Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 384
Filtrar
1.
Biomed Pharmacother ; 174: 116503, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38565060

RESUMEN

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.


Asunto(s)
Alopecia , Cuernos de Venado , Proteína Morfogenética Ósea 4 , Folículo Piloso , Cabello , Animales , Cuernos de Venado/química , Alopecia/tratamiento farmacológico , Alopecia/patología , Folículo Piloso/efectos de los fármacos , Folículo Piloso/metabolismo , Ratones , Masculino , Proteína Morfogenética Ósea 4/metabolismo , Cabello/efectos de los fármacos , Cabello/crecimiento & desarrollo , Proteínas Hedgehog/metabolismo , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proliferación Celular/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Extractos de Tejidos/farmacología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Regeneración/efectos de los fármacos , Ciervos , Proteína Smad5/metabolismo
2.
Cell Rep ; 43(5): 114162, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38678558

RESUMEN

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.


Asunto(s)
Proteína Morfogenética Ósea 7 , Proliferación Celular , Miocitos Cardíacos , Regeneración , Pez Cebra , Animales , Pez Cebra/metabolismo , Miocitos Cardíacos/metabolismo , Proteína Morfogenética Ósea 7/metabolismo , Proteína Morfogenética Ósea 7/genética , Ratones , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Neurregulina-1/metabolismo , Neurregulina-1/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Ratones Endogámicos C57BL , Proteína Smad5/metabolismo
3.
Elife ; 122024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38536963

RESUMEN

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.


Asunto(s)
Endometrio , Útero , Embarazo , Femenino , Humanos , Ratones , Animales , Útero/metabolismo , Endometrio/metabolismo , Transducción de Señal/fisiología , Implantación del Embrión , Proteína Smad5/genética , Proteína Smad5/metabolismo
4.
Oral Health Prev Dent ; 21(1): 211-218, 2023 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-37272598

RESUMEN

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.


Asunto(s)
MicroARNs , Pulpitis , Humanos , Pulpitis/metabolismo , Interleucina-6/metabolismo , Interleucina-6/farmacología , Lipopolisacáridos/farmacología , Pulpa Dental/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Proteína Smad5/metabolismo , Proteína Smad5/farmacología
5.
J Biol Chem ; 298(12): 102684, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36370851

RESUMEN

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.


Asunto(s)
Transducción de Señal , Proteína Smad1 , Proteína Smad5 , Animales , Humanos , Ratones , Proteína Morfogenética Ósea 2/metabolismo , Diferenciación Celular , Línea Celular , Proteínas de la Membrana , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/genética , Estabilidad Proteica , Transducción de Señal/genética , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteína Smad8/genética , Proteína Smad8/metabolismo
6.
Sci Rep ; 12(1): 16310, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36175474

RESUMEN

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.


Asunto(s)
Adenocarcinoma del Pulmón , Adenocarcinoma , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Proteína Smad5/metabolismo , Adenocarcinoma/genética , Animales , Proteína Morfogenética Ósea 2 , Carcinoma de Pulmón de Células no Pequeñas/genética , Humanos , Neoplasias Pulmonares/genética , Metástasis Linfática , Ratones , Proteínas Proto-Oncogénicas p21(ras) , Proteína Smad1
7.
J Biol Chem ; 298(9): 102297, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35872017

RESUMEN

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.


Asunto(s)
Endoglina , Factor 2 de Diferenciación de Crecimiento , Insulina , Neovascularización Patológica , Proteínas del Tejido Nervioso , Receptores de Factores de Crecimiento Transformadores beta , Animales , Humanos , Ratones , Receptores de Activinas Tipo II/metabolismo , Chlorocebus aethiops , Células COS , Endoglina/genética , Endoglina/metabolismo , Factor 2 de Diferenciación de Crecimiento/genética , Insulina/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fosfatidilinositol 3-Quinasas , Proteómica , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
8.
Biochim Biophys Acta Mol Cell Res ; 1869(10): 119316, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35724807

RESUMEN

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.


Asunto(s)
Aborto Espontáneo , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Aborto Espontáneo/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Animales , Desmetilación , Femenino , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Ratones , Embarazo , ARN Mensajero/genética , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Trofoblastos/metabolismo
9.
PLoS One ; 17(5): e0266409, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35580109

RESUMEN

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.


Asunto(s)
Haptoglobinas , Neoplasias Hepáticas , Proteínas Smad , Factor de Crecimiento Transformador beta , Línea Celular Tumoral , Movimiento Celular , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal , Haptoglobinas/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Precursores de Proteínas/metabolismo , Transducción de Señal , Proteínas Smad/metabolismo , Proteína Smad1/metabolismo , Proteína smad3/metabolismo , Proteína Smad5/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
10.
Bioengineered ; 13(5): 12350-12364, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35599595

RESUMEN

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.


Asunto(s)
Glioma , Proteínas Inhibidoras de la Diferenciación , Proteínas Circadianas Period , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Glioma/metabolismo , Humanos , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Ratones , Ratones Desnudos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero , Proteína Smad5/genética , Proteína Smad5/metabolismo
11.
Ren Fail ; 44(1): 191-203, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35170385

RESUMEN

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.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , MicroARNs/metabolismo , Fibrosis Peritoneal/metabolismo , Proteína Smad5/metabolismo , Animales , Cadherinas/metabolismo , Humanos , Hibridación Fluorescente in Situ , Modelos Animales , Diálisis Peritoneal/efectos adversos , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo , Vimentina/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo
12.
Gene ; 819: 146220, 2022 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-35093446

RESUMEN

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.


Asunto(s)
Bovinos/genética , Bovinos/metabolismo , Estudios de Asociación Genética , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Proteína Smad1/genética , Proteína Smad5/genética , Alelos , Animales , Tamaño Corporal/genética , Pesos y Medidas Corporales , Genotipo , Haplotipos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo
13.
Bioengineered ; 13(1): 1447-1458, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34974806

RESUMEN

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.


Asunto(s)
Queloide/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Proteína Smad5/genética , Movimiento Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Citoplasma/genética , Citoplasma/metabolismo , Progresión de la Enfermedad , Regulación hacia Abajo , Fibroblastos/química , Fibroblastos/citología , Humanos , Queloide/metabolismo , Cultivo Primario de Células , Proteína Smad5/metabolismo
14.
Hum Cell ; 35(1): 83-97, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34585365

RESUMEN

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.


Asunto(s)
Exosomas/trasplante , Riñón/patología , Células Madre Mesenquimatosas/citología , MicroARNs/administración & dosificación , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/terapia , Animales , Apoptosis/genética , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Transición Epitelial-Mesenquimal/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibrosis , Ratones , MicroARNs/metabolismo , Ratas , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/patología , Proteína Smad5/genética , Proteína Smad5/metabolismo , Obstrucción Ureteral/complicaciones
15.
Cells ; 10(12)2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34944071

RESUMEN

Chronic venous diseases, including varicose veins, are characterized by hemodynamic disturbances due to valve defects, venous insufficiency, and orthostatism. Veins are physiologically low shear stress systems, and how altered hemodynamics drives focal endothelial dysfunction and causes venous remodeling is unknown. Here we demonstrate the occurrence of endothelial to mesenchymal transition (EndMT) in human varicose veins. Moreover, the BMP4-pSMAD5 pathway was robustly upregulated in varicose veins. In vitro flow-based assays using human vein, endothelial cells cultured in microfluidic chambers show that even minimal disturbances in shear stress as may occur in early stages of venous insufficiency induce BMP4-pSMAD5-based phenotype switching. Furthermore, low shear stress at uniform laminar pattern does not induce EndMT in venous endothelial cells. Targeting the BMP4-pSMAD5 pathway with small molecule inhibitor LDN193189 reduced SNAI1/2 expression in venous endothelial cells exposed to disturbed flow. TGFß inhibitor SB505124 was less efficient in inhibiting EndMT in venous endothelial cells exposed to disturbed flow. We conclude that disturbed shear stress, even in the absence of any oscillatory flow, induces EndMT in varicose veins via activation of BMP4/pSMAD5-SNAI1/2 signaling. The present findings serve as a rationale for the possible use of small molecular mechanotherapeutics in the management of varicose veins.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Células Endoteliales/patología , Mesodermo/patología , Transducción de Señal , Proteína Smad5/metabolismo , Estrés Mecánico , Várices/metabolismo , Várices/patología , Adulto , Anciano , Biomarcadores/metabolismo , Células Endoteliales/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Neointima/patología , Fosforilación/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Reología/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Adulto Joven
16.
Cell Transplant ; 30: 9636897211052959, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34693745

RESUMEN

Dental pulp stem cells (DPSCs) are one promising cell source of mesenchymal stem cells in bone tissue engineering. However, it remains unknown that the molecules and signaling pathways involved in osteogenesis of DPSCs. Hence, this study investigated the functional roles and underlying mechanisms of circRFWD2 during osteogenesis of DPSCs. Knockdown of circRFWD2 suppressed osteogenesis of DPSCs significantly. Mechanistically, circRFWD2 could crosstalk with miR-6817-5p, which was an inhibitor of DPSCs osteogenesis. MiR-6817-5p functioned as a sponge of BMPR2, which regulated the phosphorylation of Smad5 and p38 to impact osteogenesis activity of DPSCs. Collectively, circRFWD2/miR-6817-5p/BMPR2 axis could regulate DPSCs osteogenesis via BMP-Smad and p38 MAPK pathway, which are novel mechanisms in the osteogenic differentiation of DPSCs and suggest potential therapeutic methods for bone defects regeneration.


Asunto(s)
Pulpa Dental/metabolismo , Osteogénesis/genética , ARN Circular/genética , Proteína Smad5/metabolismo , Células Madre/metabolismo , Ingeniería de Tejidos/métodos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Diferenciación Celular , Humanos , Transfección
17.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34445177

RESUMEN

Adipose tissues (AT) expand in response to energy surplus through adipocyte hypertrophy and hyperplasia. The latter, also known as adipogenesis, is a process by which multipotent precursors differentiate to form mature adipocytes. This process is directed by developmental cues that include members of the TGF-ß family. Our goal here was to elucidate, using the 3T3-L1 adipogenesis model, how TGF-ß family growth factors and inhibitors regulate adipocyte development. We show that ligands of the Activin and TGF-ß families, several ligand traps, and the SMAD1/5/8 signaling inhibitor LDN-193189 profoundly suppressed 3T3-L1 adipogenesis. Strikingly, anti-adipogenic traps and ligands engaged the same mechanism of action involving the simultaneous activation of SMAD2/3 and inhibition of SMAD1/5/8 signaling. This effect was rescued by the SMAD2/3 signaling inhibitor SB-431542. By contrast, although LDN-193189 also suppressed SMAD1/5/8 signaling and adipogenesis, its effect could not be rescued by SB-431542. Collectively, these findings reveal the fundamental role of SMAD1/5/8 for 3T3-L1 adipogenesis, and potentially identify a negative feedback loop that links SMAD2/3 activation with SMAD1/5/8 inhibition in adipogenic precursors.


Asunto(s)
Adipogénesis , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Células 3T3-L1 , Animales , Ratones , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo
18.
Int J Mol Sci ; 22(16)2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-34445305

RESUMEN

Pulmonary arterial hypertension (PAH) is characterized by pulmonary vascular remodeling. Recent evidence supports that inflammation plays a key role in triggering and maintaining pulmonary vascular remodeling. Recent studies have shown that garlic extract has protective effects in PAH, but the precise role of allicin, a compound derived from garlic, is unknown. Thus, we used allicin to evaluate its effects on inflammation and fibrosis in PAH. Male Wistar rats were divided into three groups: control (CON), monocrotaline (60 mg/kg) (MCT), and MCT plus allicin (16 mg/kg/oral gavage) (MCT + A). Right ventricle (RV) hypertrophy and pulmonary arterial medial wall thickness were determined. IL-1ß, IL-6, TNF-α, NFκB p65, Iκß, TGF-ß, and α-SMA were determined by Western blot analysis. In addition, TNF-α and TGF-ß were determined by immunohistochemistry, and miR-21-5p and mRNA expressions of Cd68, Bmpr2, and Smad5 were determined by RT-qPCR. Results: Allicin prevented increases in vessel wall thickness due to TNF-α, IL-6, IL-1ß, and Cd68 in the lung. In addition, TGF-ß, α-SMA, and fibrosis were lower in the MCT + A group compared with the MCT group. In the RV, allicin prevented increases in TNF-α, IL-6, and TGF-ß. These observations suggest that, through the modulation of proinflammatory and profibrotic markers in the lung and heart, allicin delays the progression of PAH.


Asunto(s)
Antiinflamatorios/uso terapéutico , Disulfuros/uso terapéutico , Hipertensión Pulmonar/tratamiento farmacológico , Ácidos Sulfínicos/uso terapéutico , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Citocinas/genética , Citocinas/metabolismo , Fibrosis , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Masculino , FN-kappa B/genética , FN-kappa B/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Ratas , Ratas Wistar , Proteína Smad5/genética , Proteína Smad5/metabolismo
19.
Dev Biol ; 478: 183-204, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34216573

RESUMEN

The mechanisms regulating nervous system development are still unknown for a wide variety of taxa. In insects and vertebrates, bone morphogenetic protein (BMP) signaling plays a key role in establishing the dorsal-ventral (D-V) axis and limiting the neuroectoderm to one side of that axis, leading to speculation about the conserved evolution of centralized nervous systems. Studies outside of insects and vertebrates show a more diverse picture of what, if any role, BMP signaling plays in neural development across Bilateria. This is especially true in the morphologically diverse Spiralia (≈Lophotrochozoa). Despite several studies of D-V axis formation and neural induction in spiralians, there is no consensus for how these two processes are related, or whether BMP signaling may have played an ancestral role in either process. To determine the function of BMP signaling during early development of the spiralian annelid Capitella teleta, we incubated embryos and larvae in BMP4 protein for different amounts of time. Adding exogenous BMP protein to early-cleaving C. teleta embryos had a striking effect on formation of the brain, eyes, foregut, and ventral midline in a time-dependent manner. However, adding BMP did not block brain or VNC formation or majorly disrupt the D-V axis. We identified three key time windows of BMP activity. 1) BMP treatment around birth of the 3rd-quartet micromeres caused the loss of the eyes, radialization of the brain, and a reduction of the foregut, which we interpret as a loss of A- and C-quadrant identities with a possible trans-fate switch to a D-quadrant identity. 2) Treatment after the birth of micromere 4d induced formation of a third ectopic brain lobe, eye, and foregut lobe, which we interpret as a trans-fate switch of B-quadrant micromeres to a C-quadrant identity. 3) Continuous BMP treatment from late cleavage (4d â€‹+ â€‹12 â€‹h) through mid-larval stages resulted in a modest expansion of Ct-chrdl expression in the dorsal ectoderm and a concomitant loss of the ventral midline (neurotroch ciliary band). Loss of the ventral midline was accompanied by a collapse of the bilaterally-symmetric ventral nerve cord, although the total amount of neural tissue was not greatly affected. Our results compared with those from other annelids and molluscs suggest that BMP signaling was not ancestrally involved in delimiting neural tissue to one region of the D-V axis. However, the effects of ectopic BMP on quadrant-identity during cleavage stages may represent a non-axial organizing signal that was present in the last common ancestor of annelids and mollusks. Furthermore, in the last common ancestor of annelids, BMP signaling may have functioned in patterning ectodermal fates along the D-V axis in the trunk. Ultimately, studies on a wider range of spiralian taxa are needed to determine the role of BMP signaling during neural induction and neural patterning in the last common ancestor of this group. Ultimately, these comparisons will give us insight into the evolutionary origins of centralized nervous systems and body plans.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Proteínas Morfogenéticas Óseas/metabolismo , Poliquetos/embriología , Poliquetos/metabolismo , Proteínas de Pez Cebra/farmacología , Animales , Tipificación del Cuerpo/efectos de los fármacos , Proteínas Morfogenéticas Óseas/genética , Encéfalo/embriología , Sistema Digestivo/embriología , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Ojo/embriología , Proteínas del Tejido Nervioso/metabolismo , Sistema Nervioso/embriología , Poliquetos/efectos de los fármacos , Poliquetos/crecimiento & desarrollo , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteína Smad8/genética , Proteína Smad8/metabolismo
20.
Bull Exp Biol Med ; 171(3): 305-311, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34302205

RESUMEN

We studied the effects and mechanisms of action of conophylline in different concentrations in the original in vitro model of myocardial fibrosis (treatment of cardiac fibroblasts isolated form the hearts of newborn rats with angiotensin II). Viability, collagen content, and expression of related protein in cardiac fibroblasts were assessed using the MTT-test, Sircol assay, and Western blotting, respectively. Conophylline markedly protected the cultured cells against the development of angiotensin II-induced fibrosis, which was seen from reduced viability of fibroblasts, decreased collagen content, and down-regulation of the expression of α-smooth muscle actin (α-SMA). Conophylline did not affect the TGF-ß pathway altered by angiotensin II, but markedly decreased the level of bone morphogenetic protein-4 (BMP4) enhanced by angiotensin II and BMP4 itself. Conophylline produced no effect on phosphorylation of α-SMA and Smad homologue-1/5/8, the classic BMP4 downstream pathway elements, but reduced the level of c-Jun N-terminal kinase (JNK) elevated by BMP4. Conophylline did not inhibit the development of myocardial fibrosis in the presence of JNK activator anisomycin. Thus, conophylline inhibited angiotensin II-provoked myocardial fibrosis via the BMP4/JNK pathway.


Asunto(s)
Angiotensina II/farmacología , Antifibróticos/farmacología , Proteína Morfogenética Ósea 4/genética , Fibroblastos/efectos de los fármacos , MAP Quinasa Quinasa 4/genética , Alcaloides de la Vinca/farmacología , Animales , Animales Recién Nacidos , Proteína Morfogenética Ósea 4/antagonistas & inhibidores , Proteína Morfogenética Ósea 4/metabolismo , Colágeno/genética , Colágeno/metabolismo , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/metabolismo , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/prevención & control , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación de la Expresión Génica , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , Modelos Biológicos , Miocardio/metabolismo , Miocardio/patología , Fosforilación/efectos de los fármacos , Cultivo Primario de Células , Ratas , Ratas Wistar , Transducción de Señal , Proteína Smad1/genética , Proteína Smad1/metabolismo , Proteína Smad5/genética , Proteína Smad5/metabolismo , Proteína Smad8/genética , Proteína Smad8/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA