Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Phytomedicine ; 121: 155118, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37801895

RESUMO

BACKGROUND: With an increasing number of myocardial infarction (MI) patients, myocardial fibrosis is becoming a widespread health concern. It's becoming more and more urgent to conduct additional research and investigations into efficient treatments. Ethyl ferulate (EF) is a naturally occurring substance with cardioprotective properties. However, the extent of its impact and the underlying mechanism of its treatment for myocardial fibrosis after MI remain unknown. PURPOSE: The goal of this study was to look into how EF affected the signaling of the TGF-receptor 1 (TGFBR1) in myocardial fibrosis after MI. METHODS: Echocardiography, hematoxylin-eosin (HE) and Masson trichrome staining were employed to assess the impact of EF on heart structure and function in MI-affected mice in vivo. Cell proliferation assay (MTS), 5-Ethynyl-2'-deoxyuridine (EdU), and western blot techniques were employed to examine the influence of EF on native cardiac fibroblast (CFs) proliferation and collagen deposition. Molecular simulation and surface plasmon resonance imaging (SPRi) were utilized to explore TGFBR1 and EF interaction. Cardiac-specific Tgfbr1 knockout mice (Tgfbr1ΔMCK) were utilized to testify to the impact of EF. RESULTS: In vivo experiments revealed that EF alleviated myocardial fibrosis, improved cardiac dysfunction after MI and downregulated the TGFBR1 signaling in a dose-dependent manner. Moreover, in vitro experiments revealed that EF significantly inhibited CFs proliferation, collagen deposition and TGFBR1 signaling followed by TGF-ß1 stimulation. More specifically, molecular simulation, molecular dynamics, and SPRi collectively showed that EF directly targeted TGFBR1. Lastly, knocking down of Tgfbr1 partially reversed the inhibitory activity of EF on myocardial fibrosis in MI mice. CONCLUSION: EF attenuated myocardial fibrosis post-MI by directly suppressing TGFBR1 and its downstream signaling pathway.


Assuntos
Infarto do Miocárdio , Miocárdio , Humanos , Camundongos , Animais , Miocárdio/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/uso terapêutico , Fibroblastos/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Colágeno/metabolismo , Fibrose , Fator de Crescimento Transformador beta1/metabolismo
2.
Phytomedicine ; 114: 154779, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37023527

RESUMO

BACKGROUND: Gramine, also named 3-(N,N-dimethylaminomethyl) indole, is a indole alkaloid. It is mainly extracted from various natural raw plants. Despite being the simplest 3-aminomethylindole, Gramine has broad pharmaceutical and therapeutic effects, such as vasodilatation, antioxidation, mitochondrial bioenergetics-related effects, and angiogenesis via modulation of TGFß signaling. However, there is little information available about Gramine's role in heart disease, especially pathological cardiac hypertrophy. PURPOSE: To investigate Gramine's effect on pathological cardiac hypertrophy and clarify the mechanisms behind its action. METHODS: In the in vitro experiment, Gramine (25 µM or 50 µM) was used to investigate its role in Angiotensin II-induced primary neonatal rat cardiomyocytes (NRCMs) hypertrophy. In the in vivo experiment, Gramine (50 mg/kg or 100 mg/kg) was administrated to investigate its role in transverse aortic constriction (TAC) surgery mice. Additionally, we explored the mechanisms underlying these roles through Western blot, Real-time PCR, genome-wide transcriptomic analysis, chromatin immunoprecipitation and molecular docking studies. RESULTS: The in vitro data demonstrated that Gramine treatment obviously improved primary cardiomyocyte hypertrophy induced by Angiotensin II, but had few effects on the activation of fibroblasts. The in vivo experiments indicated that Gramine significantly mitigated TAC-induced myocardial hypertrophy, interstitial fibrosis and cardiac dysfunction. Mechanistically, RNA sequencing and further bioinformatics analysis demonstrated that transforming growth factor ß (TGFß)-related signaling pathway was enriched significantly and preferentially in Gramine-treated mice as opposed to vehicle-treated mice during pathological cardiac hypertrophy. Moreover, this cardio-protection of Gramine was found to mainly involved in TGFß receptor 1 (TGFBR1)- TGFß activated kinase 1 (TAK1)-p38 MAPK signal cascade. Further exploration showed that Gramine restrained the up-regulation of TGFBR1 by binding to Runt-related transcription factor 1 (Runx1), thereby alleviating pathological cardiac hypertrophy. CONCLUSION: Our findings provided a substantial body of evidence that Gramine possessed a potential druggability in pathological cardiac hypertrophy via suppressing the TGFBR1-TAK1-p38 MAPK signaling axis through interaction with transcription factor Runx1.


Assuntos
Angiotensina II , Subunidade alfa 2 de Fator de Ligação ao Core , Ratos , Camundongos , Animais , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Angiotensina II/farmacologia , Simulação de Acoplamento Molecular , Cardiomegalia/metabolismo , Miócitos Cardíacos , Transdução de Sinais , Alcaloides Indólicos/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
3.
Biomed Pharmacother ; 149: 112906, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-36068772

RESUMO

Delphinium trichophorum Franch (DTF), a species endemic to China, has been widely used for centuries in Tibet as an indigenous medicine for treating cough, pneumonia, and pulmonary fibrosis. Hetisine-type C20-diterpenoid alkaloids have been reported to be characteristic and active ingredients. Herein, five ones with relatively high contents in D. trichophorum, including 2α,11α,13ß-triacetylhetisine (DTF1), trichodelphinine A (DTF2), trichodelphinine D (DTF3), 2α-acetyl-11α,13ß-dihydroxyhetisine (DTF4), and trichodelphinine C (DTF5), were investigated for anti-fibrosis effects using fibroblasts induced by TGF-ß1 or LPS for the first time. The results showed that all five tested compounds decreased hydroxyproline (HYP) levels and inhibited the abnormal proliferation of 3T6 and HFL-1 cells induced by either TGF-ß1 or LPS. Moreover, DTF1 and DTF2 attenuated the production of collagen (Col-1 and Col-3) at relatively low doses, suggesting their higher efficiency among the five alkaloids. Based on large-scale ligand-based pharmacophore modeling, TGFBR1 was screened as a potential target for these tested alkaloids. The molecular docking results also exhibited high-affinity interactions between TGFBR1 and five alkaloids, especially DTF1 and DTF2. Further experiments revealed that DTF1 and DTF2 could inhibit the expression of TGF-ß1 and α-SMA and the phosphorylation of Smad3 and Smad4 while restoring the expression of Smad7 protein. Overall, DTF1 and DTF2 may reduce collagen generation and delay the development of pulmonary fibrosis by inhibiting the activation of the TGF-ß/Smad signaling pathway. Our results provide experimental and theoretical evidence for DTF1 and DTF2 as superior candidates for further development of anti-fibrotic drugs.


Assuntos
Alcaloides , Delphinium , Diterpenos , Fibrose Pulmonar , Alcaloides/farmacologia , Alcaloides/uso terapêutico , Delphinium/metabolismo , Diterpenos/uso terapêutico , Fibrose , Lipopolissacarídeos/farmacologia , Simulação de Acoplamento Molecular , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
4.
Phytomedicine ; 104: 154277, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35752078

RESUMO

BACKGROUND: Excessive myocardial fibrosis is the pathological basis of heart failure following myocardial infarction (MI). Although calycosin improves cardiac function, its effect on cardiac fibrosis and cardiac function after MI in mice and its precise mechanism remain unclear. PURPOSE: Here, we firstly investigated the effects of calycosin on cardiac fibrosis and ventricular function in mice after MI and the role of transforming growth factor-beta receptor 1 (TGFBR1) signaling in the amelioration of cardiac fibrosis and ventricular function. METHODS: In vivo effects of calycosin on cardiac structure and function in mice with MI induced by left anterior descending coronary artery ligation were determined by hematoxylin and eosin staining, Masson trichrome staining, and echocardiography. The molecular mechanism of the interaction between TGFBR1 and calycosin was investigated using molecular docking, molecular dynamics (MD) simulation, surface plasmon resonance imaging (SPRi), immunohistochemistry, and western blotting (WB). Subsequently, cardiac-specific Tgfbr1 knockout mice were used to verify the effects of calycosin. The effect of calycosin on primary cardiac fibroblasts (CFs) proliferation and collagen deposition was detected using cell counting (CCK-8), EdU assay, and WB in vitro. CFs infected with an adenovirus that encodes TGFBR1 were used to verify the effects of calycosin. RESULTS: In vivo, calycosin attenuated myocardial fibrosis and cardiac dysfunction following MI in a dose-dependent pattern. Calycosin-TGFBR1 complex was found to have a binding energy of -9.04 kcal/mol based on molecular docking. In addition, calycosin bound steadily in the cavity of TGFBR1 during the MD simulation. Based on SPRi results, the solution equilibrium dissociation constant for calycosin and TGFBR1 was 5.11 × 10-5 M. Calycosin inhibited the expression of TGFBR1, Smad2/3, collagen I, and collagen III. The deletion of TGFBR1 partially counteracted these effects. In vitro, calycosin suppressed CFs proliferation and collagen deposition after TGF-ß1 stimulation by suppressing the TGFBR1 signaling pathway. The suppressive effects of calycosin were partially rescued by overexpression of TGFBR1. CONCLUSION: Calycosin attenuates myocardial fibrosis and cardiac dysfunction following MI in mice in vivo via suppressing the TGFBR1 signaling pathway. Calycosin suppresses CFs proliferation and collagen deposition induced by TGF-ß1 via inhibition of the TGFBR1 signaling pathway in vitro.


Assuntos
Infarto do Miocárdio , Animais , Colágeno/metabolismo , Fibrose , Isoflavonas , Camundongos , Simulação de Acoplamento Molecular , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo
5.
Diabetes Metab J ; 46(5): 677-688, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35124687

RESUMO

BACKGROUND: Neonatal porcine pancreatic cell clusters (NPCCs) have been proposed as an alternative source of ß cells for islet transplantation because of their low cost and growth potential after transplantation. However, the delayed glucose lowering effect due to the immaturity of NPCCs and immunologic rejection remain as a barrier to NPCC's clinical application. Here, we demonstrate accelerated differentiation and immune-tolerant NPCCs by in vitro chemical treatment and microencapsulation. METHODS: NPCCs isolated from 3-day-old piglets were cultured in F-10 media and then microencapsulated with alginate on day 5. Differentiation of NPCCs is facilitated by media supplemented with activin receptor-like kinase 5 inhibitor II, triiodothyronine and exendin-4 for 2 weeks. Marginal number of microencapsulated NPCCs to cure diabetes with and without differentiation were transplanted into diabetic mice and observed for 8 weeks. RESULTS: The proportion of insulin-positive cells and insulin mRNA levels of NPCCs were significantly increased in vitro in the differentiated group compared with the undifferentiated group. Blood glucose levels decreased eventually after transplantation of microencapsulated NPCCs in diabetic mice and normalized after 7 weeks in the differentiated group. In addition, the differentiated group showed nearly normal glucose tolerance at 8 weeks after transplantation. In contrast, neither blood glucose levels nor glucose tolerance were improved in the undifferentiated group. Retrieved graft in the differentiated group showed greater insulin response to high glucose compared with the undifferentiated group. CONCLUSION: in vitro differentiation of microencapsulated immature NPCCs increased the proportion of insulin-positive cells and improved transplant efficacy in diabetic mice without immune rejection.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Ilhotas Pancreáticas , Alginatos/metabolismo , Alginatos/farmacologia , Animais , Animais Recém-Nascidos , Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/cirurgia , Exenatida/farmacologia , Insulina/metabolismo , Camundongos , RNA Mensageiro/metabolismo , RNA Mensageiro/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Suínos , Transplante Heterólogo , Tri-Iodotironina/metabolismo , Tri-Iodotironina/farmacologia
6.
Asian Pac J Cancer Prev ; 22(8): 2471-2478, 2021 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-34452560

RESUMO

OBJECTIVE: Angiogenesis plays a dominant role in many pathophysiologic disorders, including cancer. Tranilast, which is an anti-fibrotic drug, is also suggested as an anti-angiogenesis agent. As Teucrium polium (TP) is known as an herbal medicine with antitumor properties, this study aimed to investigate the effects of TP and Tranilast on human umbilical vein endothelial cells (HUVECs), in vitro model of angiogenesis, as well as rat's aortic ring ex vivo model. METHODS: In this study, The HUVECs were treated with various doses of TP and Tranilast each one alone or in combination together. Cell survival test, aortic ring ex-vivo assay, and evaluating mRNA expressions of VEGFA and TGF-ß ligands and receptors were performed. RESULTS: The survival rate of HUVECs has significantly (p <0.05) reduced by TP and Tranilast. The combination of both TP and Tranilast significantly reduced cell viability as compared to the administration of TP or Tranilast alone. As well, the treatment of HUVECs with TP and/or Tranilast significantly (p <0.05) decreased TGF-ß1, TGF-ß 2, TGF-ßRI, and TGF-ßRII mRNA expression levels, but not the expression of TGF-ß3 and TGF-ßRIII in the TP-treated cells. Image analysis showed that TP and/or Tranilast inhibited vascular growth in the aortic ring assay. CONCLUSION: Our results strongly support the anti-angiogenic effects of the TP and Tranilast combination on both in vitro and ex vivo models of angiogenesis. However, further investigations in in vivo models and human studies are needed before human use.


Assuntos
Inibidores da Angiogênese/farmacologia , Fibrina/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Neovascularização Patológica/tratamento farmacológico , Extratos Vegetais/farmacologia , Teucrium/química , ortoaminobenzoatos/farmacologia , Inibidores da Angiogênese/química , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Sinergismo Farmacológico , Quimioterapia Combinada , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Masculino , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , ortoaminobenzoatos/química
7.
Molecules ; 26(6)2021 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-33808650

RESUMO

Pulmonary fibrosis is a severe and irreversible interstitial pulmonary disease with high mortality and few treatments. Magnesium lithospermate B (MLB) is a hydrosoluble component of Salvia miltiorrhiza and has been reported to have antifibrotic effects in other forms of tissue fibrosis. In this research, we studied the effects of MLB on pulmonary fibrosis and the underlying mechanisms. Our results indicated that MLB treatment (50 mg/kg) for seven days could attenuate bleomycin (BLM)-induced pulmonary fibrosis by reducing the alveolar structure disruption and collagen deposition in the C57 mouse model. MLB was also found to inhibit transforming growth factor-beta (TGF-ß)-stimulated myofibroblastic transdifferentiation of human lung fibroblast cell line (MRC-5) cells and collagen production by human type II alveolar epithelial cell line (A549) cells, mainly by decreasing the expression of TGF-ß receptor I (TGF-ßRI) and regulating the TGF-ß/Smad pathway. Further studies confirmed that the molecular mechanisms of MLB in BLM-induced pulmonary fibrosis mice were similar to those observed in vitro. In summary, our results demonstrated that MLB could alleviate experimental pulmonary fibrosis both in vivo and in vitro, suggesting that MLB has great potential for pulmonary fibrosis treatment.


Assuntos
Bleomicina/efeitos adversos , Medicamentos de Ervas Chinesas/farmacologia , Fibrose Pulmonar , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Animais , Bleomicina/farmacologia , Modelos Animais de Doenças , Masculino , Camundongos , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia
8.
J Ethnopharmacol ; 270: 113838, 2021 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-33460756

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Myocardial fibrosis after myocardial infarction (MI) leads to cardiac remodeling and loss of function. Taohong siwu decoction (THSWD), a well-known traditional Chinese medicinal prescription, has been clinically used to treat various cardiovascular and cerebrovascular diseases, but its potential functions in myocardial fibrosis after MI remain uncharacterized. AIM OF THE STUDY: The purpose of current study was to explore the potential mechanism action and anti-myocardial fibrosis effects of treatment with THSWD in vivo and in vitro. MATERIALS AND METHODS: Mouse underwent ligation of coronary artery to induce MI and divided equally into the sham group, model group and THSWD treatment groups. After 4 weeks, the effects of THSWD treatment on cardiac function were estimated by echocardiography. HE staining was used to detect the pathologic changes and Masson trichrome staining was used to estimate tissue fibrosis. To further explore the regulatory molecular mechanisms of THSWD, transcriptome analysis was performed. Furthermore, in vitro, we investigated the effect of THSWD on cell proliferation and collagen deposition in primary cardiac fibrosis cells and its possible mechanism of action. Overexpression of TGFBR1 was achieved by infection with an adenovirus vector encoding TGFBR1. RESULTS: Treatment with THSWD significantly decreased myocardial fibrosis and recovered cardiac function in the post-MI mouse. The transcriptomics data imply that the TGF-ß pathway might be a target in the anti-fibrosis effect of THSWD. THSWD inhibits TGF-ß1-induced proliferation of primary cardiac fibroblasts. THSWD decreased collagen expression and TGFBR1 and Smad2/3 phosphorylation. Moreover, the inhibitory effect of THSWD on CFs proliferation and collagen deposition, as well as TGFBR1 signaling pathway-associated proteins expression was partially abrogated by overexpression of TGFBR1. CONCLUSION: Collectively, the results implicate that THSWD attenuates myocardial fibrosis by inhibiting fibrosis proliferation and collagen deposition via inhibiting TGFBR1, and might be a potential therapeutic agent for treatment of myocardial fibrosis post-MI.


Assuntos
Colágeno/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Fibrose/tratamento farmacológico , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Colágeno/antagonistas & inibidores , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/química , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose/etiologia , Fibrose/metabolismo , Fibrose/patologia , Masculino , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/complicações , Infarto do Miocárdio/diagnóstico por imagem , Miocárdio/metabolismo , Miocárdio/patologia , Cultura Primária de Células , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/metabolismo , Transcriptoma/efeitos dos fármacos
9.
J Nutr Biochem ; 87: 108518, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33017609

RESUMO

The galloyl moiety is a specific structural feature which dictates, in part, the chemopreventive properties of diet-derived catechins. In ovarian cancer cells, galloylated catechins were recently demonstrated to target the transforming growth factor (TGF)-ß-mediated control of the epithelial-mesenchymal transition process. The specific impact of the galloyl moiety on such signaling, however, remains poorly understood. Here, we questioned whether the sole galloyl moiety interacted with TGF-ß-receptors to alter signal transduction and chemotactic migratory response in an ES-2 serous carcinoma-derived ovarian cancer cell model. In line with the LogP and LogS values of the tested molecules, we found that TGF-ß-induced Smad-3 phosphorylation and cell migration were optimally inhibited, provided that the lateral aliphatic chain of the galloyl moiety reached 8-10 carbons. Functional inhibition of the TGF-ß receptor (TGF-ßR1) kinase activity was supported by surface plasmon resonance assays showing direct physical interaction between TGF-ßR1 and the galloyl moiety. In silico molecular docking analysis predicted a model where galloylated catechins may bind TGF-ßR1 within its adenosine triphosphate binding cleft in a site analogous to that of Galunisertib, a selective adenosine triphosphate-mimetic competitive inhibitor of TGF-ßR1. In conclusion, our data suggest that the galloyl moiety of the diet-derived catechins provides specificity of action to galloylated catechins by positioning them within the kinase domain of the TGF-ßR1 in order to antagonize TGF-ß-mediated signaling that is required for ovarian cancer cell invasion and metastasis.


Assuntos
Catequina/farmacologia , Ácido Gálico/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , Chá/química , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/isolamento & purificação , Antineoplásicos Fitogênicos/farmacologia , Catequina/química , Catequina/isolamento & purificação , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Feminino , Ácido Gálico/análogos & derivados , Ácido Gálico/isolamento & purificação , Humanos , Neoplasias Ovarianas/metabolismo , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/isolamento & purificação , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo
10.
Bioorg Chem ; 105: 104368, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33091671

RESUMO

The discovery of the antiproliferative potential of tranilast prompted additional studies directed at understanding the mechanisms of tranilast action. Its inhibitory effect on cell proliferation depends principally on the capacity of tranilast to interfere with transforming growth factor beta (TGFßR1) signaling. This work summarizes design, synthesis and biological evaluation of sixteen novel tranilast analogs on different tumors such as PC-3, HepG-2 and MCF-7 cell lines. The in vitro cytotoxicity was evaluated using MTT assay showed that, twelve compounds out of sixteen showed higher cytotoxic activities (IC50's 1.1-6.29 µM), than that of the reference standard, 5-FU (IC50 7.53 µM). The promising cytotoxic hits (4b, 7a, b and 14c-e), proved to be selective to cancer cells when their cytotoxicity's are examined on human normal cell line (WI-38). Then they are investigated for their possible mode of action as TGFßR1 inhibitors; remarkable inhibition of TGFßR1 by these hits was observed at the range of IC50 0.087-3.276 µM. The cell cycle analysis of the most potent TGFßR1 inhibitor, 4b revealed cell cycle arrest at G2/M phase on prostate cancer cells. Additionally, it is clearly indicated apoptosis induction at Pre-G1 phase, this is substantiated by significant increase in the expression on the tumor suppressor gene, p53 and up regulation the level of apoptosis mediator, caspase-3. In addition, in silico study was performed for validating the physicochemical and ADME properties which revealed that, all compounds are orally bioavailable with no side effects complying with Lipinski rule. The proposed mode of action can be further explored on the light of molecular modeling simulation of the most potent compounds, 4b and 14e which were docked into the active sites of TGFßR1 to predict their affinities toward the receptor.


Assuntos
Antineoplásicos/farmacologia , Desenho de Fármacos , Simulação de Acoplamento Molecular , Inibidores de Proteínas Quinases/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , ortoaminobenzoatos/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Relação Estrutura-Atividade , Células Tumorais Cultivadas , ortoaminobenzoatos/síntese química , ortoaminobenzoatos/química
11.
FASEB J ; 34(7): 9207-9222, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32469097

RESUMO

MicroRNAs have been reported to play a role in adipogenesis and obesity. This study was performed to investigate the role of miR-196b-5p in adipogenesis and the mechanism involved. The data revealed that miR-196b-5p expression increased in primary or established marrow stromal progenitor cells after adipogenic treatment. Supplementing miR-196b-5p in the progenitor cells stimulated adipogenic differentiation and lipogenesis, along with the induction of adipogenic and lipogenic factors. Conversely, inhibition of endogenous miR-196b-5p blocked adipogenesis and lipogenesis. Tuberous sclerosis 1 (Tsc1) and transforming growth factor-ß receptor 1 (TGFBR1) were demonstrated to be the direct target genes of miR-196b-5p. Supplementing miR-196b-5p activity in progenitor cells reduced the protein level of TSC1 and activated mammalian target of rapamycin complex 1 (mTORC1) signaling. We further demonstrated that the perturbation of TSC1 in progenitor cells altered the trend of adipogenic differentiation and lipogenesis. Overexpression of Tsc1 or inactivation of mTORC1 signaling attenuated the stimulation of adipogenic differentiation and lipogenesis by miR-196b-5p. Overexpression of Tgfbr1 also partially blocked the adipogenic effect of miR-196b-5p. Further investigations demonstrated that zinc finger E-box-binding homeobox 1 (ZEB1) transcriptionally upregulated miR-196b-5p expression. The current study suggests that miR-196b-5p promotes adipogenic differentiation and lipogenesis in progenitor cells through targeting TSC1 and TGFBR1 and therefore regulating mTORC1 and TGF-ß signaling.


Assuntos
Adipócitos/citologia , Adipogenia , Diferenciação Celular , Lipogênese , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , MicroRNAs/genética , Fator de Crescimento Transformador beta/metabolismo , Adipócitos/metabolismo , Animais , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Camundongos Endogâmicos C57BL , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/genética , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa/metabolismo
12.
Mol Cell Biochem ; 470(1-2): 41-51, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32394311

RESUMO

Astragaloside IV (AS/IV) is one of the extracted components from the traditional Chinese medicine Astragalus which has been demonstrated to have potential capacity for anti-inflammation activity and for treating cardiovascular disease. Our purpose was to determine the function and underlying molecular mechanism of AS/IV in hypoxia/reoxygenation (H/R) injured in cardiomyocytes. Differentially expressed genes (DEGs) were screened using bioinformatic analysis, and the molecular targeting relationship was verified by the dual-luciferase report system. H/R injured cardiomyocytes were employed to explore the effect of AS/IV. QRT-PCR and Western blot analysis were applied to detect the expression of mRNA and proteins, respectively. Additionally, superoxide dismutase (SOD), lactic dehydrogenase (LDH) and MDA (malondialdehyde) levels were detected to determine the oxidative damage. Cell viability was assessed by CCK-8, and flow cytometry was used to evaluate cell apoptosis ratio. TGFBR1 and TLR2 were selected as DEGs. Additionally, AS/IV could enhance cell proliferation and upregulated miR-101a expression, which suppressed TGFBR1 and TLR2 expression in H/R injured cardiomyocytes. Moreover, the results of Western blot exhibited that the downstream genes (p-ERK and p-p38) in the MAPK signaling pathway were suppressed, which meant AS/IV could inhibit this pathway in H/R injured cardiomyocytes. Overall, this study demonstrated AS/IV could attenuate H/R injury in human cardiomyocytes via the miR-101a/TGFBR1/TLR2/MAPK signaling pathway axis, which means that it could serve as a possible alternate for H/R treatment.


Assuntos
Hipóxia Celular/efeitos dos fármacos , MicroRNAs/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Saponinas/farmacologia , Triterpenos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células , Sobrevivência Celular , Biologia Computacional , Perfilação da Expressão Gênica , Humanos , Inflamação , L-Lactato Desidrogenase/metabolismo , Sistema de Sinalização das MAP Quinases , Malondialdeído/metabolismo , Traumatismo por Reperfusão Miocárdica , Miócitos Cardíacos/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Receptor 2 Toll-Like/metabolismo
13.
Am J Chin Med ; 48(3): 535-558, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32345031

RESUMO

The Th17/Treg axis plays a crucial role in immune-mediated inflammatory diseases (IMID) and might represent an interesting drug target of treatment strategy for these diseases. Accumulating evidence suggests a role for traditional Chinese medicine (TCM) in the modulation of Th17/Treg axis, but a comprehensive overview which summarizes this field hitherto is lacked. This paper performs a systematic literature review of the regulatory effects of TCM on the imbalance of Th17/Treg axis and its potential mechanisms. In addition, the frequency analysis and network pharmacology for the collected TCM herbs from clinical trial data were performed. The studies reported the changes in the ratio of Th17 and/or Treg cells as well as their transcription factor and related cytokines were included. Frequency analysis of composition of the 39 assessed TCM prescriptions showed that Astragalus membranaceus var.mongholicus (5.20%), Glycyrrhiza uralensis (3.67%), Paeonia obovate (3.06%), Salvia digitaloides (3.06%), and Angelica sinensis (2.75%) were the top five herbal components, which were closely associated to the treatment of IMID. Network pharmacology showed that six target proteins (transforming growth factor (TGF)-beta receptor type-1, TGF-beta receptor type-2, retineic-acid-receptor-related orphan nuclear receptor gamma (ROR-gamma), TGFB2, IL-17 and IL-2, respectively) might be involved in the regulatory effects of TCM on Th17/Treg axis. Moreover, there were nine active ingredients (including Oxymatrine, Baicalin, Triptolide, Paeoniflorin, Sinomenine, Celastrol, Emodin, Diosgenin and Chlorogenic acid) originating from TCM reported to have an immunological regulation effect on the Th17/Treg axis. The highlight of this systematic review is to reveal the pharmacological basis of TCM treating IMID and is helpful for supporting future pharmacologic-driven studies. Further research elucidates the immune-modulating mechanisms on Th17/Treg axis by TCM might provide a broader insight for the treatment of IMID.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/imunologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Medicina Tradicional Chinesa , Fitoterapia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Angelica sinensis , Astrágalo , Medicamentos de Ervas Chinesas/química , Glycyrrhiza uralensis , Humanos , Doenças do Sistema Imunitário/metabolismo , Inflamação/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Paeonia , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Salvia
14.
PLoS One ; 14(6): e0218178, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31170244

RESUMO

The role of the microenvironment in driving connective tissue disease is being increasingly appreciated. Matricellular proteins of the CCN family are signaling modifiers that are secreted by cells into the extracellular matrix microenvironment where they have profound, context-dependent effects on organ development, homeostasis and disease. Indeed, CCN proteins are emergent targets for therapeutic intervention. Recent evidence suggests that, in vivo, CCN3 has effects opposing CCN2. Moreover, when CCN3 expression is high, CCN2 expression is low. That is, they appear to be regulated in a yin/yang fashion, leading to the hypothesis that the CCN2:CCN3 ratio is important to control tissue homeostasis. To begin to test the hypothesis that alterations in CCN2:CCN3 expression might be important in skin biology in vivo, we evaluated the relative ex vivo effects of the profibrotic protein TGFbeta1 on dermal fibroblasts on protein and RNA expression of CCN3 and CCN2, as well as the related protein CCN1. We also used signal transduction inhibitors to begin to identify the signal transduction pathways controlling the ability of fibroblasts to respond to TGFbeta1. As anticipated, CCN1 and CCN2 protein and mRNA were induced by TGFbeta1 in human dermal fibroblasts. This induction was blocked by TAK1, FAK, YAP1 and MEK inhibition. Conversely, TGFbeta1 suppressed CCN3 mRNA expression in a fashion insensitive to FAK, MEK, TAK1 or YAP1 inhibition. Unexpectedly, CCN3 protein was not detected in human dermal fibroblasts basally. These data suggest that, in dermal fibroblasts, the profibrotic protein TGFbeta1 has a divergent effect on CCN3 relative to CCN2 and CCN1, both at the mRNA and protein level. Given that the major source in skin in vivo of CCN proteins are fibroblasts, our data are consistent that alterations in CCN2/CCN1: CCN3 ratios in response to profibrotic agents such as TGFbeta1 may play a role in connective tissue pathologies including fibrosis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fator de Crescimento do Tecido Conjuntivo/biossíntese , Proteína Rica em Cisteína 61/biossíntese , Quinase 1 de Adesão Focal/metabolismo , Regulação da Expressão Gênica , MAP Quinase Quinase Quinases/metabolismo , Proteína Sobre-Expressa em Nefroblastoma/biossíntese , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Derme , Fibroblastos , Humanos , Proteínas de Sinalização YAP
15.
Biomed Pharmacother ; 106: 426-433, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29990830

RESUMO

Scabiosa comosa inflorescence is a traditional Mongolian medicine in the treatment of liver diseases. In the study, we investigated the anti-fibrotic efficacy of flavonoid-rich Scabiosa comosa inflorescence extract (TF-SC) in a rat model of CCl4-induced hepatic fibrosis and explored its underlying mechanism in vitro and in vivo. Rats (Wistar, Male, weight 200-250 g) were injected intraperitoneally with CCl4 (1:1v/v in peanut oil, 2 mL/kg body weight) to induce liver fibrosis, followed by treatment with TF-SC or vehicle. In addition, transforming growth factor-ß1 (TGF-ß1)-activated hepatic stellate cells (HSCs) were used for measuring Smad3 phosphorylation. We found decrease in liver function and liver fibrosis markers in serums. Also, TF-SC decreased hydroxyproline content and collagen deposition in liver tissues. TF-SC also decreased the expression of α-SMA, collagen I and fibronectin in CCl4-induced hepatic fibrosis rats. Mechanistically, TF-SC attenuated liver fibrosis by selectively inhibiting Smad3 phosphorylation. In TGF-ß1-stimulated HSCs, TF-SC blocked the interaction between Smad3 and TGF-ß type I receptor (TßRI), suppressed subsequent phosphorylation and nuclear translocation of Smad3, and down-regulated the transcription of fibrotic genes. In conclusion, the study demonstrated that TF-SC was an effective therapeutic agent for treatment of hepatic fibrosis, and provided a molecular basis through which TF-SC exerts its anti-fibrotic effects.


Assuntos
Tetracloreto de Carbono , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Flavonoides/farmacologia , Cirrose Hepática Experimental/prevenção & controle , Fígado/efeitos dos fármacos , Extratos Vegetais/farmacologia , Substâncias Protetoras/farmacologia , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Biomarcadores/sangue , Doença Hepática Induzida por Substâncias e Drogas/sangue , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citoproteção , Dipsacaceae/química , Relação Dose-Resposta a Droga , Flavonoides/isolamento & purificação , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Células Estreladas do Fígado/patologia , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática Experimental/sangue , Cirrose Hepática Experimental/induzido quimicamente , Cirrose Hepática Experimental/patologia , Masculino , Fosforilação , Fitoterapia , Extratos Vegetais/isolamento & purificação , Plantas Medicinais , Substâncias Protetoras/isolamento & purificação , Ratos Wistar , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 34(1): 69-73, 2018 Jan 08.
Artigo em Chinês | MEDLINE | ID: mdl-29926663

RESUMO

OBJECTIVE: To investigate the effects of centella asiatica (CA) granule on the expression of transform growth factor-ß1(TGF-ß1) and related down-stream signals in rats with early diabetic nephropathy(DN) and to clarify the molecular mechanisms of CA molecular mechanism of on preventing and curing early diabetic kidney disease DN by studying the effects of centella asiatica on TGF-ß1 expression and related down-stream signals. METHODS: Sixty male SD rats were divided into control group(n=10) and DN model group(n=50). The model rats were made a right nephrectomy. One week later, diabetic nephropathy was induced by intraperitoneal injection of streptocozin(30 mg/kg) for three consecutive days. High blood glucose level of Tail vein (fasting glucose ≥ 16.7 mmol/L) and high urinary protein level(total protein level in DN group was more than twice higher than the control group) were measured to confirm early DN in rats. In the sham operation group, the right renal capsule was damaged and the corresponding amount of saline was injected. The model rats were administrated by the means of intragastric administration. The DN model group were divided into DN group, DN+fosinopril group(1.6 mg/kg·d), DN+high CA group(16.8 mg/kg·d), DN+medium CA group(11.2 mg/kg·d) and DN+low CA group(5.6 mg/kg·d), and each group was intragastric administration one time every morning last for 16 weeks. The expressions of mRNA and protein of TGF-ß1, TßR1, TßR2, Smad2/3, Smad7 and the level of Smad2/3 phosphorylation were detected by using real time quantitative polymerase chain reaction and Western blot. RESULTS: The expressions of mRNA and protein of TGF-ß1, TßR1, TßR2, Smad2/3 and the level of Smad2/3 phosphorylation were significantly increased, the expressions of mRNA and protein of Smad7 were dramatically decreased. The fosinopril and high dosage CA could reverse the effects of DN. CONCLUSIONS: CA plays an important role in preventing and curing DN through regulating the TGF-ß1/Smad signaling pathways.


Assuntos
Centella/química , Nefropatias Diabéticas/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Diabetes Mellitus Experimental , Nefropatias Diabéticas/induzido quimicamente , Nefropatias Diabéticas/metabolismo , Rim/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/metabolismo , Transdução de Sinais , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Proteína Smad7/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA