Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Cell Immunol ; 395-396: 104781, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38159414

RESUMO

Pulmonary sarcoidosis is an immune-mediated disorder closely related to Th17/Treg cell imbalance. Dexamethasone has been shown to regulate inflammation and immune responses in sarcoidosis patients. However, the underlying mechanisms of dexamethasone regulating Th17/Treg balance in sarcoidosis remain elusive. Herein, we elucidated the function role of TGF-ß/Smad3 signaling in pulmonary sarcoidosis development and explored the underlying mechanism of dexamethasone in treating pulmonary sarcoidosis. We found that the TGF-ß/Smad3 pathway was inactivated in pulmonary sarcoidosis patients. Propionibacterium acnes (PA) induced mouse model was generated to investigate the function of TGF-ß/Smad3 signaling in vivo. Data indicated that IL17A inhibition with neutralizing antibody and activation of TGF-ß/Smad3 signaling with SRI-011381 alleviated granuloma formation in the sarcoidosis mouse model. Moreover, we revealed that the Th17/Treg cell ratio was increased with PA treatment in mouse bronchoalveolar lavage fluid (BALF) and peripheral blood. The concentration of cytokines produced by Th17 cells (IL-17A, IL-23) was up-regulated in the BALF of PA-treated mice, while those produced by Tregs (IL-10, TGF-ß1) presented significant reduction. The treatment of IL-17A neutralizing antibody or SRI-011381 was demonstrated to rescue the PA-induced changes in the concentration of IL-17A, IL-23, IL-10, and TGF-ß1. Additionally, we demonstrated that dexamethasone treatment activated the TGF-ß/Smad3 signaling in the lung tissues of pulmonary sarcoidosis mice. Dexamethasone was also revealed to promote the rebalancing of the Th17/Treg ratio and attenuated the granuloma formation in pulmonary sarcoidosis. In conclusion, dexamethasone activates the TGF-ß/Smad3 signaling and induces Th17/Treg rebalance, alleviating pulmonary sarcoidosis, which suggests the potential of dexamethasone in treating pulmonary sarcoidosis.


Assuntos
Dexametasona , Sarcoidose Pulmonar , Animais , Humanos , Camundongos , Anticorpos Neutralizantes/farmacologia , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Granuloma/prevenção & controle , Interleucina-10/metabolismo , Interleucina-17 , Interleucina-23/metabolismo , Sarcoidose Pulmonar/tratamento farmacológico , Linfócitos T Reguladores , Células Th17 , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
2.
Toxicol Appl Pharmacol ; : 117099, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39260469

RESUMO

BACKGROUND AND AIM: Hepatic fibrosis, one of the main reasons for death globally, is a serious complication of chronic liver disorders. However, the available therapies for liver fibrosis are limited, ineffective, and often associated with adverse events. Hence, seeking for a novel, effective therapy is warranted. Our objective was to investigate the potential efficacy of ferulic acid (FA), a phenolic phytochemical, at different doses in hindering the progress of concanavalin A (Con A)-induced hepatic fibrosis and explore the involved mechanisms. METHODS: Thirty-six mice were assorted into 6 groups (n = 6): Group I (control); group II received FA (20 mg/kg/day orally for 4 weeks); group III received Con A (6 mg/kg/week/i.v.) for 4 weeks; groups IV, V, and VI received Con A and were offered FA at 5, 10, and 20 mg/kg/day, respectively. RESULTS: The data showed the palliative effect of FA against Con A-induced fibrosis in a dose-dependent manner. This was obvious from the recovery of liver markers and hepatic architecture with the regression of fibrosis in FA-treated mice. FA abolished Con A-mediated oxidative insults and promoted the antioxidant enzyme activities, which run through the Nrf2/HO-1 signaling. Additionally, FA suppressed Con A-induced increase in NF-kB and IL-ß levels, and TNF-α immune-expression. The anti-fibrotic effect of FA was evident from the drop in TGF-ß, smad3 levels, α-SMA expression, and hydroxyproline content. CONCLUSION: FA attenuated Con A-induced liver fibrosis through stimulating Nrf2 signaling, suppressing NF-kB, and inhibiting the TGF-ß/smad3 signaling pathway. Thus FA can be considered as a promising therapy for combating liver fibrosis.

3.
Mol Ther ; 31(2): 569-584, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36307990

RESUMO

Myeloid-derived suppressor cells (MDSCs) are a group of immature myeloid cells that play an important role in diseases. MDSCs promote Th17 differentiation and aggravate systemic lupus erythematosus (SLE) progression by producing arginase-1 to metabolize arginine. However, the metabolic regulators remain unknown. Here, we report that MDSC derivative polyamines can promote Th17 differentiation via miR-542-5p in vitro. Th17 polarization was enhanced in response to polyamine treatment or upon miR-542-5p overexpression. The TGF-ß/SMAD3 pathway was shown to be involved in miR-542-5p-facilitated Th17 differentiation. Furthermore, miR-542-5p expression positively correlated with the levels of polyamine synthetases in peripheral blood mononuclear cells of patients with SLE as well as disease severity. In humanized SLE model mice, MDSC depletion decreased the levels of Th17 cells, accompanied by reduced expression of miR-542-5p and these polyamine synthetases. In addition, miR-542-5p expression positively correlated with the Th17 level and disease severity in both patients and humanized SLE mice. Together, our data reveal a novel molecular pathway by which MDSC-derived polyamine metabolism enhances Th17 differentiation and aggravates SLE.


Assuntos
Lúpus Eritematoso Sistêmico , MicroRNAs , Células Supressoras Mieloides , Animais , Camundongos , Células Supressoras Mieloides/metabolismo , Células Th17/metabolismo , Leucócitos Mononucleares/metabolismo , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Progressão da Doença , Ligases/metabolismo
4.
Pestic Biochem Physiol ; 200: 105831, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38582594

RESUMO

Paraquat (PQ) causes fatal poisoning that leads to systemic multiple organ fibrosis, and transforming growth factor (TGF)-ß1 plays a critical role in this process. In this study, we aimed to investigate the effects of AZ12601011 (a small molecular inhibitor of TGFßRI) on PQ-induced multiple organ fibrosis. We established a mouse model of PQ in vivo and used PQ-treated lung epithelial cell (A549) and renal tubular epithelial cells (TECs) in vitro. Haematoxylin-eosin and Masson staining revealed that AZ12601011 ameliorated pulmonary, hepatic, and renal fibrosis, consistent with the decrease in the levels of fibrotic indicators, alpha-smooth muscle actin (α-SMA) and collagen-1, in the lungs and kidneys of PQ-treated mice. In vitro data showed that AZ12601011 suppressed the induction of α-SMA and collagen-1 in PQ-treated A549 cells and TECs. In addition, AZ12601011 inhibited the release of inflammatory factors, interleukin (IL)-1ß, IL-6, and tumour necrosis factor-α. Mechanistically, TGF-ß and TGFßRI levels were significantly upregulated in the lungs and kidneys of PQ-treated mice. Cellular thermal shift assay and western blotting revealed that AZ12601011 directly bound with TGFßRI and blocked the activation of Smad3 downstream. In conclusion, our findings revealed that AZ12601011 attenuated PQ-induced multiple organ fibrosis by blocking the TGF-ß/Smad3 signalling pathway, suggesting its potential for PQ poisoning treatment.


Assuntos
Lesão Pulmonar Aguda , Paraquat , Fibrose Pulmonar , Camundongos , Animais , Paraquat/toxicidade , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Fator de Crescimento Transformador beta/toxicidade , Fator de Crescimento Transformador beta1/toxicidade , Fator de Crescimento Transformador beta1/metabolismo , Colágeno/toxicidade , Colágeno/metabolismo , Fatores de Crescimento Transformadores/toxicidade
5.
Pharmacol Res ; 176: 105962, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34756923

RESUMO

OBJECTIVE: Acute kidney injury (AKI) is a serious complication of sepsis. This study was performed to explore the mechanism that THBS1 mediated pyroptosis by regulating the TGF-ß signaling pathway in sepsis-induced AKI. METHODS: Gene expression microarray related to sepsis-induced AKI was obtained from the GEO database, and the mechanism in sepsis-induced AKI was predicted by bioinformatics analysis. qRT-PCR and ELISA were performed to detect expressions of THBS1, USF2, TNF-α, IL-1ß, and IL-18 in sepsis-induced AKI patients and healthy volunteers. The mouse model of sepsis-induced AKI was established, with serum creatinine, urea nitrogen, 24-h urine output measured, and renal tissue lesions observed by HE staining. The cell model of sepsis-induced AKI was cultured in vitro, with expressions of TNF-α, IL-1ß, and IL-18, pyroptosis, Caspase-1 and GSDMD-N, and activation of TGF-ß/Smad3 pathway detected. The upstream transcription factor USF2 was knocked down in cells to explore its effect on sepsis-induced AKI. RESULTS: THBS1 and USF2 were highly expressed in patients with sepsis-induced AKI. Silencing THBS1 protected mice against sepsis-induced AKI, and significantly decreased the expressions of NLRP3, Caspase-1, GSDMD-N, IL-1ß, and IL-18, increased cell viability, and decreased LDH activity, thus partially reversing the changes in cell morphology. Mechanistically, USF2 promoted oxidative stress responses by transcriptionally activating THBS1 to activate the TGF-ß/Smad3/NLRP3/Caspase-1 signaling pathway and stimulate pyroptosis, and finally exacerbated sepsis-induced AKI. CONCLUSION: USF2 knockdown downregulates THBS1 to inhibit the TGF-ß/Smad3 signaling pathway and reduce pyroptosis and further ameliorate sepsis-induced AKI.


Assuntos
Injúria Renal Aguda/etiologia , Citocinas/genética , Sepse/complicações , Trombospondina 1/genética , Fatores Estimuladores Upstream/genética , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Animais , Caspase 1/metabolismo , Linhagem Celular , Sobrevivência Celular , Citocinas/metabolismo , Regulação para Baixo , Feminino , Humanos , Rim/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose , Sepse/genética , Sepse/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo
6.
Mol Biol Rep ; 49(4): 2839-2845, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35067813

RESUMO

BACKGROUND: Hepatic fibrosis is one of the main reasons for mortality in the world. Hepatic stellate cells (HSCs) activate during chronic liver injury, express more Transforming growth factor beta (TGF-ß), Collagen1α (COLA1) and actin-alpha smooth muscle (αSMA) that lead to hepatic fibrosis. Quercetin is a flavonoid in vegetables and fruits that has shown hepatoprotective potential, but little is known about its effects on HSCs activation. In this study, we investigated the antifibrotic activity of Quercetin on fructose-activated human HSCs and its underlying mechanism in vitro. METHODS: First, the human HSCs were treated with fructose (25 mM) for 48 h and then with Quercetin for 24 h. Total RNAs were extracted, reversely transcribed into cDNA, Quantitative Real-time PCR and western blot were performed. RESULTS: The results showed that the levels of mRNA expression of TGF-ß, αSMA, Collagen1 genes, and phosphorylated smad3 protein were significantly reduced in fructose-activated HSCs after treatment with Quercetin compared to fructose-activated HSCs. CONCLUSION: Quercetin is effective in reducing the expression of fibrogenic genes in fructose-activated human HSCs through downregulation of the TGF-ß/smad3 signaling pathway. Therefore, Quercetin possesses significant antifibrotic properties in hepatic fibrosis.


Assuntos
Células Estreladas do Fígado , Quercetina , Frutose/metabolismo , Frutose/farmacologia , Células Estreladas do Fígado/metabolismo , Humanos , Cirrose Hepática/patologia , Quercetina/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
7.
J Cell Mol Med ; 24(22): 13093-13103, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32971570

RESUMO

Angiotensin-converting enzyme-2 (ACE2) and Mas receptor are the major components of the ACE2/Ang 1-7/Mas axis and have been shown to play a protective role in hypertension and hypertensive nephropathy individually. However, the effects of dual deficiency of ACE2 and Mas (ACE2/Mas) on Ang II-induced hypertensive nephropathy remain unexplored, which was investigated in this study in a mouse model of hypertension induced in either ACE2 knockout (KO) or Mas KO mice and in double ACE2/Mas KO mice by subcutaneously chronic infusion of Ang II. Compared with wild-type (WT) animals, mice lacking either ACE2 or Mas significantly increased blood pressure over 7-28 days following a chronic Ang II infusion (P < .001), which was further exacerbated in double ACE2/Mas KO mice (P < .001). Furthermore, compared to a single ACE2 or Mas KO mice, mice lacking ACE2/Mas developed more severe renal injury including higher levels of serum creatinine and a further reduction in creatinine clearance, and progressive renal inflammation and fibrosis. Mechanistically, worsen hypertensive nephropathy in double ACE2/Mas KO mice was associated with markedly enhanced AT1-ERK1/2-Smad3 and NF-κB signalling, thereby promoting renal fibrosis and renal inflammation in the hypertensive kidney. In conclusion, ACE2 and Mas play an additive protective role in Ang II-induced hypertension and hypertensive nephropathy. Thus, restoring the ACE2/Ang1-7/Mas axis may represent a novel therapy for hypertension and hypertensive nephropathy.


Assuntos
Angiotensina II/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Hipertensão Renal/metabolismo , Nefrite/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Pressão Sanguínea , Fibrose , Deleção de Genes , Hipertensão Renal/genética , Inflamação , Rim/metabolismo , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nefrite/genética , Proteinúria/genética , Proto-Oncogene Mas , Transdução de Sinais
8.
Toxicol Mech Methods ; 30(8): 620-633, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32718261

RESUMO

Protection against liver injury and its consequences is considered an essential issue to minimize the number of annual deaths caused by liver diseases. The present study was designed to evaluate the potential role of pomegranate extract (PE) and/or curcumin in the regression of thioacetamide (TAA)-induced liver fibrosis, focusing on their modulatory effects on Nrf2/HO-1, NF-κB, and TGF-ß/Smad3 signaling pathways. Liver fibrosis was induced in male Wistar rats by intraperitoneal injection of TAA (100 mg/kg) three times a week, for 8 weeks. To assess the protective effects of PE and/or curcumin against TAA-induced liver fibrosis, rats were treated on a daily basis with oral doses of PE (200 mg/kg) and/or curcumin (200 mg/kg) for 8 weeks. The results indicated that PE and/or curcumin attenuated TAA-induced liver fibrogenesis, as evidenced by a significant improvement in the liver function tests (AST, ALT, ALP, and albumin), oxidative stress biomarkers (MDA, SOD, and GSH), and inflammatory biomarkers (NF-ĸB, TNF-α, IL-1ß, iNOS, TGF-ß, and MPO), compared to TAA group. Moreover, treatment with PE and/or curcumin exerted a significant upregulation of Nrf2/HO-1 gene expressions along with significant downregulation of NF-ĸB, TGF-ß, and phospho-Smad3 protein expressions, as well as α-SMA and collagen-1 gene expressions. The histopathological examination has corroborated these findings. In conclusion, hepatoprotective activities of PE and/or curcumin could be linked to their abilities to modulate Nrf2/HO-1, NF-κB, and TGF-ß/Smad3 signaling pathways. It is worth noting that the combination of PE and curcumin exerted superior hepatoprotective effects against TAA-induced liver fibrosis, as compared to monotherapy.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Curcumina/farmacologia , Cirrose Hepática/prevenção & controle , Fígado/efeitos dos fármacos , NF-kappa B/metabolismo , Extratos Vegetais/farmacologia , Punica granatum , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Quimioterapia Combinada , Frutas , Humanos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Extratos Vegetais/isolamento & purificação , Punica granatum/química , Ratos Wistar , Transdução de Sinais , Tioacetamida
9.
J Cell Mol Med ; 23(8): 5576-5587, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31211499

RESUMO

Obstructive nephropathy is the end result of a variety of diseases that block drainage from the kidney(s). Transforming growth factor-ß1 (TGF-ß1)/Smad3-driven renal fibrosis is the common pathogenesis of obstructive nephropathy. In this study, we identified petchiether A (petA), a novel small-molecule meroterpenoid from Ganoderma, as a potential inhibitor of TGF-ß1-induced Smad3 phosphorylation. The obstructive nephropathy was induced by unilateral ureteral obstruction (UUO) in mice. Mice received an intraperitoneal injection of petA/vehicle before and after UUO or sham operation. An in vivo study revealed that petA protected against renal inflammation and fibrosis by reducing the infiltration of macrophages, inhibiting the expression of proinflammatory cytokines (interleukin-1ß and tumour necrosis factor-α) and reducing extracellular matrix deposition (α-smooth muscle actin, collagen I and fibronectin) in the obstructed kidney of UUO mice; these changes were associated with suppression of Smad3 and NF-κB p65 phosphorylation. Petchiether A inhibited Smad3 phosphorylation in vitro and down-regulated the expression of the fibrotic marker collagen I in TGF-ß1-treated renal epithelial cells. Further, we found that petA dose-dependently suppressed Smad3-responsive promoter activity, indicating that petA inhibits gene expression downstream of the TGF-ß/Smad3 signalling pathway. In conclusion, our findings suggest that petA protects against renal inflammation and fibrosis by selectively inhibiting TGF-ß/Smad3 signalling.


Assuntos
Nefropatias/tratamento farmacológico , Nefropatias/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo , Terpenos/uso terapêutico , Fator de Crescimento Transformador beta/metabolismo , Actinas/metabolismo , Animais , Linhagem Celular , Colágeno/metabolismo , Fibronectinas/metabolismo , Fibrose , Humanos , Inflamação/patologia , Rim/lesões , Rim/patologia , Nefropatias/patologia , Masculino , Camundongos Endogâmicos C57BL , Fosforilação , Terpenos/química , Terpenos/farmacologia , Terpenos/toxicidade , Obstrução Ureteral/tratamento farmacológico , Obstrução Ureteral/patologia
10.
Mol Ther ; 26(1): 148-161, 2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29102563

RESUMO

Transforming growth factor ß (TGF-ß)/Smad3 signaling plays a role in tissue fibrosis. We report here that Erbb4-IR is a novel long non-coding RNA (lncRNA) responsible for TGF-ß/Smad3-mediated renal fibrosis and is a specific therapeutic target for chronic kidney disease. Erbb4-IR was induced by TGF-ß1 via a Smad3-dependent mechanism and was highly upregulated in the fibrotic kidney of mouse unilateral ureteral obstructive nephropathy (UUO). Silencing Erbb4-IR blocked TGF-ß1-induced collagen I and alpha-smooth muscle actin (α-SMA) expressions in vitro and effectively attenuated renal fibrosis in the UUO kidney by blocking TGF-ß/Smad3 signaling. Mechanistic studies revealed that Smad7, a downstream negative regulator of TGF-ß/Smad signaling, is a target gene of Erbb4-IR because a binding site of Erbb4-IR was found on the 3' UTR of Smad7 gene. Mutation of this binding site prevented the suppressive effect of Erbb4-IR on the Smad7 reporter activity; in contrast, overexpression of Erbb4-IR largely inhibited Smad7 but increased collagen I and α-SMA transcriptions. Thus, kidney-specific silencing of Erbb4-IR upregulated renal Smad7 and thus blocked TGF-ß/Smad3-mediated renal fibrosis in vivo and in vitro. In conclusion, the present study identified that Erbb4-IR is a novel lncRNA responsible for TGF-ß/Smad3-mediated renal fibrosis by downregulating Smad7. Targeting Erbb4-IR may represent a precise therapeutic strategy for progressive renal fibrosis.


Assuntos
Nefropatias/genética , Nefropatias/metabolismo , RNA Longo não Codificante/genética , Receptor ErbB-4/genética , Proteína Smad3/genética , Fator de Crescimento Transformador beta/metabolismo , Animais , Biópsia , Linhagem Celular , Fibrose , Técnicas de Silenciamento de Genes , Inativação Gênica , Nefropatias/patologia , Camundongos , Transcrição Gênica
11.
Immunopharmacol Immunotoxicol ; 41(3): 438-445, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31119954

RESUMO

Objective: Paeonol is a natural phenolic component isolated from the root bark of peony with multiple pharmacological activities. We investigated the anti-fibrotic effect and underlying mechanism of paeonol. Methods: Twenty-four male C57BL/6J mice were divided into 4 groups (n = 6 in each group), injected with CCl4 to induce liver fibrosis and administrated with paeonol according to the regimen. The serum activity of ALT and AST, and H&E staining were to assess liver injury. Sirius and Masson staining, and hydroxyproline content were to evaluate the degree of liver fibrosis. TNF-α, IL-6, TGF-ß, MDA, GSH-PX, SOD, and CAT were detected to reflect inflammation and oxidative stress. RT-qPCR and Western blot analysis to assess the activation of HSCs and TGF-ß/Smad3 signaling. Results: Paeonol ameliorated liver injury and liver fibrosis, reflected by the decrease of ALT, AST, less lesion in H&E staining, mitigated fibrosis in Sirius and Masson staining, lessened content of hydroxyproline. Paeonol attenuated the level of IL-6 and TNF-α, and elevated the activity of GSH-PX, SOD, and CAT with reducing the level of MDA. The expression of col 1a, α-SMA, vimentin, and desmin were down-regulated and TGF-ß/Smad3 signaling pathway was inhibited. Conclusion: These data demonstrated that paeonol could alleviate CCl4-induced liver fibrosis through suppression of hepatic stellate cells activation via inhibiting the TGF-ß/Smad3 signaling.


Assuntos
Acetofenonas/farmacologia , Intoxicação por Tetracloreto de Carbono/tratamento farmacológico , Células Estreladas do Fígado/imunologia , Cirrose Hepática/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Intoxicação por Tetracloreto de Carbono/imunologia , Intoxicação por Tetracloreto de Carbono/patologia , Células Estreladas do Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Masculino , Camundongos , Transdução de Sinais/imunologia
12.
J Physiol ; 596(16): 3493-3503, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29781524

RESUMO

Transforming growth factor-ß (TGF-ß) is the key player in tissue fibrosis. However, antifibrotic therapy targeting this multifunctional protein may interfere with other physiological processes to cause side effects. Thus, precise therapeutic targets need to be identified by further understanding the underlying mechanisms of TGF-ß1 signalling during fibrogenesis. Equilibrium of Smad signalling is crucial for TGF-ß-mediated renal fibrosis, where Smad3 is pathogenic but Smad2 and Smad7 are protective. The activation of TGF-ß1/Smad signalling triggers extracellular matrix deposition, and local myofibroblast generation and activation. Mechanistic studies have shown that TGF-ß/Smad3 transits the microRNA profile from antifibrotic to profibrotic and therefore promotes renal fibrosis via regulating non-coding RNAs at transcriptional levels. More importantly, disease-specific Smad3-dependent long non-coding RNAs have been recently uncovered from mouse kidney disease models and may represent novel precision therapeutic targets for chronic kidney disease. In this review, mechanisms of TGF-ß-driven renal fibrosis via non-coding RNAs and their translational capacities will be discussed in detail.


Assuntos
Fibrose/patologia , Regulação da Expressão Gênica , Nefropatias/patologia , RNA não Traduzido/genética , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Fibrose/genética , Fibrose/metabolismo , Humanos , Nefropatias/genética , Nefropatias/metabolismo
13.
Clin Sci (Lond) ; 132(13): 1403-1415, 2018 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-29875262

RESUMO

It is well established that Smad3 is a key downstream effector of transforming growth factor-ß (TGF-ß) signaling in tissue fibrogenesis. We reported here that targetting Smad3 specifically with a Smad3 inhibitor SIS3 is able to prevent or halt the progression of renal fibrosis in a mouse model of unilateral ureteral obstructive nephropathy (UUO). We found that preventive treatment with SIS3 at the time of disease induction largely suppressed progressive renal fibrosis by inhibiting α-smooth muscle actin (α-SMA) + myofibroblast accumulation and extracellular matrix (collagen I (Col.I) and fibronectin (FN)) production. Importantly, we also found that treatment with SIS3 on established mouse model of UUO from day 4 after UUO nephropathy halted the progression of renal fibrosis. Mechanistically, the preventive and therapeutic effects of SIS3 on renal fibrosis were associated with the inactivation of Smad3 signaling and inhibition of TGF-ß1 expression in the UUO kidney. In conclusion, results from the present study suggest that targetting Smad3 may be a specific and effective therapy for renal fibrosis.


Assuntos
Isoquinolinas/farmacologia , Rim/patologia , Piridinas/farmacologia , Pirróis/farmacologia , Proteína Smad3/antagonistas & inibidores , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Fibrose , Isoquinolinas/administração & dosagem , Isoquinolinas/uso terapêutico , Rim/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Piridinas/administração & dosagem , Piridinas/uso terapêutico , Pirróis/administração & dosagem , Pirróis/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Obstrução Ureteral/complicações , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
14.
Ren Fail ; 40(1): 447-454, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30101622

RESUMO

3,3'-Diindolylmethane (DIM), a natural acid condensation extracted from cruciferous plants, exhibits anti-fibrotic effects in hepatic and cardiac fibrosis models. The effects of DIM on renal fibrosis, however, are unclear. This study aimed to explore the protective effects of DIM on renal fibrosis. Unilateral ureteral obstruction (UUO) and transforming growth factor (TGF)-ß1-stimulated normal rat kidney (NRK)-49F fibroblast cell mouse models were established. The models were then treated with DIM for the assessment of its anti-fibrotic effects and mechanisms. Results of HE and Masson staining showed that DIM reduced kidney injury and production of interstitial collagens fibrosis. CTS also inhibited expression of fibronectin, collagen-1 but retain E-cadherin in the UUO model. Furthermore, DIM suppressed local fibroblast activation, as evidenced by the suppressed expression of the myofibroblast markers α-SMA and vimentin in vivo and in vitro. In addition, DIM significantly inhibited the TGF-ß1-induced proliferation of NRK49F cells in a time- and dose-dependent manner. DIM decreased Smad2/3 phosphorylation but increased Smad7 expression. Results suggested that DIM inhibits TGF-ß/Smad2/3 signaling to attenuate renal interstitial fibrosis via inhibiting local fibroblast activation. This mechanism is likely related to Smad7 induction.


Assuntos
Indóis/farmacologia , Nefropatias/etiologia , Rim/patologia , Miofibroblastos/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo , Animais , Caderinas/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Fibronectinas/metabolismo , Fibrose , Nefropatias/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/efeitos dos fármacos , Fator de Crescimento Transformador beta1/efeitos dos fármacos
15.
Ren Fail ; 38(6): 945-51, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27050799

RESUMO

This study investigated whether oxymatrine (OMT) treatment can ameliorate renal interstitial fibrosis in unilateral ureteral obstruction (UUO) mice model. Moreover, the potential mechanisms of such treatment were analyzed. Twenty-four C57/BL6 mice were randomly divided into three groups, namely sham group, vehicle plus unilateral ureteral obstruction (UUO)-treated group, and 100 mg/kg/d OMT plus UUO-treated group. All mice were euthanized seven days after surgery, and their kidneys were harvested. Renal injury, fibrosis, expression of proinflammatory cytokines, and the transforming growth factor-ß1/Smads (TGF-ß/Smads) and nuclear factor-kappa B (NF-κB)-signaling pathways were assessed. The results showed OMT significantly prevented kidney injury and fibrosis, as evidenced by decreased expression of collagen-1 and fibronectin. Furthermore, OMT administration inhibited the release of inflammatory factors including tumor necrosis factor-α, (TNF-α) interleukin-1ß (IL-1ß), and interleukin-6 (IL-6), as well as phosphorylated NF-κB p65. In addition, OMT blocked the activation of myofibroblasts by inhibiting the TGF-ß/Smad3-signaling pathway. The findings indicate that OMT-attenuated renal fibrosis and inflammation, and this renoprotective effect may be ascribed to the inactivation of the TGF-ß/Smad3 and NF-κB p65 pathways.


Assuntos
Alcaloides/farmacologia , Citocinas/metabolismo , Rim/patologia , Quinolizinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Injúria Renal Aguda/prevenção & controle , Animais , Modelos Animais de Doenças , Regulação para Baixo , Fibrose/prevenção & controle , Inflamação/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Fator de Transcrição RelA/metabolismo , Obstrução Ureteral/patologia
16.
Pathol Res Pract ; 260: 155384, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38850874

RESUMO

OBJECTIVE: To investigate the association between Helicobacter pylori infection and GDF6 expression in gastric cancer patients, and to determine its influence on prognosis and resistance to capecitabine. METHODS: Tumor and adjacent non-tumor tissues were collected from 148 gastric cancer patients who underwent surgery in our department from October 2019 to June 2022. Of these patients, 78 tested positive for Helicobacter pylori and 70 tested negative. Hematoxylin-eosin (HE) and immunofluorescence staining were utilized to quantify GDF6 expression in cancerous and adjacent tissues. Patient prognosis was monitored via follow-up. Western blotting analyzed GDF6 expression in common gastric cancer cell lines. HGC27 cells exhibiting high GDF6 expression and BGC823 cells with low expression were used to create GDF6-silenced and overexpressed cell lines. The impact of GDF6 on the proliferation, migration, invasion, and cloning abilities of gastric cancer cells was evaluated using the CCK-8 assay, scratch test, Transwell assay, and plate colony formation assay. Fluorescent quantitative PCR and Western blotting assessed the effects of GDF6 levels on epithelial-mesenchymal transition (EMT) and tumor cell stemness. RESULTS: GDF6 expression in gastric cancer tissues was significantly correlated with cancer grading and staging (P<0.05). Helicobacter pylori-positive tissues exhibited significantly higher GDF6 expression levels than negative samples (P<0.05). Kaplan-Meier survival analysis indicated that high GDF6 expression was associated with poor survival prognosis. Overexpressed GDF6 enhanced the proliferation, migration, and invasion abilities of gastric cancer cells, while silencing GDF6 yielded opposite results. Increased GDF6 expression upregulated TGF-ß expression and the phosphorylation levels of SMAD3, leading to an elevation in mesenchymal cell markers N-cadherin, vimentin, and a reduction in epithelial cell markers cytokeratins, E-cadherin. Moreover, high GDF6 levels contributed to increased resistance to capecitabine and enhanced the expression of tumor stem cell markers Nanog, Sox-2, Oct-4, CD44, amplifying tumor cell stemness. CONCLUSION: Helicobacter pylori infection is associated with increased GDF6 expression in gastric cancer tissue, correlating with poor survival prognosis. Elevated GDF6 expression promotes the proliferation, migration, and invasion abilities of gastric cancer cells, facilitates EMT via the TGF-ß/SMAD3 pathway, and intensifies cell stemness and capecitabine resistance. Consequently, GDF6 presents itself as a potential new target for gastric cancer treatment. DATA AVAILABILITY STATEMENT: The data that support the findings of this study are available from the corresponding author upon reasonable request.


Assuntos
Transição Epitelial-Mesenquimal , Infecções por Helicobacter , Helicobacter pylori , Transdução de Sinais , Proteína Smad3 , Neoplasias Gástricas , Fator de Crescimento Transformador beta , Neoplasias Gástricas/patologia , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/metabolismo , Humanos , Feminino , Masculino , Pessoa de Meia-Idade , Infecções por Helicobacter/patologia , Infecções por Helicobacter/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína Smad3/metabolismo , Idoso , Regulação para Cima , Proliferação de Células , Prognóstico , Linhagem Celular Tumoral , Capecitabina/farmacologia , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Resistencia a Medicamentos Antineoplásicos , Adulto
17.
Iran J Basic Med Sci ; 27(9): 1096-1104, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39055878

RESUMO

Objectives: Liver diseases, including non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), pose significant global public health challenges. This study investigates the therapeutic effects of quercetin (QC), Capparis spinosa (CS), a QC and CS combination, and Saroglitazar (SARO) on NASH in a Wistar rat model. Materials and Methods: NASH was induced by a 42-day high-fat diet regimen in male Wistar rats. Post-induction, rats were divided into five groups receiving SARO, QC, CS, and CS+QC combination. We monitored changes in liver and body weights and evaluated the expression of genes associated with fatty acid biosynthesis (e.g., ACC and FAS), ß-oxidation (e.g., CPT1, PPAR α), inflammation (e.g., TNF-α and IL-6), and fibrosis (e.g., TGF-ß and COL1A), as well as protein expression levels of p-Smad2/3 and p-Smad3. Results: Treatment with QC+CS significantly decreased liver weight, body mass gain, and liver triglyceride (TG) compared to other treatments. The QC and CS combined therapy also resulted in a greater normalization of hepatic enzymatic activities, including decreases in ALT and AST levels, coupled with improvements in lipid profile indicated by decreased LDL-C and increased HDL-C concentrations, as compared to SARO and QC alone. Furthermore, this combined treatment significantly down-regulated the expression of TGF-ß, TNF-α, IL-6 genes, and Smad2/3 and Smad3 protein levels. Conclusion: Our study demonstrates that an interactive effect between QC and CS can effectively reduce liver fibrosis and steatosis by inhibiting the TGF-ß/Smad3 signaling pathway in a diet-induced model of nonalcoholic steatohepatitis and fibrosis in rats.

18.
Front Cardiovasc Med ; 11: 1323329, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38798919

RESUMO

Cardiac fibrosis is an important pathological manifestation of various cardiac diseases such as hypertension, coronary heart disease, and cardiomyopathy, and it is also a key link in heart failure. Previous studies have confirmed that exercise can enhance cardiac function and improve cardiac fibrosis, but the molecular target is still unclear. In this review, we introduce the important role of miR-126 in cardiac protection, and find that it can regulate TGF-ß/Smad3 signaling pathway, inhibit cardiac fibroblasts transdifferentiation, and reduce the production of collagen fibers. Recent studies have shown that exosomes secreted by cells can play a specific role through intercellular communication through the microRNAs carried by exosomes. Cardiac endothelial progenitor cell-derived exosomes (EPC-Exos) carry miR-126, and exercise training can not only enhance the release of exosomes, but also up-regulate the expression of miR-126. Therefore, through derivation and analysis, it is believed that exercise can inhibit TGF-ß/Smad3 signaling pathway by up-regulating the expression of miR-126 in EPC-Exos, thereby weakening the transdifferentiation of cardiac fibroblasts into myofibroblasts. This review summarizes the specific pathways of exercise to improve cardiac fibrosis by regulating exosomes, which provides new ideas for exercise to promote cardiovascular health.

19.
Adv Sci (Weinh) ; 10(32): e2304360, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37749872

RESUMO

Podocyte injury plays a critical role in the progression of focal segmental glomerulosclerosis (FSGS). Here, it is reported that B-cell translocation gene 2 (Btg2) promotes Adriamycin (ADR)-induced FSGS via Smad3-dependent podocyte-mesenchymal transition. It is found that in FSGS patients and animal models, Btg2 is markedly upregulated by podocytes and correlated with progressive renal injury. Podocyte-specific deletion of Btg2 protected against the onset of proteinuria and glomerulosclerosis in ADR-treated mice along with inhibition of EMT markers such as α-SMA and vimentin while restoring epithelial marker E-cadherin. In cultured MPC5 podocytes, overexpression of Btg2 largely promoted ADR and TGF-ß1-induced EMT and fibrosis, which is further enhanced by overexpressing Btg2 but blocked by disrupting Btg2. Mechanistically, Btg2 is rapidly induced by TGF-ß1 and then bound Smad3 but not Smad2 to promote Smad3 signaling and podocyte EMT, which is again exacerbated by overexpressing Btg2 but blocked by deleting Btg2 in MPC5 podocytes. Interestingly, blockade of Smad3 signaling with a Smad3 inhibitor SIS3 is also capable of inhibiting Btg2 expression and Btg2-mediated podocyte EMT, revealing a TGF-ß/Smad3-Btg2 circuit mechanism in Btg2-mediated podocyte injury in FSGS. In conclusion, Btg2 is pathogenic in FSGS and promotes podocyte injury via a Smad3-dependent EMT pathway.


Assuntos
Glomerulosclerose Segmentar e Focal , Podócitos , Animais , Humanos , Camundongos , Doxorrubicina/farmacologia , Glomerulosclerose Segmentar e Focal/induzido quimicamente , Glomerulosclerose Segmentar e Focal/metabolismo , Glomerulosclerose Segmentar e Focal/patologia , Rim/metabolismo , Podócitos/metabolismo , Podócitos/patologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
20.
Cell Signal ; 105: 110611, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36708753

RESUMO

TGF-ß signaling mediates its biological effects by engaging canonical Smad proteins and crosstalking extensively with other signaling networks, including the NF-kB pathway. The paracaspase MALT1 is an intracellular signaling molecule essential for NF-kB activation downstream of several key cell surface receptors. Despite intensive research on TGF-ß and NF-kB interactions, the significance of MALT1 in this context remains undecoded. Here we provide experimental evidence supporting that MALT1 functions to converge these pathways. Using A549 and Huh7 cancer cell line models, we report that TGF-ß stimulation enhances MALT1 protein and transcript levels in a time- and dose-dependent manner. Systematic and selective perturbation of TGF-ß signaling components identifies MALT1 as a downstream target of Smad3. Rescue experiments in SMAD3 knockout cells confirm that C-terminal phosphorylation of Smad3 is central to MALT1 induction. Corroborating these data, we document that the expression of SMAD3 and MALT1 genes are positively correlated in TCGA cohorts, and we trace the molecular basis of MALT1 elevation to promoter activation. Functional studies in parental as well as NF-kB p65 signaling reporter engineered cells conclusively reveal that MALT1 is paramount for TGF-ß-stimulated nuclear translocation and transcriptional activation of NF-kB p65. Furthermore, we find that BCL10 is also implicated in TGF-ß activation of NF-kB target genes, potentially coupling the TGF-ß-MALT1-NF-kB signaling axis to the CARMA-BCL10-MALT1 (CBM) signalosome. The novel findings of this study indicate that MALT1 is a downstream target of the canonical TGF-ß/Smad3 pathway and plays a critical role in modulating TGF-ß and NF-kB crosstalk in cancer.


Assuntos
NF-kappa B , Neoplasias , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa , NF-kappa B/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA