Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 390
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
J Orthop Surg Res ; 19(1): 444, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39075522

RESUMO

BACKGROUND: Dysregulation of osteogenic differentiation is a crucial event during osteoporosis. The bioactive phytochemical icariin has become an anti-osteoporosis candidate. Here, we elucidated the mechanisms underlying the promoting function of icariin in osteogenic differentiation. METHODS: Murine pre-osteoblast MC3T3-E1 cells were stimulated with dexamethasone (DEX) to induce osteogenic differentiation, which was evaluated by an Alizarin Red staining assay and ALP activity measurement. The mRNA amounts of SPI1 and SMAD5 were detected by real-time quantitative PCR. Expression analysis of proteins, including osteogenic markers (OPN, OCN and RUNX2) and autophagy-associated proteins (LC3, Beclin-1, and ATG5), was performed by immunoblotting. The binding of SPI1 and the SMAD5 promoter was predicted by the Jaspar2024 algorithm and confirmed by chromatin immunoprecipitation (ChIP) experiments. The regulation of SPI1 in SMAD5 was examined by luciferase assays. RESULTS: During osteogenic differentiation of MC3T3-E1 cells, SPI1 and SMAD5 were upregulated. Functionally, SPI1 overexpression enhanced autophagy and osteogenic differentiation of MC3T3-E1 cells, while SMAD5 downregulation exhibited opposite effects. Mechanistically, SPI1 could enhance SMAD5 transcription and expression. Downregulation of SMAD5 also reversed SPI1 overexpression-induced autophagy and osteogenic differentiation in MC3T3-E1 cells. In MC3T3-E1 cells under DEX stimulation, icariin increased SMAD5 expression by upregulating SPI1. Furthermore, icariin could attenuate SPI1 depletion-imposed inhibition of autophagy and osteogenic differentiation of MC3T3-E1 cells. CONCLUSION: Our findings demonstrate that the SPI1/SMAD5 cascade, with the ability to enhance osteogenic differentiation, underlies the promoting effect of icariin on osteogenic differentiation of MC3T3-E1 cells.


Assuntos
Diferenciação Celular , Flavonoides , Osteoblastos , Osteogênese , Proteína Smad5 , Flavonoides/farmacologia , Animais , Camundongos , Osteogênese/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proteína Smad5/metabolismo , Proteína Smad5/genética , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Autofagia/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células 3T3
2.
Bone ; 187: 117175, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38917963

RESUMO

While previous studies have demonstrated the role of ubiquitin-conjugating enzyme 2C (UBE2C) in promoting ß-cell proliferation and cancer cell lineage expansion, its specific function and mechanism in bone marrow mesenchymal stem/stromal cells (BMSCs) growth and differentiation remain poorly understood. Our findings indicate that mice with conditional Ube2c deletions in BMSCs and osteoblasts exhibit reduced skeletal bone mass and impaired bone repair. A significant reduction in the proliferative capacity of BMSCs was observed in conditional Ube2c knockout mice, with no effect on apoptosis. Additionally, conditional Ube2c knockout mice exhibited enhanced osteoclastic activity and reduced osteogenic differentiation. Furthermore, human BMSCs with stable UBE2C knockdown exhibited diminished capacity for osteogenic differentiation. Mechanistically, we discovered that UBE2C binds to and stabilizes SMAD1/5 protein expression levels. Interestingly, UBE2C's role in regulating osteogenic differentiation and SMAD1/5 expression levels appears to be independent of its enzymatic activity. Notably, UBE2C regulates osteogenic differentiation through SMAD1/5 signaling. In conclusion, our findings underscore the pivotal role of UBE2C in bone formation, emphasizing its contribution to enhanced osteogenic differentiation through the stabilization of SMAD1/5. These results propose UBE2C as a promising target for BMSC-based bone regeneration.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais , Camundongos Knockout , Osteogênese , Proteína Smad1 , Proteína Smad5 , Enzimas de Conjugação de Ubiquitina , Animais , Osteogênese/fisiologia , Proteína Smad1/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Humanos , Diferenciação Celular/fisiologia , Proteína Smad5/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Transdução de Sinais , Proliferação de Células , Estabilidade Proteica , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoclastos/citologia
3.
Cell Metab ; 36(6): 1169-1171, 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38838638

RESUMO

In this issue of Cell Metabolism, Fang et al.1 report a novel pH-sensitive cellular signaling mechanism involving the transcription factor SMAD5 that regulates the vesicular secretion of insulin from pancreatic ß cells in response to dietary challenges. Dysregulation of this pathway may contribute to metabolic disorders such as type 2 diabetes mellitus.


Assuntos
Secreção de Insulina , Células Secretoras de Insulina , Insulina , Transdução de Sinais , Proteína Smad5 , Insulina/metabolismo , Animais , Células Secretoras de Insulina/metabolismo , Proteína Smad5/metabolismo , Humanos , Diabetes Mellitus Tipo 2/metabolismo , Camundongos , Concentração de Íons de Hidrogênio
4.
Vascul Pharmacol ; 155: 107381, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38795838

RESUMO

AIMS: Bone morphogenetic protein-9 (BMP9) is critical for bone morphogenetic protein receptor type-2 (BMPR2) signalling in pulmonary vascular endothelial cells. Furthermore, human genetics studies support the central role of disrupted BMPR2 mediated BMP9 signalling in vascular endothelial cells in the initiation of pulmonary arterial hypertension (PAH). In addition, loss-of-function mutations in BMP9 have been identified in PAH patients. BMP9 is considered to play an important role in vascular homeostasis and quiescence. METHODS AND RESULTS: We identified a novel BMP9 target as the class-3 semaphorin, SEMA3G. Although originally identified as playing a role in neuronal development, class-3 semaphorins may have important roles in endothelial function. Here we show that BMP9 transcriptional regulation of SEMA3G occurs via ALK1 and the canonical Smad pathway, requiring both Smad1 and Smad5. Knockdown studies demonstrated redundancy between type-2 receptors in that BMPR2 and ACTR2A were compensatory. Increased SEMA3G expression by BMP9 was found to be regulated by the transcription factor, SOX17. Moreover, we observed that SEMA3G regulates VEGF signalling by inhibiting VEGFR2 phosphorylation and that VEGF, in contrast to BMP9, negatively regulated SEMA3G transcription. Functional endothelial cell assays of VEGF-mediated migration and network formation revealed that BMP9 inhibition of VEGF was abrogated by SEMA3G knockdown. Conversely, treatment with recombinant SEMA3G partially mimicked the inhibitory action of BMP9 in these assays. CONCLUSIONS: This study provides further evidence for the anti-angiogenic role of BMP9 in microvascular endothelial cells and these functions are mediated at least in part via SOX17 and SEMA3G induction.


Assuntos
Movimento Celular , Células Endoteliais , Fator 2 de Diferenciação de Crescimento , Semaforinas , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular , Humanos , Movimento Celular/efeitos dos fármacos , Semaforinas/metabolismo , Semaforinas/genética , Fator 2 de Diferenciação de Crescimento/genética , Fator 2 de Diferenciação de Crescimento/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Proteína Smad5/metabolismo , Proteína Smad5/genética , Receptores de Ativinas Tipo I/metabolismo , Receptores de Ativinas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Proteína Smad1/metabolismo , Proteína Smad1/genética , Pulmão/metabolismo , Pulmão/irrigação sanguínea , Neovascularização Fisiológica/efeitos dos fármacos , Células Cultivadas
5.
Am J Physiol Cell Physiol ; 327(1): C124-C139, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38766767

RESUMO

Protein synthesis regulation is critical for skeletal muscle hypertrophy, yet other established cellular processes are necessary for growth-related cellular remodeling. Autophagy has a well-acknowledged role in muscle quality control, but evidence for its role in myofiber hypertrophy remains equivocal. Both mammalian target of rapamycin complex I (mTORC1) and bone morphogenetic protein (BMP)-Smad1/5 (Sma and Mad proteins from Caenorhabditis elegans and Drosophila, respectively) signaling are reported regulators of myofiber hypertrophy; however, gaps remain in our understanding of how this regulation is integrated with growth processes and autophagy regulation. Therefore, we investigated the mTORC1 and Smad1/5 regulation of protein synthesis and autophagy flux during serum-stimulated myotube growth. Chronic serum stimulation experiments were performed on day 5 differentiated C2C12 myotubes incubated in differentiation medium [2% horse serum (HS)] or growth medium [5% fetal bovine serum (FBS)] for 48 h. Rapamycin or LDN193189 was dosed for 48 h to inhibit mTORC1 and BMP-Smad1/5 signaling, respectively. Acute serum stimulation was examined in day 7 differentiated myotubes. Protein synthesis was measured by puromycin incorporation. Bafilomycin A1 and immunoblotting for LC3B were used to assess autophagy flux. Chronic serum stimulation increased myotube diameter 22%, total protein 21%, total RNA 100%, and Smad1/5 phosphorylation 404% and suppressed autophagy flux. Rapamycin, but not LDN193189, blocked serum-induced myotube hypertrophy and the increase in total RNA. Acute serum stimulation increased protein synthesis 111%, Smad1/5 phosphorylation 559%, and rpS6 phosphorylation 117% and suppressed autophagy flux. Rapamycin increased autophagy flux during acute serum stimulation. These results provide evidence for mTORC1, but not BMP-Smad1/5, signaling being required for serum-induced myotube hypertrophy and autophagy flux by measuring LC3BII/I expression. Further investigation is warranted to examine the role of autophagy flux in myotube hypertrophy.NEW & NOTEWORTHY The present study demonstrates that myotube hypertrophy caused by chronic serum stimulation requires mammalian target of rapamycin complex 1 (mTORC1) signaling but not bone morphogenetic protein (BMP)-Smad1/5 signaling. The suppression of autophagy flux was associated with serum-induced myotube hypertrophy and mTORC1 regulation of autophagy flux by measuring LC3BII/I expression. Rapamycin is widely investigated for beneficial effects in aging skeletal muscle and sarcopenia; our results provide evidence that rapamycin can regulate autophagy-related signaling during myotube growth, which could benefit skeletal muscle functional and metabolic health.


Assuntos
Autofagia , Hipertrofia , Alvo Mecanístico do Complexo 1 de Rapamicina , Fibras Musculares Esqueléticas , Transdução de Sinais , Animais , Camundongos , Autofagia/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Hipertrofia/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Soro/metabolismo , Proteína Smad1/metabolismo , Proteína Smad1/genética , Proteína Smad5/metabolismo , Proteína Smad5/genética
6.
Cell Rep ; 43(5): 114162, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38678558

RESUMO

Zebrafish have a lifelong cardiac regenerative ability after damage, whereas mammals lose this capacity during early postnatal development. This study investigated whether the declining expression of growth factors during postnatal mammalian development contributes to the decrease of cardiomyocyte regenerative potential. Besides confirming the proliferative ability of neuregulin 1 (NRG1), interleukin (IL)1b, receptor activator of nuclear factor kappa-Β ligand (RANKL), insulin growth factor (IGF)2, and IL6, we identified other potential pro-regenerative factors, with BMP7 exhibiting the most pronounced efficacy. Bmp7 knockdown in neonatal mouse cardiomyocytes and loss-of-function in adult zebrafish during cardiac regeneration reduced cardiomyocyte proliferation, indicating that Bmp7 is crucial in the regenerative stages of mouse and zebrafish hearts. Conversely, bmp7 overexpression in regenerating zebrafish or administration at post-mitotic juvenile and adult mouse stages, in vitro and in vivo following myocardial infarction, enhanced cardiomyocyte cycling. Mechanistically, BMP7 stimulated proliferation through BMPR1A/ACVR1 and ACVR2A/BMPR2 receptors and downstream SMAD5, ERK, and AKT signaling. Overall, BMP7 administration is a promising strategy for heart regeneration.


Assuntos
Proteína Morfogenética Óssea 7 , Proliferação de Células , Miócitos Cardíacos , Regeneração , Peixe-Zebra , Animais , Feminino , Masculino , Camundongos , Proteína Morfogenética Óssea 7/metabolismo , Proteína Morfogenética Óssea 7/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Neuregulina-1/metabolismo , Neuregulina-1/genética , Transdução de Sinais , Proteína Smad5/metabolismo , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética
7.
Eur J Clin Invest ; 54(8): e14212, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38591651

RESUMO

BACKGROUND: Bone morphogenetic protein 9 (BMP9) is a hepatokine that plays a pivotal role in the progression of liver diseases. Moreover, an increasing number of studies have shown that BMP9 is associated with hepatopulmonary syndrome (HPS), but its role in HPS is unclear. Here, we evaluated the influence of CBDL on BMP9 expression and investigated potential mechanisms of BMP9 signalling in HPS. METHODS: We profiled the circulating BMP9 levels in common bile duct ligation-induced HPS rat model, and then investigated the effects and mechanisms of HPS rat serum on pulmonary vascular endothelial dysfunction in rat model, as well as in primarily cultured rat pulmonary microvascular endothelial cells. RESULTS: Our data revealed that circulating BMP9 levels were significantly increased in the HPS rats compared to control group. Besides, the elevated BMP9 in HPS rat serum was not only crucial for promoting endothelial cell proliferation and tube formation through the activin receptor-like kinase1 (ALK1)-Endoglin-Smad1/5/9 pathway, but also important for accumulation of monocytes. Treatments with ALK1-Fc or silencing ALK1 expression to inhibit the BMP9 signalling pathway effectively eliminated these effects. In agreement with these observations, increased circulating BMP9 was associated with an increase in lung vessel density and accumulation of pro-angiogenic monocytes in the microvasculature in HPS rats. CONCLUSIONS: This study provided evidence that elevated circulating BMP9, secreted from the liver, promote pulmonary angiogenesis in HPS rats via ALK1-Endoglin-Smad1/5/9 pathway. In addition, BMP9-regulated pathways are also involved in accumulation of pro-angiogenic monocytes in the pulmonary microvasculature in HPS rats.


Assuntos
Receptores de Activinas Tipo II , Endoglina , Fator 2 de Diferenciação de Crescimento , Síndrome Hepatopulmonar , Pulmão , Neovascularização Patológica , Transdução de Sinais , Proteína Smad1 , Animais , Síndrome Hepatopulmonar/metabolismo , Fator 2 de Diferenciação de Crescimento/metabolismo , Ratos , Receptores de Activinas Tipo II/metabolismo , Pulmão/metabolismo , Masculino , Proteína Smad1/metabolismo , Endoglina/metabolismo , Neovascularização Patológica/metabolismo , Células Endoteliais/metabolismo , Modelos Animais de Doenças , Proteína Smad5/metabolismo , Ratos Sprague-Dawley , Proliferação de Células , Ducto Colédoco , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Monócitos/metabolismo , Angiogênese , Receptores de Ativinas
8.
Biomed Pharmacother ; 174: 116503, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38565060

RESUMO

Androgenetic alopecia (AGA) is a prevalent disease in worldwide, local application or oral are often used to treat AGA, however, effective treatments for AGA are currently limited. In this work, we observed the promoting the initial anagen phase effect of pilose antler extract (PAE) on hair regeneration in AGA mice. We found that PAE accelerated hair growth and increased the degree of skin blackness by non-invasive in vivo methods including camera, optical coherence tomography and dermoscopy. Meanwhile, HE staining of sagittal and coronal skin sections revealed that PAE augmented the quantity and length of hair follicles, while also enhancing skin thickness and hair papilla diameter. Furthermore, PAE facilitated the shift of the growth cycle from the telogen to the anagen phase and expedited the proliferation of hair follicle stem cells and matrix cells in mice with AGA. This acceleration enabled the hair follicles to enter the growth phase at an earlier stage. PAE upregulated the expression of the sonic hedgehog (SHH), smoothened receptor, glioma-associated hemolog1 (GLI1), and downregulated the expression of bone morphogenetic protein 4 (BMP4), recombinant mothers against decapentaplegic homolog (Smad) 1 and 5 phosphorylation. This evidence suggests that PAE fosters hair growth and facilitates the transition of the growth cycle from the telogen to the anagen phase in AGA mice. This effect is achieved by enhancing the proliferation of follicle stem cells and matrix cells through the activation of the SHH/GLI pathway and suppression of the BMP/Smad pathway.


Assuntos
Alopecia , Chifres de Veado , Proteína Morfogenética Óssea 4 , Folículo Piloso , Cabelo , Animais , Chifres de Veado/química , Alopecia/tratamento farmacológico , Alopecia/patologia , Folículo Piloso/efeitos dos fármacos , Folículo Piloso/metabolismo , Camundongos , Masculino , Proteína Morfogenética Óssea 4/metabolismo , Cabelo/efeitos dos fármacos , Cabelo/crescimento & desenvolvimento , Proteínas Hedgehog/metabolismo , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína GLI1 em Dedos de Zinco/genética , Proliferação de Células/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Extratos de Tecidos/farmacologia , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Regeneração/efeitos dos fármacos , Cervos , Proteína Smad5/metabolismo
9.
Elife ; 122024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38536963

RESUMO

Endometrial decidualization, a prerequisite for successful pregnancies, relies on transcriptional reprogramming driven by progesterone receptor (PR) and bone morphogenetic protein (BMP)-SMAD1/SMAD5 signaling pathways. Despite their critical roles in early pregnancy, how these pathways intersect in reprogramming the endometrium into a receptive state remains unclear. To define how SMAD1 and/or SMAD5 integrate BMP signaling in the uterus during early pregnancy, we generated two novel transgenic mouse lines with affinity tags inserted into the endogenous SMAD1 and SMAD5 loci (Smad1HA/HA and Smad5PA/PA). By profiling the genome-wide distribution of SMAD1, SMAD5, and PR in the mouse uterus, we demonstrated the unique and shared roles of SMAD1 and SMAD5 during the window of implantation. We also showed the presence of a conserved SMAD1, SMAD5, and PR genomic binding signature in the uterus during early pregnancy. To functionally characterize the translational aspects of our findings, we demonstrated that SMAD1/5 knockdown in human endometrial stromal cells suppressed expressions of canonical decidual markers (IGFBP1, PRL, FOXO1) and PR-responsive genes (RORB, KLF15). Here, our studies provide novel tools to study BMP signaling pathways and highlight the fundamental roles of SMAD1/5 in mediating both BMP signaling pathways and the transcriptional response to progesterone (P4) during early pregnancy.


Assuntos
Endométrio , Útero , Gravidez , Feminino , Humanos , Camundongos , Animais , Útero/metabolismo , Endométrio/metabolismo , Transdução de Sinais/fisiologia , Implantação do Embrião , Proteína Smad5/genética , Proteína Smad5/metabolismo
10.
Oral Health Prev Dent ; 21(1): 211-218, 2023 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-37272598

RESUMO

PURPOSE: To research the role of microRNA (miR)-152 in the pathogenesis of pulpitis using a cell model based on human dental pulp cells (HDPCs) treated with lipopolysaccharides (LPS). MATERIALS AND METHODS: The biological activity of HDPCs infected by LPS was measured using a cell counting kit (CCK-8), Transwell test, flow cytometry, and fluorescent quantitative PCR. The concentration of superoxide dismutase (SOD) and malondialdehyde (MDA) was evaluated using an assay kit, the levels of interleukin (IL)-1ß and IL-6 were measured by enzyme-linked immunosorbent assay (ELISA), and the targeting relationship between SMAD5 and miR-152 was measured by the double-luciferase report test. The expression of cell cycle-related CyclinD1 and BAX was assessed by PCR. By plotting a receiver operating characteristic (ROC) curve, the diagnostic value of miR-152 was shown. RESULTS: The level of miR-152 in HDPCs induced by LPS decreased, while the level of SMAD5 increased. After overexpressing miR-152 in LPS-induced HDPCs, the viability was elevated, the apoptosis rate decreased, CyclinD1 was elevated, BAX diminished, the inflammatory cytokines (IL-6 and IL-1ß) were inhibited, the activity of SOD increased, and the MDA content decreased. miR-152 targeted regulation of SMAD5, and SMAD5 modulated the effects of miR-152 on cell viability, apoptosis, inflammation, and the oxidative response of HDPCs. Reduced miR-152 expression was verified in patients with pulpitis, which could be a biomarker for pulpitis. CONCLUSION: miR-152 was found to be a biomarker correlated with the pathogenesis of pulpitis and the biological behaviour of HDPCs.


Assuntos
MicroRNAs , Pulpite , Humanos , Pulpite/metabolismo , Interleucina-6/metabolismo , Interleucina-6/farmacologia , Lipopolissacarídeos/farmacologia , Polpa Dentária/metabolismo , Proteína X Associada a bcl-2/metabolismo , Proteína X Associada a bcl-2/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína Smad5/metabolismo , Proteína Smad5/farmacologia
11.
J Biol Chem ; 298(12): 102684, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36370851

RESUMO

The bone morphogenetic protein (BMP) signaling pathway plays pivotal roles in various biological processes during embryogenesis and adult homeostasis. Transmembrane anterior posterior transformation 1 (TAPT1) is an evolutionarily conserved protein involved in murine axial skeletal patterning. Genetic defects in TAPT1 result in complex lethal osteochondrodysplasia. However, the specific cellular activity of TAPT1 is not clear. Herein, we report that TAPT1 inhibits BMP signaling and destabilizes the SMAD1/5 protein by facilitating its interaction with SMURF1 E3 ubiquitin ligase, which leads to SMAD1/5 proteasomal degradation. In addition, we found that the activation of BMP signaling facilitates the redistribution of TAPT1 and promotes its association with SMAD1. TAPT1-deficient murine C2C12 myoblasts or C3H/10T1/2 mesenchymal stem cells exhibit elevated SMAD1/5/9 protein levels, which amplifies BMP activation, in turn leading to a boost in the transdifferentiation or differentiation processing of these distinct TAPT1-deficient cell lines changing into mature osteoblasts. Furthermore, the enhancing effect of TAPT1 deficiency on osteogenic differentiation of C3H/10T1/2 cells was observed in an in vivo ectopic bone formation model. Importantly, a subset of TAPT1 mutations identified in humans with lethal skeletal dysplasia exhibited gain-of-function activity on SMAD1 protein levels. Thus, this finding elucidates the role of TAPT1 in the regulation of SMAD1/5 protein stability for controlling BMP signaling.


Assuntos
Transdução de Sinais , Proteína Smad1 , Proteína Smad5 , Animais , Humanos , Camundongos , Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular , Linhagem Celular , Proteínas de Membrana , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/genética , Estabilidade Proteica , Transdução de Sinais/genética , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteína Smad8/genética , Proteína Smad8/metabolismo
12.
Sci Rep ; 12(1): 16310, 2022 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-36175474

RESUMO

Bone morphogenetic protein 2 (BMP2) is highly overexpressed in human non-small cell lung cancer (NSCLC) and correlates with tumor stage and metastatic burden. Although several lines of evidence suggest that BMP2 promotes cell migration and invasiveness in vitro, the in vivo role of BMP2 in the metastasis of lung adenocarcinoma cells remains less well understood. Here, we revealed that BMP2 is highly overexpressed in lung adenocarcinoma patients with lymph node metastasis compared with patients without lymph node metastasis. Using an in vivo orthotopic mouse model, we clearly demonstrated that BMP2 promotes lung adenocarcinoma metastasis. The depletion of BMP2 or its receptor BMPR2 significantly reduced cell migration and invasiveness. We further identified that BMP2/BMPR2-mediated cell migration involves the activation of the SMAD1/5/8 signaling pathway, independent of the KRAS signaling pathway. Significantly, the depletion of SMAD1/5/8 or the inhibition of SMAD1/5/8 by LDN193189 inhibitor significantly reduced cell migration. These findings show that BMP2 promotes NSCLC metastasis, indicating that targeting the BMP2 signaling pathway may represent a potential therapeutic strategy for treating patients with metastatic NSCLC.


Assuntos
Adenocarcinoma de Pulmão , Adenocarcinoma , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Proteína Smad5/metabolismo , Adenocarcinoma/genética , Animais , Proteína Morfogenética Óssea 2 , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Neoplasias Pulmonares/genética , Metástase Linfática , Camundongos , Proteínas Proto-Oncogênicas p21(ras) , Proteína Smad1
13.
J Biol Chem ; 298(9): 102297, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35872017

RESUMO

Insulin signaling in blood vessels primarily functions to stimulate angiogenesis and maintain vascular homeostasis through the canonical PI3K and MAPK signaling pathways. However, angiogenesis is a complex process coordinated by multiple other signaling events. Here, we report a distinct crosstalk between the insulin receptor and endoglin/activin receptor-like kinase 1 (ALK1), an endothelial cell-specific TGF-ß receptor complex essential for angiogenesis. While the endoglin-ALK1 complex normally binds to TGF-ß or bone morphogenetic protein 9 (BMP9) to promote gene regulation via transcription factors Smad1/5, we show that insulin drives insulin receptor oligomerization with endoglin-ALK1 at the cell surface to trigger rapid Smad1/5 activation. Through quantitative proteomic analysis, we identify ependymin-related protein 1 (EPDR1) as a major Smad1/5 gene target induced by insulin but not by TGF-ß or BMP9. We found endothelial EPDR1 expression is minimal at the basal state but is markedly enhanced upon prolonged insulin treatment to promote cell migration and formation of capillary tubules. Conversely, we demonstrate EPDR1 depletion strongly abrogates these angiogenic effects, indicating that EPDR1 is a crucial mediator of insulin-induced angiogenesis. Taken together, these results suggest important therapeutic implications for EPDR1 and the TGF-ß pathways in pathologic angiogenesis during hyperinsulinemia and insulin resistance.


Assuntos
Endoglina , Fator 2 de Diferenciação de Crescimento , Insulina , Neovascularização Patológica , Proteínas do Tecido Nervoso , Receptores de Fatores de Crescimento Transformadores beta , Animais , Humanos , Camundongos , Receptores de Activinas Tipo II/metabolismo , Chlorocebus aethiops , Células COS , Endoglina/genética , Endoglina/metabolismo , Fator 2 de Diferenciação de Crescimento/genética , Insulina/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Fosfatidilinositol 3-Quinases , Proteômica , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Fator de Crescimento Transformador beta/metabolismo
14.
Biochim Biophys Acta Mol Cell Res ; 1869(10): 119316, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35724807

RESUMO

The molecules induced by hypoxia have been supposed to be important regulators of first trimester trophoblast activity, but the key mechanism mediating invasion of trophoblast cells is not fully illustrated. Here, we found that the expression of RNA demethylase ALKBH5 was upregulated in trophoblast upon hypoxia treatment and decreased in extravillous trophoblast (EVT) of patients with recurrent spontaneous abortion (RSA). Furthermore, we found that trophoblast-specific knockdown of ALKBH5 in mouse placenta suppressed the invasion of trophoblast and significantly led to fetus abortion in vivo. Then ALKBH5 was identified to promote the invasion of trophoblast. Mechanistically, we identified transcripts with altered methylation in trophoblast induced by hypoxia via m6A-seq, ALKBH5 translocated from nucleus to cytoplasm upon hypoxia treatment and demethylated certain target transcripts, such as m6A-modified SMAD1/SMAD5, consequently enhanced the translation of SMAD1/SMAD5 and then promoted MMP9 and ITGA1 production. Thus, we demonstrated that ALKBH5 promoted the activity of trophoblasts by enhancing SMAD1/5 expression via erasing their m6A modifications. Our research revealed a new m6A epigenetic way to regulate the invasion of trophoblast, which suggested a novel potential therapeutic target for spontaneous abortion prevention.


Assuntos
Aborto Espontâneo , Homólogo AlkB 5 da RNA Desmetilase/metabolismo , Aborto Espontâneo/metabolismo , Homólogo AlkB 5 da RNA Desmetilase/genética , Animais , Desmetilação , Feminino , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Camundongos , Gravidez , RNA Mensageiro/genética , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Trofoblastos/metabolismo
15.
Bioengineered ; 13(5): 12350-12364, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35599595

RESUMO

In this study, we employed multiple laboratory techniques to acknowledge the biological activities and processes of Per2 and Id3 in glioma. We analyzed TCGA and CGGA databases for seeking association among Per2, Id3, and clinical features in glioma. Immunohistochemistry and Western blot were used to detect protein expression levels. CCK-8 assay, colony formation assay, Transwell assay, the wound healing assay, flow cytometric, and Xenograft nude mice were used to acknowledge the impact of Per2 and Id3 on biological behavior of glioma. The results showed that the Per2 mRNA expression was negatively correlated with the WHO grade, while the Id3 mRNA expression was positively correlated with the WHO grade in patients with glioma in TCGA and CGGA databases. Per2 and Id3 maintained separate prognostic abilities and had a negative connection in human glioma. In the clinical sample study, Per2 and Id3 were validated at the protein level with the same results compared to the mRNA expression level in TCGA and CGGA. By using a wide range of functional examples, overexpression of Per2 restrains malignant biological behaviors in glioma cells by many ways, while Id3 promotes malignant biological behaviors in glioma cells. Furthermore, overexpression of Per2 can inhibit Id3 expression via regulating PTEN/AKT/Smad5 signaling pathway and thereby abolish malignant biological behaviors that are caused by Id3 overexpression. These results suggested that Per2 inhibits glioma cell proliferation through regulating PTEN/AKT/Smad5/Id3 signaling pathway, which may be a viable therapeutic target for glioma.


Assuntos
Glioma , Proteínas Inibidoras de Diferenciação , Proteínas Circadianas Period , Animais , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Glioma/metabolismo , Humanos , Proteínas Inibidoras de Diferenciação/genética , Proteínas Inibidoras de Diferenciação/metabolismo , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro , Proteína Smad5/genética , Proteína Smad5/metabolismo
16.
PLoS One ; 17(5): e0266409, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35580109

RESUMO

Transforming growth factor-ß (TGF-ß) is an important inducer of the epithelial-to-mesenchymal transition (EMT) in various cancers. Our previous study demonstrated that prohaptoglobin (proHp) stimulates Smad1/5 activation via ALK1, a TGF-ß type I receptor, in endothelial cells, suggesting that proHp plays a role in TGF-ß signaling. However, the function of proHp in cellular events downstream of Smads remains unclear. The current study investigated the effects of proHp on TGF-ß-mediated Smad-dependent EMT induction and cell invasion in vitro using proHp-overexpressing SK-Hep1 liver cancer cells. The results of Western blotting, quantitative real-time RT-PCR, and immunocytochemistry indicated that proHp downregulated expression of mesenchymal marker and EMT regulator such as N-cadherin, vimentin, and twist, and upregulated expression of the epithelial marker E-cadherin. Compared with control cells, proHp-overexpressing cells exhibited high levels of ALK1/2/3 receptors and markedly increased Smad1/5 phosphorylation. Interestingly, proHp attenuated TGF-ß-induced expression of mesenchymal markers and Smad2/3 phosphorylation. It also significantly suppressed cell invasion and migration. Knockdown of Smad1/5 abolished the inhibitory effects of proHp on TGF-ß-stimulated Smad2/3 phosphorylation and mesenchymal marker expression. These findings indicate that proHp suppresses the TGF-ß-induced EMT and cell invasion in vitro by enhancing Smad1/5 activation via ALK1/2/3 receptors and thus suppressing the Smad2/3 signaling pathway in SK-Hep1 cells. This study suggests that proHp may prevent a de-differentiation of hepatic cells and induce a cell differentiation by regulating the Smad signaling pathway.


Assuntos
Haptoglobinas , Neoplasias Hepáticas , Proteínas Smad , Fator de Crescimento Transformador beta , Linhagem Celular Tumoral , Movimento Celular , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal , Haptoglobinas/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Precursores de Proteínas/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Proteína Smad1/metabolismo , Proteína Smad3/metabolismo , Proteína Smad5/metabolismo , Fator de Crescimento Transformador beta/metabolismo
17.
Ren Fail ; 44(1): 191-203, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35170385

RESUMO

Peritoneal fibrosis (PF) is the main reason leading to declining efficiency and ultrafiltration failure of peritoneum, which restricts the application of peritoneal dialysis (PD). We aimed to investigate the effects and mechanisms of miR-122-5p on the PF. Sprague-Dawley (SD) rats were infused with glucose-based standard PD fluid to establish PF model. HE staining was performed to evaluate the extent of PF. Real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) and fluorescence in situ hybridization (FISH) were performed to measure the expression level of miR-122-5p. Western blot was used to test the expression of transforming growth factor (TGF)-ß1, platelet-derived growth factor (PDGF)-A, Fibronectin 1 (FN1), extracellular matrix protein 1 (ECM1), Smad5, α-smooth muscle actin (SMA), collagen type 1(COL-1), Vimentin, E-Cadherin, Wnt1, ß-catenin, p-ß-catenin, c-Myc, c-Jun, and Cyclin D1. Immunohistochemistry (IHC) staining was used to detect type I collagen alpha 1 (Col1α1), α-SMA, and E-Cadherin expression. We found PF was glucose concentration-dependently enhanced in peritoneum of PD rat. The PD rats showed increased miR-122-5p and decreased Smad5 expression. MiR-122-5p silencing improved PF and epithelial-mesenchymal transition (EMT) process in PD rats. MiR-122-5p silencing attenuated the activity of the Wnt/ß-catenin signaling pathway. Importantly, dual-luciferase reporter assay showed Smad5 was a target gene of miR-122-5p. Smad5 overexpression significantly reversed the increases of PF and EMT progression induced by miR-122-5p overexpression. Moreover, miR-122-5p mimic activated Wnt/ß-catenin activity, which was blocked by Smad5 overexpression. Overall, present results demonstrated that miR-122-5p overexpression showed a deterioration effect on PD-related PF by targeting Smad5 to activate Wnt/ß-catenin pathway.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , MicroRNAs/metabolismo , Fibrose Peritoneal/metabolismo , Proteína Smad5/metabolismo , Animais , Caderinas/metabolismo , Humanos , Hibridização in Situ Fluorescente , Modelos Animais , Diálise Peritoneal/efeitos adversos , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta1/metabolismo , Vimentina/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
18.
Bioengineered ; 13(1): 1447-1458, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34974806

RESUMO

Accumulating evidence suggests that long non-coding RNAs (lncRNAs) participate in the formation and development of keloids, a benign tumor. In addition, lncRNA H19 has been shown to act on the biological processes of keloids. This study aimed to identify other important mechanisms of the effect of lncRNA H19 on keloid formation. The H19, miR-196b-5p, and SMAD family member 5 (SMAD5) expression levels were detected using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and Western blotting. Subcellular localization of lncRNA H19 was detected using a nuclear-cytoplasmic separation assay. Cell viability and proliferation were measured using counting kit-8 and colony formation assays. Bax and Bcl-2 levels were examined using Western blot analysis. The interaction between H19 and miR-196b-5p or SMAD5 was verified using a dual-luciferase reporter assay. H19 and SMAD5 expression was upregulated in keloid tissue and fibroblasts, whereas miR-196b-5p expression was downregulated. Knockdown of H19, overexpression of miR-196b-5p, or knockdown of SMAD5 inhibited the viability and proliferation of keloid fibroblasts and promoted apoptosis. Overexpression of H19 or SMAD5 and knockdown of miR-196b-5p promoted viability and proliferation and inhibited apoptosis. miR-196b-5p was identified as a H19 sponge, and SMAD5 was identified as a miR-196b-5p target. The combination of lncRNA H19 and miR-196b-5p regulates SMAD5 expression and promotes keloid formation, thus providing a new direction for keloid treatment.


Assuntos
Queloide/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Proteína Smad5/genética , Movimento Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Citoplasma/genética , Citoplasma/metabolismo , Progressão da Doença , Regulação para Baixo , Fibroblastos/química , Fibroblastos/citologia , Humanos , Queloide/metabolismo , Cultura Primária de Células , Proteína Smad5/metabolismo
19.
Gene ; 819: 146220, 2022 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-35093446

RESUMO

The SMAD1 and SMAD5 genes belong to mothers against decapentaplegic proteins family, which participate in the BMP pathway to control skeletal myogenesis and growth. In the present study, we analyzed the associations between polymorphisms of SMAD1 and SMAD5 genes promoter and important economical traits in Qinchuan cattle. Four SNPs in the SMAD1 gene promoter and three SNPs in the SMAD5 promoter were identified by sequencing of 448 Qinchuan cattles. Allelic and frequency analyses of these SNPs resulted in eight haplotypes both in the promoters of the two genes promoter and identified potential cis-regulatory transcription factor (TF) components. In addition, correlation analysis showed that cattle SMAD1 promoter activity of individuals with Hap4 (P < 0.01) was stronger than that of individuals with Hap2. while the transcriptional activity of individuals with Hap3 within SMAD5 gene promoter was significantly (P < 0.01) higher followed by H2. Uniformly, diplotypes H4-H6 of SMAD1 gene and H1-H3 of SMAD5 gene performed significant (P < 0.01) associations with body measurement and improved carcass quality traits. All these results have indicated that polymorphisms in SMAD1 and SMAD5 genes promoter could impact the transcriptional regulation and then affect muscle content in beef cattle. Moreover, both the SMAD1 and SMAD5 genes were expressed ubiquitously in 10 tissues and had higher expression in the longissimus thoracis tissue from 6-month-old and 12-month-old cattle than in cattle of other ages. We can conclude that SMAD1 and SMAD5 genes may play an important role in muscle growth and development, and the variants mapped within SMAD1 and SMAD5 genes can be utilized in molecular marker-assisted selection for cattle carcass quality and body measurement traits in breed improvement programs of Qinchuan cattle.


Assuntos
Bovinos/genética , Bovinos/metabolismo , Estudos de Associação Genética , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Proteína Smad1/genética , Proteína Smad5/genética , Alelos , Animais , Tamanho Corporal/genética , Pesos e Medidas Corporais , Genótipo , Haplótipos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo
20.
Hum Cell ; 35(1): 83-97, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34585365

RESUMO

Evidence has shown that mesenchymal stem cells' (MSCs) therapy has potential application in treating chronic kidney disease (CKD). In addition, MSCs-derived exosomes can improve the renal function and prevent the progression of CKD. However, the mechanisms by which MSCs-derived exosomes (MSCs-Exo) ameliorate renal fibrosis in CKD remain largely unclear. To mimic an in vitro model of renal fibrosis, rat kidney tubular epithelial cells (NRK52E) were stimulated with transforming growth factor (TGF)-ß1. In addition, we established an in vivo model of unilateral ureteric obstruction (UUO)-induced renal fibrosis. Meanwhile, we exploited exosomes derived from MSCs for delivering miR-186-5p agomir into NRK52E cells or kidneys in vitro and in vivo. In this study, we found that level of miR-186-5p was significantly downregulated in TGF-ß1-stimulated NRK52E cells and the obstructed kidneys of UUO mice. In addition, miR-186-5p can be transferred from MSCs to NRK52E cells via exosomes. MSCs-delivered miR-186-5p markedly reduced the accumulation of extracellular matrix (ECM) protein, and inhibited epithelial-to-mesenchymal transition (EMT) and apoptosis in TGF-ß1-stimulated NRK52E cells. Moreover, exosomal miR-186-5p from MSCs attenuated kidney injury and fibrosis in a UUO mouse model via inhibition of the ECM protein accumulation and EMT process. Meanwhile, dual-luciferase assay showed that miR-186-5p downregulated Smad5 expression via direct binding with the 3'-UTR of Smad5. Collectively then, these findings indicated that exosomal miR-186-5p derived from MSCs could attenuate renal fibrosis in vitro and in vivo by downregulation of Smad5. These findings may help to understand the role of MSCs' exosomes in alleviating renal fibrosis in CKD.


Assuntos
Exossomos/transplante , Rim/patologia , Células-Tronco Mesenquimais/citologia , MicroRNAs/administração & dosagem , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/terapia , Animais , Apoptose/genética , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Proteínas da Matriz Extracelular/metabolismo , Fibrose , Camundongos , MicroRNAs/metabolismo , Ratos , Insuficiência Renal Crônica/etiologia , Insuficiência Renal Crônica/patologia , Proteína Smad5/genética , Proteína Smad5/metabolismo , Obstrução Ureteral/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...