Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Biochem Cell Biol ; 99(5): 578-586, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33646885

RESUMO

Duloxetine, a selective serotonin-norepinephrine reuptake inhibitor, is currently recommended for the treatment of chronic painful disorders such as fibromyalgia, chronic musculoskeletal pain, and diabetic peripheral neuropathy. We previously demonstrated that bone morphogenetic protein-4 (BMP-4) stimulates osteoprotegerin (OPG) production in osteoblast-like MC3T3-E1 cells, and that p70 S6 kinase positively regulates OPG synthesis. The present study aimed to investigate the effect of duloxetine on BMP-4-stimulated OPG synthesis in these cells. Duloxetine dose-dependently suppressed OPG release stimulated by BMP-4. Fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), reduced BMP-4-stimulated OPG release, whereas a selective and specific norepinephrine reuptake inhibitor, reboxetine, failed to affect OPG release. In addition, another SSRI sertraline also inhibited BMP-4-stimulated OPG release. On the other hand, siRNA of SMAD1 reduced the OPG release stimulated by BMP-4, indicating the involvement of the SMAD1/5/8 pathway in OPG release. Rapamycin inhibited BMP-4-stimulated p70 S6 kinase phosphorylation, and compound C suppressed the SMAD1/5/8 phosphorylation stimulated by BMP-4. Duloxetine did not affect BMP-4-induced phosphorylation of p70 S6 kinase but suppressed SMAD1/5/8 phosphorylation. Both fluvoxamine and sertraline also inhibited BMP-4-elicited phosphorylation of SMAD1/5/8. These results strongly suggest that duloxetine suppresses BMP-4-stimulated OPG release via inhibition of the Smad1/5/8 signaling pathway in osteoblasts.


Assuntos
Proteína Morfogenética Óssea 4/antagonistas & inibidores , Cloridrato de Duloxetina/farmacologia , Osteoblastos/efeitos dos fármacos , Osteoprotegerina/antagonistas & inibidores , Células 3T3 , Animais , Proteína Morfogenética Óssea 4/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Camundongos , Osteoblastos/metabolismo , Osteoprotegerina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/metabolismo , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/metabolismo , Proteína Smad8/antagonistas & inibidores , Proteína Smad8/metabolismo
2.
Eur Rev Med Pharmacol Sci ; 24(10): 5691-5696, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32495904

RESUMO

OBJECTIVE: To elucidate the role of Prunella vulgaris L (PVL) in protecting glucocorticoids (GC)-induced osteogenesis inhibition, thereafter, protecting the deterioration of osteoporosis (OP). MATERIALS AND METHODS: Cell Counting Kit-8 (CCK-8) assay was conducted to assess the influence of PVL treatment on MSCs viability. Osteogenesis in MSCs was induced by Dexamethasone (DEX) stimulation. Regulatory effects of PVL on osteogenesis-related gene expressions, ALP activity, and mineralization ability in DEX-induced MSCs were determined. At last, protein levels of p-Smad1/5/9 and total-Smad1/5/9 influenced by DEX and PVL were measured by Western blot. RESULTS: PVL treatment did not pose a time- or dose-dependent influence on MSCs viability. DEX induction in MSCs downregulated ALP, RUNX2, Bglap, and Osterix. ALP activity and mineralization in DEX-induced MSCs were suppressed. Downregulated osteogenesis-related genes decreased ALP activity and mineralization in MSCs undergoing DEX stimulation were partially reversed by PVL treatment. Moreover, the downregulated p-Smad1/5/9 level in DEX-induced MSCs was elevated by PVL treatment, while total-Smad1/5/9 was not affected. CONCLUSIONS: PVL alleviated GC-induced suppression in MSCs osteogenesis by activating the Smad pathway, thereafter, protecting the deterioration of OP.


Assuntos
Dexametasona/antagonistas & inibidores , Glucocorticoides/antagonistas & inibidores , Células-Tronco Mesenquimais/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Prunella/química , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Células Cultivadas , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Substâncias Protetoras/química , Proteína Smad1/antagonistas & inibidores , Proteína Smad5/antagonistas & inibidores , Proteína Smad8/antagonistas & inibidores
3.
Mol Med Rep ; 21(1): 478-484, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31939623

RESUMO

Endothelial to mesenchymal transition (EndMT) has been confirmed to participate in several cardiovascular diseases. In addition, EndMT of circulating endothelial cells (CECs) contributes to the pathology of musculoskeletal injury. However, little is known about the molecular mechanism of CECs undergoing EndMT. In the present study, human CECs were isolated and identified using anti­CD146­coupled magnetic beads. CECs were exposed to transforming growth factor (TGF)­ß1 or TGF­ß1 + recombinant human bone morphogenetic protein 7 (rhBMP­7) or TGF­ß1 + rhBMP­7 + Smad5 antagonist Jun activation domain­binding protein 1. Vascular endothelial (VE)­cadherin and vimentin expression were detected by immunofluorescence staining in TGF­ß1­treated CECs. The expression levels of von Willebrand factor (vWF), E­selectin, VE­cadherin, vimentin, fibronectin, α smooth muscle actin (α­SMA) and Smad2/3 were detected by reverse transcription­quantitative PCR or western blot analysis. It was identified that rhBMP­7 attenuated TGF­ß1­induced endothelial cell injury. TGF­ß1 could induce the EndMT process in CECs, as confirmed by the co­expression of VE­cadherin and vimentin. TGF­ß1 significantly reduced the expression of VE­cadherin, and induced the expression of vimentin, fibronectin and α­SMA. rhBMP­7 reversed the effects of TGF­ß1 on the expression of these genes. Additionally, Smad5 antagonist reversed the effects of rhBMP­7 on TGF­ß1­induced EndMT, and upregulated rhBMP­7­inhibited Smad2/3 expression. In conclusion, TGF­ß1 could induce EndMT in CECs and rhBMP­7 may suppress this process by regulating Smad5.


Assuntos
Proteína Morfogenética Óssea 7/metabolismo , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal/genética , Proteína Smad5/antagonistas & inibidores , Fator de Crescimento Transformador beta1/farmacologia , Adulto , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/farmacologia , Antígeno CD146/metabolismo , Complexo do Signalossomo COP9/farmacologia , Caderinas/metabolismo , Técnicas de Cocultura , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Masculino , Peptídeo Hidrolases/farmacologia , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Proteína Smad5/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Vimentina/metabolismo
4.
Mol Med Rep ; 21(1): 61-68, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31939626

RESUMO

Considering the increased resistance to antibiotics in the clinic and the ideal antibacterial properties of KR­12, the effects of KR­12­a6, an important analogue of KR­12, on the osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) were investigated. Osteogenic differentiation­associated experiments were conducted in hBMSCs, and KR­12­a6 was used as an additional stimulating factor during osteogenic induction. Quantitative analysis of alkaline phosphatase (ALP) and alizarin red staining, and reverse transcription­quantitative PCR analysis of the expression of osteogenesis­associated genes were performed to determine the effects of KR­12­a6 on the osteogenic differentiation of hBMSCs. LDN­212854 was selected to selectively suppress BMP/SMAD signaling. Western blotting was performed to investigate the underlying mechanisms. The intensity of ALP and alizarin red staining gradually increased with increasing KR­12­a6 concentrations. KR­12­a6 induced the strongest staining at 40 µg/ml, whereas 60 µg/ml and 80 µg/ml concentrations did not further increase the intensity of staining. The mRNA expression levels of RUNX2 and ALP increased in a dose­dependent manner as early as 3 days post­KR­12­a6 treatment. The mRNA expression of COL1A1, BSP and BMP2 exhibited significant upregulation from day 7 post­KR­12­a6 treatment. In contrast, the mRNA levels of OSX, OCN and OPN were enhanced dramatically at day 14 following KR­12­a6 stimulation. Additionally, KR­12­a6 significantly promoted the phosphorylation of Smad1/5. Furthermore, LDN­212854 suppressed the activation of Smad1/5 and inhibited the upregulation of several osteogenic differentiation­associated genes in KR­12­a6­treated hBMSCs. KR­12­a6 promoted the osteogenic differentiation of hBMSCs via BMP/SMAD signaling.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Imidazóis/farmacologia , Nylons/farmacologia , Osteogênese/efeitos dos fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Adulto , Fosfatase Alcalina/metabolismo , Proteína Morfogenética Óssea 2/genética , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Feminino , Humanos , Imidazóis/farmacocinética , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Nylons/farmacocinética , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese/genética , Osteopontina/genética , Fragmentos de Peptídeos/genética , Transdução de Sinais/genética , Proteína Smad1/antagonistas & inibidores , Proteína Smad5/antagonistas & inibidores , Fator de Transcrição Sp7/genética , Fator de Transcrição Sp7/metabolismo
5.
Endocrinology ; 159(1): 297-309, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29029022

RESUMO

The goal of this study was to characterize the function of microRNA-27a-3p (miR-27a-3p) in polycystic ovary syndrome (PCOS). miR-27a-3p expression was analyzed in excised granulosa cells (GCs) from 21 patients with PCOS and 12 normal patients undergoing in vitro fertilization cycle treatments and in 17 nontreated cuneiform ovarian resection PCOS samples and 13 control ovarian samples from patients without PCOS. We found that the expression of miR-27a-3p was significantly increased in both excised GCs and the ovaries of patients with PCOS compared with the controls. Insulin treatment of the human granulosa-like tumor cell line (KGN) resulted in decreased downregulated expression of miR-27a-3p, and this effect appeared to be mediated by signal transducer and activator of transcription STAT1 and STAT3. The overexpression of miR-27a-3p in KGN cells inhibited SMAD5, which in turn decreased cell proliferation and promoted cell apoptosis. After KGN cells were stimulated with insulin for 48 hours, there was increased expression of SMAD5 protein and decreased apoptosis. Additionally, knockdown/overexpression of SMAD5 in KGN cells reduced/increased cell number and promoted/inhibited cell apoptosis. Insulin-stimulated primary GCs isolated from patients with PCOS, in contrast to normal GCs or KGN cells, did not exhibit decreased miR-27a-3p expression. The differences in the expression levels in KGN cells and human PCOS GCs are likely explained by increased miR-27a-3p expression in the GCs caused by insulin resistance in PCOS. Taken together, our data provided evidence for a functional role of miR-27a-3p in the GCs' dysfunction that occurs in patients with PCOS.


Assuntos
Apoptose , Regulação da Expressão Gênica , Células da Granulosa/metabolismo , MicroRNAs/metabolismo , Síndrome do Ovário Policístico/metabolismo , Proteína Smad5/antagonistas & inibidores , Adulto , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Tumor de Células da Granulosa/tratamento farmacológico , Tumor de Células da Granulosa/metabolismo , Tumor de Células da Granulosa/patologia , Células da Granulosa/efeitos dos fármacos , Células da Granulosa/patologia , Humanos , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Resistência à Insulina , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/patologia , RNA/metabolismo , Interferência de RNA , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Proteína Smad5/agonistas , Proteína Smad5/genética , Proteína Smad5/metabolismo
6.
Mol Hum Reprod ; 23(3): 155-165, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27986931

RESUMO

STUDY QUESTION: Does bone morphogenetic protein 2 (BMP2) regulate connexin43 (Cx43) and modulate cell-cell communication in luteinized human granulosa cells? SUMMARY ANSWER: BMP2 decreases gap junction intercellular communication (GJIC) of luteinized human granulosa cells by down-regulating Cx43 expression through an activin receptor-like kinase (ALK)2/ALK3-mediated Sma- and Mad-related protein (SMAD)-dependent signaling pathway. WHAT IS KNOWN ALREADY: BMP2 and its putative receptors are highly expressed in the human corpus luteum and are involved in the process of luteolysis. Cx43-coupled gap junctions play a critical role in the development and maintenance of corpus luteum. STUDY DESIGN DURATION: This is a laboratory study conducted over a 1-year period. At least three independent experiments with three replicates were conducted and the experimental samples were compared with the appropriate vehicle controls for all of the inhibition-approach, concentration-dependent or time-course studies. PARTICIPANTS/MATERIALS, SETTING, METHODS: SVOG cell line (immortalized human granulosa-lutein cells derived from in vitro fertilization patients in an academic research center) was used as the study model. The changes of Cx43 expression and levels of phosphorylated SMAD1/5/8 protein were evaluated after exposure to recombinant human BMP2. Real-time quantitative PCR and Western blot analysis were used to examine the specific mRNA and protein levels, respectively. The BMP/TGF-ß type I receptor inhibitors (Dorsomorphin, DMH-1 and SB431542) and target depletion small interfering RNAs (ALK2, ALK3, ALK6 and SMAD4) were used to investigate the underlying molecular mechanisms. A scrape loading and dye transfer assay was used to evaluate the GJIC between the SVOG cells. MAIN RESULTS AND THE ROLE OF CHANCE: Treatment with BMP2 down-regulated the expression of Cx43 and decreased the GJIC activity, whereas it increased the phosphorylated SMAD1/5/8 protein in SVOG cells (P < 0.05). These biological effects were abolished by pre-treatment with the BMP type I receptor inhibitors, Dorsomorphin and DMH-1 (P < 0.05), but not SB431542. Additionally, the individual or concomitant small interfering RNA-mediated knockdown of ALK2 and ALK3, but not ALK6 attenuated the BMP2-induced increases in phosphorylated SMAD1/5/8 and down-regulation of Cx43 expression (P < 0.05). The knockdown of SMAD4 completely abolished the BMP2-induced down-regulation of Cx43 expression (P < 0.05). LIMITATIONS REASONS FOR CAUTION: This experimental study was conducted in an in vitro cell culture system, and may not reflect a realistic intra-ovarian environment. WIDER IMPLICATIONS OF THE FINDINGS: Our results suggested that BMP2 may be involved in the local modulation of cell-cell communication in the luteal phase. This study also represents the first comprehensive research of molecular mechanisms of BMP2 in the down-regulation Cx43 in luteinized human granulosa cells. Such data may provide valuable insights into ovarian physiology and benefit the development of potential therapeutic methods for patients suffering from luteal insufficiency. LARGE SCALE DATA: N/A. STUDY FUNDING AND COMPETING INTEREST(s): This research was supported by an operating grant from the China-Canadian Joint Health Research Initiative Grants Program to P.C.K. Leung and J.Z. Sheng. The authors declare no competing interest with the contents of this article.


Assuntos
Receptores de Ativinas Tipo I/genética , Proteína Morfogenética Óssea 2/farmacologia , Comunicação Celular/efeitos dos fármacos , Conexina 43/genética , Células Lúteas/efeitos dos fármacos , Receptores de Ativinas Tipo I/metabolismo , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Linhagem Celular Transformada , Conexina 43/antagonistas & inibidores , Conexina 43/metabolismo , Feminino , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Regulação da Expressão Gênica , Humanos , Células Lúteas/citologia , Células Lúteas/metabolismo , Fosforilação/efeitos dos fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Pirazóis/farmacologia , Pirimidinas/farmacologia , Quinolinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteína Smad8/antagonistas & inibidores , Proteína Smad8/genética , Proteína Smad8/metabolismo
7.
Dev Cell ; 34(2): 229-41, 2015 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-26190145

RESUMO

FGF, BMP, and WNT balance embryonic nephron progenitor cell (NPC) renewal and differentiation. By modulating these pathways, we have created an in vitro niche in which NPCs from embryonic kidneys or derived from human embryonic stem cells can be propagated. NPC cultures expanded up to one billion-fold in this environment can be induced to form tubules expressing nephron differentiation markers. Single-cell culture reveals phenotypic variability within the early CITED1-expressing NPC compartment, indicating that it is a mixture of cells with varying progenitor potential. Furthermore, we find that the developmental age of NPCs does not correlate with propagation capacity, indicating that cessation of nephrogenesis is related to factors other than an intrinsic clock. This in vitro nephron progenitor niche will have important applications for expansion of cells for engraftment and will facilitate investigation of mechanisms that determine the balance between renewal and differentiation in these cells.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Néfrons/embriologia , Proteínas Nucleares/biossíntese , Transativadores/biossíntese , Proteínas Wnt/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Células-Tronco Embrionárias/citologia , Ativação Enzimática , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Néfrons/citologia , Proteínas Nucleares/genética , Organogênese , Pirazóis/farmacologia , Pirimidinas/farmacologia , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/metabolismo , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/metabolismo , Transativadores/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
8.
Mol Med Rep ; 12(2): 2189-96, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25891964

RESUMO

The current study investigated the role of exogenous cytochrome c in sepsis-induced myocardial dysfunction (SIMD) using a mouse model and aimed to elucidate its effect on transforming growth factor-ß1 (TGF-ß1) expression during this process. A total of 75 male Kunming mice were randomly divided into the following five group: Normal (N, n=15); sham-operation (SHAM, n=15); sepsis (CLP, n=15); normal saline (NS, n=15); and cytochrome c (Cytc, n=15). Animals were sacrificed at 0, 6 or 12 h and the samples were analyzed using transmission electron microscopy, histopathological examination, reverse transcription-quantitative polymerase chain reaction, ELISA, protein analysis by western blotting. The SIMD model was developed and a significant downregulation of TGF-ß1 gene expression, in addition to a reduction in the plasma and protein levels of TGF-ß1 as well as the protein levels of TGF-ß1-activated SMAD 1/5/8 were observed in the CLP group. The data from the current study indicate that using exogenous cytochrome c as a therapeutic strategy for SIMD is feasible, and may function via the downregulation of TGF-ß1 expression through the SMAD 1/5/8 signaling pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Cardiomiopatias/tratamento farmacológico , Citocromos c/farmacologia , Sepse/tratamento farmacológico , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Cardiomiopatias/etiologia , Cardiomiopatias/genética , Cardiomiopatias/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Masculino , Camundongos , Miocárdio/metabolismo , Miocárdio/patologia , Miocárdio/ultraestrutura , Sepse/complicações , Sepse/genética , Sepse/patologia , Transdução de Sinais , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/sangue , Proteína Smad1/genética , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/sangue , Proteína Smad5/genética , Proteína Smad8/antagonistas & inibidores , Proteína Smad8/sangue , Proteína Smad8/genética , Fator de Crescimento Transformador beta1/sangue , Fator de Crescimento Transformador beta1/genética
9.
Blood ; 124(3): 393-402, 2014 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-24894772

RESUMO

Bone morphogenetic protein (BMP) signaling regulates early hematopoietic development, proceeding from mesoderm patterning through the progressive commitment and differentiation of progenitor cells. The BMP pathway signals largely through receptor-mediated activation of Mothers Against Decapentaplegic homolog (SMAD) proteins, although alternate pathways are modulated through various components of mitogen-activated protein kinase (MAPK) signaling. Using a conditional, short hairpin RNA (shRNA)-based knockdown system in the context of differentiating embryonic stem cells (ESCs), we demonstrated previously that Smad1 promotes hemangioblast specification, but then subsequently restricts primitive progenitor potential. Here we show that co-knockdown of Smad5 restores normal progenitor potential of Smad1-depleted cells, suggesting opposing functions for Smad1 and Smad5. This balance was confirmed by cotargeting Smad1/5 with a specific chemical antagonist, LDN193189 (LDN). However, we discovered that LDN treatment after hemangioblast commitment enhanced primitive myeloid potential. Moreover, inhibition with LDN (but not SMAD depletion) increased expression of Delta-like ligands Dll1 and Dll3 and NOTCH activity; abrogation of NOTCH activity restored LDN-enhanced myeloid potential back to normal, corresponding with expression levels of the myeloid master regulator, C/EBPα. LDN but not SMAD activity was also associated with activation of the p38MAPK pathway, and blocking this pathway was sufficient to enhance myelopoiesis. Therefore, NOTCH and p38MAPK pathways balance primitive myeloid progenitor output downstream of the BMP pathway.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Mielopoese/fisiologia , Receptores Notch/metabolismo , Proteínas Smad/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Proteínas de Ligação ao Cálcio , Células Cultivadas , Corpos Embrioides/citologia , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/efeitos dos fármacos , Células Progenitoras Mieloides/metabolismo , Mielopoese/efeitos dos fármacos , Mielopoese/genética , Pirazóis/farmacologia , Pirimidinas/farmacologia , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/genética , Proteína Smad5/metabolismo
10.
Dev Biol ; 367(2): 216-27, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22609550

RESUMO

Bone Morphogenetic Proteins (BMPs) have multiple activities in the developing spinal cord: they specify the identity of the dorsal-most neuronal populations and then direct the trajectories of dorsal interneuron (dI) 1 commissural axons. How are these activities decoded by dorsal neurons to result in different cellular outcomes? Our previous studies have shown that the diverse functions of the BMPs are mediated by the canonical family of BMP receptors and then regulated by specific inhibitory (I) Smads, which block the activity of a complex of Smad second messengers. However, the extent to which this complex translates the different activities of the BMPs in the spinal cord has remained unresolved. Here, we demonstrate that the receptor-activated (R) Smads, Smad1 and Smad5 play distinct roles mediating the abilities of the BMPs to direct cell fate specification and axon outgrowth. Smad1 and Smad5 occupy spatially distinct compartments within the spinal cord, with Smad5 primarily associated with neural progenitors and Smad1 with differentiated neurons. Consistent with this expression profile, loss of function experiments in mouse embryos reveal that Smad5 is required for the acquisition of dorsal spinal neuron identities whereas Smad1 is critical for the regulation of dI1 axon outgrowth. Thus the R-Smads, like the I-Smads, have discrete roles mediating BMP-dependent cellular processes during spinal interneuron development.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Smad Reguladas por Receptor/metabolismo , Medula Espinal/embriologia , Medula Espinal/metabolismo , Animais , Proteínas Aviárias/antagonistas & inibidores , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Axônios/metabolismo , Sequência de Bases , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Hibridização In Situ , Interneurônios/citologia , Interneurônios/metabolismo , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Modelos Neurológicos , Neurogênese , RNA Interferente Pequeno/genética , Ratos , Proteínas Smad Reguladas por Receptor/antagonistas & inibidores , Proteínas Smad Reguladas por Receptor/genética , Proteína Smad1/antagonistas & inibidores , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/genética , Proteína Smad5/metabolismo , Medula Espinal/citologia
11.
J Virol ; 86(3): 1372-81, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22013049

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) encodes 12 pre-microRNAs (pre-miRNAs). Current studies have shown that these miRNAs are involved in regulation of viral and host gene expression, implicating a role in the maintenance of viral latency and suppression of antiviral innate immunity. However, the functions of these miRNAs remain largely unknown. On the basis of the sequence homology between oncogenic miR-155 and KSHV-encoded miR-K12-11, we hypothesized that miR-K12-11 could attenuate transforming growth factor ß (TGF-ß) signaling, facilitating viral infection and tumorigenesis. In the present study, we demonstrated that ectopic expression of miR-K12-11 in Ramos, a TGF-ß-sensitive cell line, downregulated TGF-ß signaling and facilitated cell proliferation upon TGF-ß treatment by directly targeting SMAD5, an important mediator in TGF-ß signaling. In addition, the downregulation of SMAD5 by miR-K12-11 was further confirmed in a de novo KSHV infection system or latently infected KSHV-positive B-lymphoma cell lines. More importantly, repression of miR-K12-11 by a specific sponge inhibitor restored the expression of SMAD5 in both de novo-infected and latently infected cells. Finally, we found that restoration of SMAD5, in addition to the TGF-ß type II receptor, which was epigenetically silenced by the latent viral protein latency-associated nuclear antigen, sensitized BC3 cells to the cytostatic effect of TGF-ß signaling. Taken together, our findings highlight a novel mechanism in which miR-K12-11 downregulates TGF-ß signaling and suggest that viral miRNAs and proteins may exert a dichotomy regulation in virus-induced oncogenesis by targeting the same signaling pathway.


Assuntos
Herpesvirus Humano 8/fisiologia , MicroRNAs/fisiologia , Transdução de Sinais/fisiologia , Proteína Smad5/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo , Sequência de Bases , Linhagem Celular , Primers do DNA , Herpesvirus Humano 8/genética , Humanos , Reação em Cadeia da Polimerase/métodos , Proteína Smad5/metabolismo
12.
PLoS One ; 6(11): e27403, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22102893

RESUMO

The specification of the neural crest progenitor cell (NCPC) population in the early vertebrate embryo requires an elaborate network of signaling pathways, one of which is the Bone Morphogenetic Protein (BMP) pathway. Based on alterations in neural crest gene expression in zebrafish BMP pathway component mutants, we previously proposed a model in which the gastrula BMP morphogen gradient establishes an intermediate level of BMP activity establishing the future NCPC domain. Here, we tested this model and show that an intermediate level of BMP signaling acts directly to specify the NCPC. We quantified the effects of reducing BMP signaling on the number of neural crest cells and show that neural crest cells are significantly increased when BMP signaling is reduced and that this increase is not due to an increase in cell proliferation. In contrast, when BMP signaling is eliminated, NCPC fail to be specified. We modulated BMP signaling levels in BMP pathway mutants with expanded or no NCPCs to demonstrate that an intermediate level of BMP signaling specifies the NCPC. We further investigated the ability of Smad5 to act in a graded fashion by injecting smad5 antisense morpholinos and show that increasing doses first expand the NCPCs and then cause a loss of NCPCs, consistent with Smad5 acting directly in neural crest progenitor specification. Using Western blot analysis, we show that P-Smad5 levels are dose-dependently reduced in smad5 morphants, consistent with an intermediate level of BMP signaling acting through Smad5 to specify the neural crest progenitors. Finally, we performed chimeric analysis to demonstrate for the first time that BMP signal reception is required directly by NCPCs for their specification. Together these results add substantial evidence to a model in which graded BMP signaling acts as a morphogen to pattern the ectoderm, with an intermediate level acting in neural crest specification.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Embrião não Mamífero/metabolismo , Crista Neural/metabolismo , Transdução de Sinais , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Western Blotting , Padronização Corporal , Proteínas Morfogenéticas Ósseas/genética , Ectoderma/citologia , Ectoderma/metabolismo , Embrião não Mamífero/citologia , Gástrula/citologia , Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Técnicas Imunoenzimáticas , Hibridização In Situ , Morfolinos/farmacologia , Mutação/genética , Crista Neural/citologia , Neurônios/citologia , Neurônios/metabolismo , Fenótipo , Fosforilação , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/genética , Proteína Smad5/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética
13.
Am J Physiol Renal Physiol ; 293(5): F1641-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17804487

RESUMO

In early diabetic renal injury, there is podocyte drop-out (but no decrease in the number of other glomerular cells) which is thought to cause glomerular proteinuria and subsequent diabetic glomerular injury. We tested the hypothesis that early diabetic podocyte injury is caused, in part, by downregulation of bone morphogenetic protein-7 (BMP7) and loss of its autocrine function in murine podocytes. High glucose (HG; 25 mM) induces rounding of differentiated podocytes and changes in the distribution of F-actin but without quantitative changes in E-cadherin and the podocyte markers podocin, CD2AP, Neph1, or synaptopodin. HG reduces BMP7 secretion and activity but does not affect BMP receptor levels in murine podocytes. In these cells, BMP7 effectively activates smad5 (but not smad1) and raises p38 phosphorylation [which is also increased by transforming growth factor-beta (TGF-beta)]. HG as well as TGF-beta raise caspase-3 activity, increase apoptosis, and reduce cell survival which is, in part, blocked by BMP7. Knockdown and forced expression studies indicate that smad5 is required as well as sufficient for these actions of BMP7. These findings indicate that BMP7 is a differentiation and survival factor for podocytes, requires smad5, and can reduce diabetic podocyte injury.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Citoproteção , Diabetes Mellitus/patologia , Rim/patologia , Podócitos/metabolismo , Podócitos/patologia , Proteína Smad5/metabolismo , Actinas/metabolismo , Animais , Comunicação Autócrina , Proteína Morfogenética Óssea 7 , Caspase 3/metabolismo , Diferenciação Celular , Forma Celular/efeitos dos fármacos , Sobrevivência Celular , Células Cultivadas , Citoesqueleto/metabolismo , Nefropatias Diabéticas/prevenção & controle , Relação Dose-Resposta a Droga , Regulação para Baixo , Ativação Enzimática/efeitos dos fármacos , Glucose/administração & dosagem , Glucose/farmacologia , Camundongos , Fenótipo , Fosforilação , Podócitos/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/genética , Fator de Crescimento Transformador beta/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
J Mol Graph Model ; 24(4): 271-7, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16243555

RESUMO

Members of the Smad protein family function as signal transducers of the transforming growth factor (TGF-beta) superfamily proteins. The human Smad5 protein, a signal transducer downstream of TGF-beta/BMP receptors, is composed of N-terminal DNA binding domain (MH1) and C-terminal protein-protein interaction domain (MH2) connected together by a linker motif. We used homology-modeling techniques to generate a reliable molecular model of the Smad5 MH1 domain based on the crystal structure of Smad3 MH1 domain. Our study presents the structural features of a BMP-regulated, R-Smad subfamily member (consisting of Smad1, Smad5 and Smad8) for the first time. This model provides a structural basis for explaining both functional similarities and differences between Smad3 and Smad5. Also, the structural model of this molecular target would be useful for structure-based inhibitor design because of its high accuracy. The results of our study provide important insights into understanding the structure-function relationship of the members of the Smad protein family and can serve to guide future genetic and biochemical experiments in this area.


Assuntos
DNA/metabolismo , Desenho de Fármacos , Modelos Moleculares , Proteína Smad5/antagonistas & inibidores , Proteína Smad5/química , Sequência de Aminoácidos , Sequência Conservada , Humanos , Dados de Sequência Molecular , Filogenia , Estrutura Terciária de Proteína , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Proteína Smad5/genética , Proteína Smad5/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA