockdown of Myh11 had no effect on the levels of either of these proteins. Phospho-Caldesmon and p-Mlc levels were markedly reduced in colourless mutants that have intestinal motility comparable with wild type larvae. CONCLUSIONS & INFERENCES: These in vivo findings provide new information regarding the activation and stability of smooth muscle regulatory proteins in zebrafish larvae and their role in intestinal peristalsis in this model organism.


Assuntos
Citoesqueleto de Actina/fisiologia , Intestinos/citologia , Intestinos/fisiologia , Proteínas Musculares/metabolismo , Músculo Liso/citologia , Músculo Liso/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/fisiologia , Animais , Proteínas de Ligação a Calmodulina/biossíntese , Proteínas de Ligação a Calmodulina/genética , Sistema Nervoso Entérico/fisiologia , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Larva , Proteínas dos Microfilamentos/metabolismo , Microscopia Confocal , Morfolinas/farmacologia , Músculo Liso/metabolismo , Mutação/fisiologia , Cadeias Pesadas de Miosina/biossíntese , Cadeias Pesadas de Miosina/genética , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Peristaltismo/fisiologia , RNA Antissenso , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Tropomiosina/biossíntese , Tropomiosina/genética
17.
J Periodontal Res ; 45(5): 618-25, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20546111

RESUMO

BACKGROUND AND OBJECTIVE: The junctional epithelium attaches to the tooth enamel at the dentogingival junction. The attachment mechanisms of the junctional epithelium have been studied histologically, but the molecular functions of the junctional epithelium have not been elucidated. The aim of this study was to perform a comprehensive analysis of gene expression in the junctional epithelium and to search for specific genetic markers of the junctional epithelium. MATERIAL AND METHODS: A comprehensive analysis of genes expressed in the mouse junctional epithelium and oral gingival epithelium was performed using laser microdissection and microarray analysis. To extract high-quality RNA from these tissues, we made frozen sections using a modified film method. Confirmation of the differential expression of selected genes was performed by quantitative real-time PCR and immunohistochemistry. RESULTS: The modified method produced RNA of sufficient quality for microarray analysis. The result of microarray analysis showed that 841 genes were up-regulated in the junctional epithelium compared with the oral gingival epithelium, and five were increased more than 50-fold in the junctional epithelium. These five genes were secretory leukocyte protease inhibitor (Slpi), keratin 17 (Krt17), annexin A1 (Anxa1), myosin light peptide 6 (Myl6) and endoplasmic reticulum protein 29 (Erp29). In particular, Slpi expression in the junctional epithelium was approximately 100-fold higher than in the oral gingival epithelium by real-time PCR. Additionally, immunohistochemistry indicated that the Slpi protein is highly expressed in the junctional epithelium. CONCLUSION: We developed a method for generating fresh-frozen tissue sections suitable for extraction of good-quality RNA. We determined that Slpi is characteristically expressed in the junctional epithelium. Our results provide a substantial advance in the analysis of gene expression in the junctional epithelium.


Assuntos
Inserção Epitelial/metabolismo , Perfilação da Expressão Gênica/métodos , Inibidor Secretado de Peptidases Leucocitárias/biossíntese , Animais , Anexina A1/biossíntese , Anexina A1/genética , Retículo Endoplasmático , Inserção Epitelial/enzimologia , Secções Congeladas , Gengiva/metabolismo , Proteínas de Choque Térmico/biossíntese , Proteínas de Choque Térmico/genética , Queratina-17/biossíntese , Queratina-17/genética , Lasers de Gás , Camundongos , Microdissecção/métodos , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/genética , Análise de Sequência com Séries de Oligonucleotídeos , Inibidor Secretado de Peptidases Leucocitárias/genética
18.
Am J Physiol Heart Circ Physiol ; 297(1): H191-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19429828

RESUMO

The participation of nonmuscle myosin in force maintenance is controversial. Furthermore, its regulation is difficult to examine in a cellular context, as the light chains of smooth muscle and nonmuscle myosin comigrate under native and denaturing electrophoresis techniques. Therefore, the regulatory light chains of smooth muscle myosin (SM-RLC) and nonmuscle myosin (NM-RLC) were purified, and these proteins were resolved by isoelectric focusing. Using this method, intact mouse aortic smooth muscle homogenates demonstrated four distinct RLC isoelectric variants. These spots were identified as phosphorylated NM-RLC (most acidic), nonphosphorylated NM-RLC, phosphorylated SM-RLC, and nonphosphorylated SM-RLC (most basic). During smooth muscle activation, NM-RLC phosphorylation increased. During depolarization, the increase in NM-RLC phosphorylation was unaffected by inhibition of either Rho kinase or PKC. However, inhibition of Rho kinase blocked the angiotensin II-induced increase in NM-RLC phosphorylation. Additionally, force for angiotensin II stimulation of aortic smooth muscle from heterozygous nonmuscle myosin IIB knockout mice was significantly less than that of wild-type littermates, suggesting that, in smooth muscle, activation of nonmuscle myosin is important for force maintenance. The data also demonstrate that, in smooth muscle, the activation of nonmuscle myosin is regulated by Ca(2+)-calmodulin-activated myosin light chain kinase during depolarization and a Rho kinase-dependent pathway during agonist stimulation.


Assuntos
Músculo Liso/fisiologia , Miosinas/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Sequência de Aminoácidos , Angiotensina II/farmacologia , Animais , Western Blotting , Galinhas , Eletroforese em Gel Bidimensional , Regulação da Expressão Gênica/fisiologia , Espectrometria de Massas , Camundongos , Camundongos Transgênicos , Contração Muscular/fisiologia , Músculo Liso/efeitos dos fármacos , Cadeias Leves de Miosina/biossíntese , Cadeias Leves de Miosina/química , Cadeias Leves de Miosina/metabolismo , Miosinas/química , Miosinas/genética , Miosina não Muscular Tipo IIB/química , Miosina não Muscular Tipo IIB/genética , Fosforilação/efeitos dos fármacos , Proteínas Recombinantes , Quinases Associadas a rho/antagonistas & inibidores
19.
Stem Cells Dev ; 17(6): 1131-40, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19006452

RESUMO

Previous studies have suggested that mesenchymal stem cells (MSCs) can differentiate into smooth muscle-like cells. However their functionalities remain questionable. The aim of this study was to investigate the functionality of MSCs differentiated into smooth muscle (SM) in vitro by SM-inducing medium. MSCs have been isolated from rat bone marrow and cultured in SM-inducing medium. After 21 days in culture, messenger RNA and specific SM proteins such as myosin heavy chain and myosin light chain 2 were expressed in the in vitro differentiated MSCs to a similar level of that in freshly isolated SM cells (SMCs). At the electrophysiological level, MSCs presented an outward K+ current with an IK(DR) component and IK(Ca) component. In vitro differentiation induced an enhancement of the IK(Ca) current to a level similar to that observed in aortic SMCs. Calcium homeostasis measurements revealed that both differentiated and undifferentiated MSCs responded to extracellular adenosine triphosphate (ATP) in a similar fashion to SMCs. However MSCs failed to contract in response to ATP. This data shows that despite specific SM protein expression and modification of electrophysiological properties similar to that of aortic SMCs, MSCs cultured in differentiation medium failed to display contractile properties. These results underline the necessity to find the ideal cultured conditions to induce complete SMC function.


Assuntos
Diferenciação Celular/fisiologia , Potenciais da Membrana/fisiologia , Células-Tronco Mesenquimais/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Cálcio/metabolismo , Miosinas Cardíacas/biossíntese , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Homeostase/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/citologia , Cadeias Pesadas de Miosina/biossíntese , Cadeias Leves de Miosina/biossíntese , Ratos , Fatores de Tempo
20.
Brain Res Bull ; 74(6): 439-51, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17920452

RESUMO

Movement of glutamate receptors in neurons likely involves direct and indirect association of receptor subunits with microtubule- and actin-based motor proteins. We have previously shown that myosin II regulatory light chain (RLC) binds directly to subunits of the NMDA-type glutamate receptor (NR), suggesting that NMDA receptors are closely associated with a myosin II motor complex. Using a polyclonal antibody predicted to recognize all RLC isoforms previously described in rodent brain, we report the expression of RLC and the NR1 subunit in cortex, hippocampus and cerebellum of postnatal day 0 (P0) and adult mouse. Although myosin RLC was not exclusively localized with NR1 by immunohistochemistry, co-staining was striking in the neuronal soma of deep cortical neurons and Purkinje neurons of the cerebellum which showed a punctate, perinuclear pattern of immunoreactivity. These neuronal populations were identified using a monoclonal antibody directed against a nuclear-specific, transcriptional repressor, chicken ovalbumin upstream promoter-transcription factor (COUP-TF)-interacting protein 2 (CTIP2). Co-expression of NR1 and a myosin II motor was validated using an isoform specific anti-nonmuscle myosin II-B heavy chain (NMHC II-B) antibody. Our findings support the idea that there is regional heterogeneity in the molecular composition of the NMDA receptor-associated cytoskeleton, and suggest that NR subunits may be associated with an actin-based, myosin II-B motor within the endomembrane system of some neuronal populations. Differential staining patterns observed with light and heavy chain antibodies, however, suggest that there is also heterogeneity in the composition of myosin II complexes in brain.


Assuntos
Encéfalo/metabolismo , Cadeias Leves de Miosina/biossíntese , Miosina Tipo II/biossíntese , Receptores de N-Metil-D-Aspartato/biossíntese , Animais , Animais Recém-Nascidos , Western Blotting , Feminino , Imuno-Histoquímica , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA