Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Nephrol ; 52(8): 653-665, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34496361

RESUMO

INTRODUCTION: Transforming growth factor-ß1 (TGF-ß1) is a multifunctional cytokine, with diverse roles in fibrosis and inflammation, which acts through Smad signaling in renal pathology. We intended to investigate the expression of TGF-ß/Smad signaling in glomerulonephritis (GN) and to assess its role as risk factor for progression to chronic kidney disease (CKD). METHODS: We evaluated the immunohistochemical expression of TGF-ß1, phosphorylated Smad3 (pSmad3), and Smad7 semiquantitatively and quantitatively using computerized image analysis program in different compartments of 50 renal biopsies with GN, and the results were statistically analyzed with clinicopathological parameters. We also examined the associations among their expressions, the impact of their co-expression, and their role in progression to CKD. RESULTS: TGF-ß1 expression correlated positively with segmental glomerulosclerosis (p= 0.025) and creatinine level at diagnosis (p = 0.002), while pSmad3 expression with interstitial inflammation (p = 0.024). In glomerulus, concomitant expressions of high Smad7 and medium pSmad3 were observed to be correlated with renal inflammation, such as cellular crescent (p = 0.011), intense interstitial inflammation (p = 0.029), and lower serum complement (C) 3 (p = 0.028) and C4 (p = 0.029). We also reported a significant association between pSmad3 expression in glomerular endothelial cells of proliferative GN (p = 0.045) and in podocytes of nonproliferative GN (p = 0.005). Finally, on multivariate Cox-regression analysis, TGF-ß1 expression (hazard ratio = 6.078; 95% confidence interval: 1.168-31.627; p = 0.032) was emerged as independent predictor for CKD. DISCUSSION/CONCLUSION: TGF-ß1/Smad signaling is upregulated with specific characteristics in different forms of GN. TGF-ß1 expression is indicated as independent risk factor for progression to CKD, while specific co-expression pattern of pSmad3 and Smad7 in glomerulus is correlated with renal inflammation.


Assuntos
Insuficiência Renal Crônica/etiologia , Proteína Smad3/fisiologia , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Adulto , Idoso , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Transdução de Sinais
2.
J Biol Chem ; 295(15): 4858-4869, 2020 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-32122971

RESUMO

The interplay between the transforming growth factor ß (TGF-ß) signaling proteins, SMAD family member 2 (SMAD2) and 3 (SMAD3), and the TGF-ß-inhibiting SMAD, SMAD7, seems to play a vital role in proper pancreatic endocrine development and also in normal ß-cell function in adult pancreatic islets. Here, we generated conditional SMAD7 knockout mice by crossing insulin1Cre mice with SMAD7fx/fx mice. We also created a ß cell-specific SMAD7-overexpressing mouse line by crossing insulin1Dre mice with HPRT-SMAD7/RosaGFP mice. We analyzed ß-cell function in adult islets when SMAD7 was either absent or overexpressed in ß cells. Loss of SMAD7 in ß cells inhibited proliferation, and SMAD7 overexpression enhanced cell proliferation. However, alterations in basic glucose homeostasis were not detectable following either SMAD7 deletion or overexpression in ß cells. Our results show that both the absence and overexpression of SMAD7 affect TGF-ß signaling and modulates ß-cell proliferation but does not appear to alter ß-cell function. Reversible SMAD7 overexpression may represent an attractive therapeutic option to enhance ß-cell proliferation without negative effects on ß-cell function.


Assuntos
Proliferação de Células , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/fisiologia , Insulina/fisiologia , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Glucose/farmacologia , Masculino , Camundongos , Camundongos Knockout , Transdução de Sinais , Edulcorantes/farmacologia , Fator de Crescimento Transformador beta/genética
3.
Anticancer Drugs ; 31(3): 272-281, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31815762

RESUMO

Drug resistance is a major problem in the treatment of advanced cervical cancer. The oncogenic microRNA-21 (miR-21) is involved in drug resistance in various cancers. However, the regulatory role of miR-21 and its target, Smad7 in drug resistance of cervical cancer remains to be elucidated. We compared miR-21 and Smad7 levels in human samples from chemoradiotherapy-resistance cervical cancer (resistant group) and chemoradiotherapy-sensitive cervical cancer (sensitive group) patients. Then, the miR-21 level was manipulated in HeLa and SiHa cervical cancer cells and the Smad7 level was determined by PCR and western blot. We also manipulated miR-21, Smad7 or both in cells, and measured cell viability using cell counting kit-8 method and epithelial-mesenchymal transition (EMT) biomarkers using Western blot. In human samples, resistant group has significantly higher miR-21 and lower Smad7 levels than sensitive group. In-vitro analysis demonstrated downregulated Smad7 after transfection with miR-21 mimics. When cells were transfected with Smad7 inhibitor, we observed increased drug resistance and changed levels of EMT-biomarkers after chemoradiotherapy, suggesting that downregulation of Smad7 decreased the sensitivity through EMT. When the cells were transfected with miR-21 inhibitor alone, we found increased sensitivity to chemoradiotherapy through EMT. However, such effects were attenuated when Smad7 was also downregulated after cotransfection. In summary, we provided clinical and experimental evidence that decreased miR-21 may improve drug resistance through EMT by direct targeting Smad7 in cervical cancer. Our data suggest that miR-21/Smad7 pathway may be an effective target for drug resistance in cervical cancer treatment.


Assuntos
Quimiorradioterapia , MicroRNAs/fisiologia , Proteína Smad7/fisiologia , Neoplasias do Colo do Útero/terapia , Adulto , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Feminino , Células HeLa , Humanos , MicroRNAs/antagonistas & inibidores , Pessoa de Meia-Idade , Transdução de Sinais/fisiologia , Neoplasias do Colo do Útero/patologia
4.
J Dent Res ; 98(12): 1376-1385, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31499015

RESUMO

Tooth morphogenesis involves dynamic changes in shape and size as it proceeds through the bud, cap, and bell stages. This process requires exact regulation of cell proliferation and differentiation. Smad7, a general antagonist against transforming growth factor-ß (TGF-ß) signaling, is necessary for maintaining homeostasis and proper functionality in many organs. While TGF-ß signaling is widely involved in tooth morphogenesis, the precise role of Smad7 in tooth development remains unknown. In this study, we showed that Smad7 is expressed in the developing mouse molars with a high level in the dental epithelium but a moderate to weak level in the dental mesenchyme. Smad7 deficiency led to a profound decrease in tooth size primarily due to a severely compromised cell proliferation capability in the dental epithelium. Consistent with the tooth shrinkage phenotype, RNA sequencing (RNA-seq) analysis revealed that Smad7 ablation downregulated genes referred to epithelial cell proliferation and cell cycle G1/S phase transition, whereas the upregulated genes were involved in responding to TGF-ß signaling and cell cycle arrest. Among these genes, the expression of Cdkn1a (encoding p21), a negative cell proliferation regulator, was remarkably elevated in parallel with the diminution of Ccnd1 encoding the crucial cell cycle regulator cyclin D1 in the dental epithelium. Meanwhile, the expression level of p-Smad2/3 was ectopically elevated in the developing tooth germ of Smad7 null mice, indicating the hyperactivation of the canonical TGF-ß signaling. These effects were reversed by addition of TGF-ß signaling inhibitor in cell cultures of Smad7-/- molar tooth germs, with rescued expression of cyclin D1 and cell proliferation rate. In sum, our studies demonstrate that Smad7 functions primarily as a positive regulator of cell proliferation via inhibition of the canonical TGF-ß signaling during dental epithelium development and highlight a crucial role for Smad7 in regulating tooth size.


Assuntos
Proliferação de Células , Odontogênese , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Pontos de Checagem do Ciclo Celular , Ciclina D1/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Mesoderma , Camundongos , Camundongos Knockout , Transdução de Sinais , Proteína Smad7/genética , Germe de Dente/crescimento & desenvolvimento
5.
Cell Death Dis ; 10(6): 387, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-31097718

RESUMO

Recent reports indicate that Smad7 promotes skeletal muscle differentiation and growth. We previously documented a non-canonical role of nuclear Smad7 during myogenesis, independent of its role in TGF-ß signaling. Here further characterization of the myogenic function of Smad7 revealed ß-catenin as a Smad7 interacting protein. Biochemical analysis identified a Smad7 interaction domain (SID) between aa575 and aa683 of ß-catenin. Reporter gene analysis and chromatin immunoprecipitation demonstrated that Smad7 and ß-catenin are cooperatively recruited to the extensively characterized ckm promoter proximal region to facilitate its muscle restricted transcriptional activation in myogenic cells. Depletion of endogenous Smad7 and ß-catenin in muscle cells reduced ckm promoter activity indicating their role during myogenesis. Deletion of the ß-catenin SID substantially reduced the effect of Smad7 on the ckm promoter and exogenous expression of SID abolished ß-catenin function, indicating that SID functions as a trans dominant-negative regulator of ß-catenin activity. ß-catenin interaction with the Mediator kinase complex through its Med12 subunit led us to identify MED13 as an additional Smad7-binding partner. Collectively, these studies document a novel function of a Smad7-MED12/13-ß-catenin complex at the ckm locus, indicating a key role of this complex in the program of myogenic gene expression underlying skeletal muscle development and regeneration.


Assuntos
Desenvolvimento Muscular/genética , Proteína Smad7/fisiologia , beta Catenina/fisiologia , Animais , Diferenciação Celular , Linhagem Celular , Regulação da Expressão Gênica , Camundongos , Proteína Smad7/genética , Proteína Smad7/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
Transl Res ; 204: 72-81, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30342000

RESUMO

Overexpression of SMAD7-a hallmark inhibitor of transforming growth factor ß (TGFß) signaling-has been documented and related with adverse prognosis in a number of epithelial malignancies, suggesting that it may be responsible for resistance to TGFß-induced growth arrest of cancer cells. The involvement of SMAD7 in development and progression of malignant melanoma is unclear, and its expression has not been characterized so far at the protein level in clinical melanoma tissue samples. We evaluated SMAD7 expression in 205 skin melanoma primary tumors by immunohistochemistry and correlated the findings with clinicopathological profiles of patients. Melanocytic SMAD7 was evidenced in 204 cases, and the expression pattern was predominantly nuclear. High expression of SMAD7 was positively associated with several features of tumor aggressiveness, for example, presence of ulceration (P < 0.001), higher tumor thickness (P < 0.001), and mitotic rate (P < 0.001), but not presence of regional or distant metastases. Moreover, high SMAD7 expression independently predicted unfavorable outcome: melanoma-specific survival (hazard ratio = 3.16, P < 0.001) and recurrence-free survival (hazard ratio = 2.88, P < 0.001). Taken together, our results underline the importance of TGFß signaling in cancer and define SMAD7 as a marker of aggressive tumor behavior and adverse clinical outcomes in melanoma patients.


Assuntos
Melanoma/mortalidade , Neoplasias Cutâneas/mortalidade , Proteína Smad7/fisiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Imuno-Histoquímica , Masculino , Melanócitos/química , Melanoma/química , Melanoma/patologia , Pessoa de Meia-Idade , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/química , Neoplasias Cutâneas/patologia , Proteína Smad7/análise , Fator de Crescimento Transformador beta/fisiologia , Adulto Jovem
7.
Carcinogenesis ; 40(1): 112-120, 2019 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-30219864

RESUMO

SMA- and MAD-related protein 7 (SMAD7) is a general inhibitor of transforming growth factor-ß (TGF-ß) signaling that acts through interaction and degradation of TGF-ß receptors. SMAD7 has been demonstrated to be transcriptionally upregulated in chemical-induced skin tumors and TGF-ß-treated normal keratinocytes. To evaluate the function of SMAD7 in skin carcinogenesis in vivo, Smad7 transgenic mice that specifically express either wild-type (WT) SMAD7 (TG-Smad7-WT) or mutant SMAD7 (TG-Smad7-MT) in keratinocytes, as well as Smad7 keratinocyte-specific knockout (Smad72f/2f-K14Cre) mice, were subjected to chemical-induced skin carcinogenesis. WT-SMAD7-expressing transgenic mice showed significantly greater papilloma formation than did non-TG control and Smad7-MT mice. The expression of WT-SMAD7 attenuated DNA damage-induced apoptosis in epidermal keratinocytes by stimulating the ATM-dependent DNA repair pathway. Nonetheless, overexpression of WT-SMAD7 caused a susceptibility to 12-O-tetradecanoylphorbol-13-acetate-induced epidermal hyperproliferation through activation of epidermal growth factor (EGF) signaling. In agreement with the transgenic mouse data, keratinocyte-specific deletion of SMAD7 markedly suppressed the tumor formation by inhibiting ATM and epidermal growth factor receptor (EGFR) signaling. Moreover, specific inhibition of EGFR signaling attenuated the hyperproliferation and tumor formation in TG-Smad7-WT mice. Taken together, these data support a novel role for SMAD7 as a tumor promoter in skin carcinogenesis where SMAD7 stimulates the DNA repair pathway and EGFR signaling activation.


Assuntos
Reparo do DNA , Receptores ErbB/fisiologia , Queratinócitos/fisiologia , Neoplasias Cutâneas/etiologia , Proteína Smad7/fisiologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Proliferação de Células , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Acetato de Tetradecanoilforbol/farmacologia
8.
Phytother Res ; 32(12): 2568-2576, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30251285

RESUMO

Stevia has been shown to prevent oxidative stress and inflammation in carbon tetrachloride­induced cirrhosis models. This study aimed to investigate the ability of an aqueous extract of stevia (AES) to prevent thioacetamide (TAA)­induced cirrhosis in rats and to explore its mechanism of action. Liver cirrhosis was established by administering TAA (200 mg/kg by i.p. injections three times a week for 10 weeks); AES was administered (100 mg/kg by gavage daily) during the TAA treatment. Liver damage and fibrosis were evaluated, and the profibrotic pathways were analyzed by western blotting and immunohistochemistry. TAA increased nuclear factor kappa B (NF­κB) and pro­inflammatory cytokine production, as well as the malondialdehyde and 4­hydroxynonenal levels, whereas the glutathione/glutathione disulfide and nuclear factor­E2­related factor 2 (Nrf2) levels were decreased. Moreover, TAA increased collagen production, hepatic stellate cell (HSC) activation, and expression of profibrogenic mediators. TAA­treated rats that had been exposed to Mn2+ exhibited altered striatal dopamine turnover, indicating hepatic encephalopathy. AES partially or completely prevented all of these effects. AES showed antioxidant, anti­inflammatory, and antifibrotic properties, probably because of its capacity to induce Nrf2 expression, reduce NF­κB expression, and block several profibrogenic signaling pathways, subsequently inhibiting HSC activation and preventing fibrosis and dopamine turnover.


Assuntos
Células Estreladas do Fígado/efeitos dos fármacos , Cirrose Hepática Experimental/prevenção & controle , Fator 2 Relacionado a NF-E2/fisiologia , NF-kappa B/fisiologia , Extratos Vegetais/uso terapêutico , Proteína Smad7/fisiologia , Stevia , Fator de Crescimento Transformador beta/fisiologia , Animais , Células Estreladas do Fígado/fisiologia , Cirrose Hepática Experimental/induzido quimicamente , Masculino , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Tioacetamida
9.
Zhonghua Xin Xue Guan Bing Za Zhi ; 46(6): 450-457, 2018 Jun 24.
Artigo em Chinês | MEDLINE | ID: mdl-29925181

RESUMO

Objective: To explore the effect of microRNA-21 (miR-21) on myocardial fibrosis in mice with chronic viral myocarditis (CVMC) and related mechanisms. Methods: Forty 4-week-old Balb/c male mice were randomly divided into 4 groups (n=10 each): phosphate buffer saline (PBS) group, CVMC group, CVMC+miR-21 inhibitor group, CVMC+isotype control group. The first injection of Coxsackie virus B3 (CVB3) or PBS was performed on day 0, and the total study time was 42 days. Each mouse in CVMC group, CVMC+miR-21 inhibitor group and CVMC+isotype control group was intraperitoneally (i.p) injected with 100TCID50 CVB3 0.1, 0.15, and 0.2 ml on day 0, 14, and 28, respectively. The mice in PBS group were i.p injected with the same dose of PBS at the same time point. After the initial infection, each mouse in CVMC+miR-21 inhibitor group and CVMC+isotype control group was intravenously injected with 0.1 ml miR-21 inhibitor or 0.1 ml isotype control, on day 14 and 28. Cardiac function was measured on surviving mice of 4 groups by echocardiography on day 42. Then, the hearts were removed aseptically to observe the expressions of green fluorescence protein (GFP). The myocardial pathological changes were examined with HE, Masson staining and the myocardial pathological scores (PS), the collagen volume fraction (CVF) were calculated respectively. The levels of miR-21, collagen typeⅠ-A1 (COL1-A1) and collagen type Ⅲ-A1 (COL3-A1) mRNA in heart were detected by quantitative real-time polymerase chain reaction (RT-qPCR). Furthermore, the expressions of transforming growth factor-ß1 (TGF-ß1) and mothers against decapentaplegic homolog 7(Smad7) in heart were determined with Western blot assay. Results: (1) Cardiac function in 4 groups: Compared with PBS group, left ventricular end systolic diameter (LVESD) and left ventricular end diastolic diameter (LVEDD) were markedly increased in CVMC group and CVMC+isotype control group (all P<0.05), whereas the left ventricular ejection fraction (LVEF) was decreased (P<0.05). LVESD and LVEDD were significantly decreased, and LVEF was increased in CVMC+miR-21 inhibitor group compared with those in CVMC group and CVMC+isotype control group (all P<0.05). (2) Myocardial pathological changes: The expressions of GFP in CVMC+miR-21 inhibitor group and CVMC+isotype control group were visible in heart tissues frozen sections. The hearts in CVMC group and CVMC+isotype control group were enlarged and stiff, inflammatory cells were visible and significantly increased myocardial fibrosis was evidenced in mice of these two groups. Higher PS and CVF were evidenced in CVMC group (PS: 1.14±0.69 vs. 0, CVF: (17.86±2.61)% vs. (5.70±1.42)%, all P<0.05) and CVMC+isotype control group(PS: 1.00±0.63 vs. 0, CVF: (16.78±2.58)% vs. (5.70±1.42)%, all P<0.05) compared to PBS group. Compared with CVMC group and CVMC+isotype control group, degree of cardiac fibrosis was reduced in mice of CVMC+miR-21 inhibitor group (CVF: (11.01±2.55)% vs. (17.86±2.61)%, (11.01±2.55)% vs. (16.78±2.58)%, all P<0.05), whereas PS were similar between them (PS: 0.89±0.60 vs. 1.14±0.69, 0.89±0.60 vs. 1.00±0.63, all P>0.05). (3) Cardiac expressions of miR-21, COL1-A1 and COL3-A1 mRNA: The cardiac expressions of miR-21, COL1-A1 mRNA, COL3-A1mRNA in CVMC group and CVMC+isotype control group were markedly higher than those in PBS group (all P<0.05), which were significantly downregulated in CVMC+miR-21 inhibitor group (all P<0.05 vs. CVMC group and CVMC+isotype control group). (4) The cardiac expressions of TGF-ß1 and Smad7 protein: The cardiac expressions of TGF-ß1 protein in CVMC group and CVMC+isotype control group were markedly higher, whereas the cardiac Smad7 protein expressions were significantly lower (all P<0.05) than those in PBS group (all P<0.05), these changes could be reversed in CVMC+miR-21 inhibitor group (P<0.05 vs. CVMC group and CVMC+isotype control group). Conclusions: Our results suggest that miR-21 contributes to the myocardial fibrosis in CVMC mice through modulating TGF-ß1/Smad7 signaling pathway.


Assuntos
Cardiomiopatias , MicroRNAs , Miocardite , Animais , Cardiomiopatias/metabolismo , Enterovirus Humano B , Fibrose , Masculino , Camundongos , MicroRNAs/fisiologia , Miocardite/metabolismo , Miocardite/virologia , Miocárdio , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta1/fisiologia
10.
Br J Dermatol ; 177(6): 1633-1643, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28580633

RESUMO

BACKGROUND: Transforming growth factor (TGF)-ß1 exerts inhibitory effects on keratinocyte proliferation. OBJECTIVES: To examine whether Smad7, a known inhibitor of TGF-ß1 signalling, is involved in psoriasis-associated keratinocyte hyperproliferation. METHODS: Smad7 was evaluated in skin sections of patients with psoriasis and healthy controls and in mice with Aldara-induced skin pathology by real-time polymerase chain reaction and immunohistochemistry. To assess whether Smad7 positively regulates in vivo keratinocyte growth, mice treated with Aldara received daily cutaneous administration of Smad7 antisense oligonucleotide (AS). Keratin (K)6 and K16, cell-cycle-associated factors, cell-cycle and cell proliferation were evaluated in HaCaT cells either treated with Smad7 AS or transfected with Smad7 plasmid and in mice given Smad7 AS. RESULTS: Smad7 was highly expressed in keratinocytes of patients with psoriasis and of mice treated with Aldara. In HaCaT cells, Smad7 knockdown inhibited cell growth, reduced K6 and K16 expression and promoted accumulation of cells in the S-phase of the cell cycle. Smad7-deficient keratinocytes exhibited reduced levels of CDC25A protein, a phosphatase that facilitates progression of cells through the S-phase, and hyperphosphorylation of eukaryotic initiation factor 2 (eIF2)α, a negative regulator of CDC25 protein translation. Consistently, Smad7 overexpression in HaCaT cells was followed by induction of K6 and K16 and increased cell proliferation. Topical application of Smad7 AS to Aldara-treated mice reduced epidermal thickness. CONCLUSIONS: Our data show that Smad7 is overexpressed in human and murine psoriasis and suggest a key role of this molecule in the control of keratinocyte proliferation.


Assuntos
Proliferação de Células/fisiologia , Queratinócitos/patologia , Psoríase/patologia , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Dermatite/fisiopatologia , Relação Dose-Resposta a Droga , Epiderme/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia , Proteína Smad7/deficiência , Regulação para Cima/fisiologia
11.
J Pathol ; 242(3): 334-346, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28418194

RESUMO

Angiostasis mediated by interferon (IFN)-γ is a key mechanism of anti-tumour immunity; however, the effect of IFN-γ on host vascular endothelial growth factor A (VEGFA)-expressing cells during tumour progression is still elusive. Here, we developed transgenic mice with IFN-γ receptor (IFNγR) expression under control of the Vegfa promoter (V-γR). In these mice, the IFN-γ responsiveness of VEGFA-expressing cells led to dramatic growth suppression of transplanted lung carcinoma cells. Surprisingly, increased mortality and tumour metastasis were observed in the tumour-bearing V-γR mice, in comparison with the control wild-type and IFNγR-deficient mice. Further study showed that perivascular cells were VEGFA-expressing cells and potential IFN-γ targets. In vivo, tumour vascular perfusion and pericyte association with blood vessels were massively disrupted in V-γR mice. In vitro, IFN-γ inhibited transforming growth factor-ß signalling by upregulating SMAD7, and therefore downregulated N-cadherin expression in pericytes. Importantly, IFN-γ neutralization in vivo with a monoclonal antibody reduced tumour metastasis. Together, the results suggest that IFNγR-mediated dissociation of perivascular cells from blood vessels contributes to the acceleration of tumour metastasis. Thus, the inhibition of tumour growth via IFN-γ-induced angiostasis might also accelerate tumour metastasis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Neoplasias Pulmonares/fisiopatologia , Receptores de Interferon/fisiologia , Animais , Caderinas/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Fibroblastos/metabolismo , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Metástase Neoplásica , Transplante de Neoplasias , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Pericitos/metabolismo , Receptores de Interferon/deficiência , Receptores de Interferon/metabolismo , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Regulação para Cima/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
J Hepatol ; 64(1): 110-7, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26334580

RESUMO

BACKGROUND & AIM: Hydrogen peroxide-inducible clone-5 (Hic-5), also named as transforming growth factor beta-1-induced transcript 1 protein (Tgfb1i1), was found to be induced by TGF-ß. Previous studies have shown that TGF-ß is a principal mediator of hepatic stellate cell (HSC) activation in liver fibrosis. However, this process remains elusive. In this study, we aimed to define the role of Hic-5 in HSC activation and liver fibrosis. METHODS: We examined the expression levels of Hic-5 during HSCs activation and in fibrotic liver tissues by quantitative real-time reverse transcriptase polymerase chain reaction, Western blot and immunohistochemistry. Hic-5 knockout (KO) and wild-type (WT) mice were subjected to bile duct ligation (BDL) or carbon tetrachloride (CCl4) injection to induce liver fibrosis. RESULTS: Hic-5 expression was strongly upregulated in activated HSCs of the human fibrotic liver tissue and BDL or CCl4-induced mouse liver fibrosis. Hic-5 deficiency significantly attenuated mouse liver fibrosis and HSC activation. Furthermore, Hic-5 knockdown by siRNA in vivo repressed CCl4-induced liver fibrosis in mice. Mechanistically, the absence of Hic-5 significantly inhibited the TGF-ß/Smad2 signaling pathway, proved by increasing Smad7 expression, resulting in reduced collagen production and α-smooth muscle actin expression in the activated HSCs. CONCLUSION: Hic-5 deficiency attenuates the activation of HSCs and liver fibrosis though reducing the TGF-ß/Smad2 signaling by upregulation of Smad7. Thus, Hic-5 can be regarded as a potential therapeutic target for liver fibrosis.


Assuntos
Proteínas do Citoesqueleto/deficiência , Proteínas de Ligação a DNA/deficiência , Células Estreladas do Fígado/fisiologia , Proteínas com Domínio LIM/deficiência , Cirrose Hepática/etiologia , Proteína Smad7/fisiologia , Actinas/análise , Animais , Tetracloreto de Carbono , Células Cultivadas , Proteínas do Citoesqueleto/análise , Proteínas de Ligação a DNA/análise , Humanos , Proteínas com Domínio LIM/análise , Camundongos , Camundongos Endogâmicos C57BL , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Regulação para Cima
13.
J Physiol ; 593(11): 2479-97, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25854148

RESUMO

KEY POINTS: Smad7 is an intracellular antagonist of transforming growth factor-ß signalling pathways and modulates muscle growth in vivo. Loss of Smad7 results in decreased muscle mass, reduced force generation, fibre type switching from glycolytic towards oxidative type and delayed recovery from injury. Upregulated Smad2/3 signalling in Smad7(-/-) muscle results in reduced myoblast proliferation and differentiation. Smad7 is an important regulator of muscle growth and may be a potential intracellular therapeutic target for muscle disorders. ABSTRACT: The transforming growth factor-ß (TGF-ß) family of growth factors plays an essential role in mediating cellular growth and differentiation. Myostatin is a muscle-specific member of the TGF-ß superfamily and a negative regulator of muscle growth. Myostatin inhibitors are currently being pursued as therapeutic options for muscle disorders. Smad7 inhibits intracellular myostatin signalling via Smad2/3, and thus presents a means of regulating myostatin and potentiating muscle growth. We investigated the functional loss of Smad7 on muscle in vivo by examining muscle growth and differentiation in mice deficient in Smad7 (Smad7(-/-) ). Smad7(-/-) mice showed reduced muscle mass, hypotrophy and hypoplasia of muscle fibres, as well as an increase in oxidative fibre types. Examination of muscle strength showed reduced force generation in vivo and ex vivo compared to wild-type controls. Analysis of muscle regeneration showed a delay in recovery, probably as a result of decreased activation, proliferation and differentiation of satellite cells, as confirmed in vitro. Additionally, myostatin expression was upregulated in Smad7(-/-) muscle. Our findings suggest that increased Smad2/3 signalling in the absence of Smad7 inhibition impedes muscle growth and regeneration. Taken together, our experiments demonstrate that Smad7 is an important mediator of muscle growth in vivo. Our studies enhance our understanding of in vivo TGF-ß pathway modulation and suggest that Smad7 may be an important therapeutic target for muscle disorders.


Assuntos
Desenvolvimento Muscular , Músculo Esquelético/crescimento & desenvolvimento , Proteína Smad7/genética , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Feminino , Masculino , Camundongos Knockout , Força Muscular , Músculo Esquelético/citologia , Músculo Esquelético/lesões , Mioblastos/citologia , Regeneração , Proteína Smad7/fisiologia
14.
J Biol Chem ; 290(19): 12222-36, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25809484

RESUMO

The ability of the active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), to transcriptionally modulate Smads to inhibit Th17 differentiation and experimental autoimmune encephalomyelitis (EAE) has not been adequately studied. This study reports modulation of Smad signaling by the specific binding of the VDR along with its heterodimeric partner RXR to the negative vitamin D response element on the promoter of Smad7, which leads to Smad7 gene repression. The vitamin D receptor-mediated increase in Smad3 expression partially explains the IL10 augmentation seen in Th17 cells. Furthermore, the VDR axis also modulates non-Smad signaling by activating ERK during differentiation of Th17 cells, which inhibits the Th17-specific genes il17a, il17f, il22, and il23r. In vivo EAE experiments revealed that, 1,25(OH)2D3 suppression of EAE correlates with the Smad7 expression in the spleen and lymph nodes. Furthermore, Smad7 expression also correlates well with IL17 and IFNγ expression in CNS infiltered inflammatory T cells. We also observed similar gene repression of Smad7 in in vitro differentiated Th1 cells when cultured in presence of 1,25(OH)2D3. The above canonical and non-canonical pathways in part address the ability of 1,25(OH)2D3-VDR to inhibit EAE.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Enzimológica da Expressão Gênica , Proteína Smad7/antagonistas & inibidores , Linfócitos T Auxiliares-Indutores/citologia , Vitamina D/análogos & derivados , Animais , Células COS , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Chlorocebus aethiops , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Interferon gama/metabolismo , Linfonodos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Fosforilação , Regiões Promotoras Genéticas , Elementos de Resposta , Proteína Smad7/fisiologia , Células Th17 , Transcrição Gênica , Vitamina D/química
15.
Transpl Int ; 28(2): 232-45, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25266172

RESUMO

Calcineurin inhibitors (CNIs) are potent immunosuppressants with associated long-term nephrotoxicity mediated by tubular epithelial cell injury and arterial vasoconstriction. We hypothesized that CNI-induced renal injury is regulated by specific microRNAs (miRNAs). In this study, we found that 46 miRNAs were significantly altered in human proximal tubular epithelial cells (HPTECs) following exposure to cyclosporine A (CsA), particularly miR-21 (5.47 ± 0.47-fold versus vehicle, P = 0.002). This increase was accompanied by alterations in epithelial-mesenchymal transformation (EMT) markers including vimentin (2.80 ± 0.28-fold; P = 0.03), S100A4 (2.29 ± 0.29-fold; P = 0.04), and α-SMA (5.0 ± 0.31-fold; P = 0.03). Notably, transfection of HPTECs with miR-21 precursor also resulted in significant induction of EMT-associated genes, which were inhibited by a single-stranded nucleic acid inhibitor of miR-21. miR-21 induction resulted in a rapid increase of phosphorylated AKT and downregulation of PTEN. While CsA induces SMAD7 downregulation and TGF-ß1 upregulation in HPTECs, such changes were independent of miR-21. Moreover, there was no effect on ERK phosphorylation. We confirmed these changes using a mouse model of CsA toxicity. Collectively, our results suggest that miR-21 mediates CsA nephrotoxicity via PTEN/AKT signaling pathway. Further exploration into the epigenetic response to CsA exposure may provide new therapeutic targets to ameliorate CsA nephrotoxicity.


Assuntos
Inibidores de Calcineurina/efeitos adversos , Ciclosporina/efeitos adversos , Rim/efeitos dos fármacos , MicroRNAs/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Transdução de Sinais , Aloenxertos , Animais , Células Cultivadas , Transição Epitelial-Mesenquimal , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Fibrose , Humanos , Rim/patologia , Camundongos , PTEN Fosfo-Hidrolase/fisiologia , Fosforilação , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta/fisiologia
16.
Asian J Androl ; 17(3): 487-92, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25532569

RESUMO

Transforming growth factor-ß1 (TGF-ß1) has been identified as one of the most important fibrogenic cytokines associated with Peyronie's disease (PD). The mothers against decapentaplegic homolog 7 (SMAD7) is an inhibitory Smad protein that blocks TGF-ß signaling pathway. The aim of this study was to examine the anti-fibrotic effect of the SMAD7 gene in primary fibroblasts derived from human PD plaques. PD fibroblasts were pretreated with the SMAD7 gene and then stimulated with TGF-ß1. Treated fibroblasts were used for Western blotting, fluorescent immunocytochemistry, hydroxyproline determination, and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling assays. Overexpression of the SMAD7 gene inhibited TGF-ß1-induced phosphorylation and nuclear translocation of SMAD2 and SMAD3, transdifferentiation of fibroblasts into myofibroblasts, and quashed TGF-ß1-induced production of extracellular matrix protein and hydroxyproline. Overexpression of the SMAD7 gene decreased the expression of cyclin D1 (a positive cell cycle regulator) and induced the expression of poly (ADP-ribose) polymerase 1, which is known to terminate Smad-mediated transcription, in PD fibroblasts. These findings suggest that the blocking of the TGF-ß pathway by use of SMAD7 may be a promising therapeutic strategy for the treatment of PD.


Assuntos
Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Induração Peniana/patologia , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta1/farmacologia , Regulação para Cima/fisiologia , Células Cultivadas , Ciclina D1/efeitos dos fármacos , Ciclina D1/metabolismo , Proteínas da Matriz Extracelular/antagonistas & inibidores , Proteínas da Matriz Extracelular/efeitos dos fármacos , Proteínas da Matriz Extracelular/metabolismo , Fibrose/induzido quimicamente , Humanos , Hidroxiprolina/antagonistas & inibidores , Hidroxiprolina/efeitos dos fármacos , Hidroxiprolina/metabolismo , Masculino , Induração Peniana/tratamento farmacológico , Induração Peniana/fisiopatologia , Poli(ADP-Ribose) Polimerases/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteína Smad7/genética , Proteína Smad7/uso terapêutico , Transfecção , Fator de Crescimento Transformador beta1/efeitos adversos , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Regulação para Cima/genética
17.
Exp Gerontol ; 57: 149-54, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24862634

RESUMO

We recently reported that young (3 to 4months old) mice lacking Exon 1 of the Smad7 gene (S7ΔEx1 mice) show enhanced proliferation of neural stem and progenitor cells (NPCs) in the hippocampal dentate gyrus (DG) and in the subventricular zone (SVZ) of the lateral ventricles. It remained unclear, however, whether this phenotype would persist along aging, the latter typically being associated with a profound decrease in neurogenesis. Analysis of NPCs' proliferation based on the cell cycle marker PCNA in 12month-old S7ΔEx1 mice revealed a reversal of the phenotype. Hence, in contrast to their younger counterparts, 12month-old S7ΔEx1 mice had a reduced number of proliferating cells, compared to wildtype (WT) mice. At the same time, the survival of newly generated cells was enhanced in the aged transgenic animals. 12month-old S7ΔEx1 mice further displayed a reduced level of neurogenesis based on the numbers of cells expressing doublecortin (DCX), a marker for newborn neurons. The reduced neurogenesis in aged S7ΔEx1 mice was not due to a stem cell depletion, which might have occurred as a consequence of hyperproliferation in the young mice, since the number of Nestin and Sox2 positive cells was similar in WT and S7ΔEx1 mice. Instead, Nestin positive cells in the DG as well as primary neurosphere cultures derived from 12month-old S7ΔEx1 mice had a reduced capability to proliferate. However, after passaging, when released from their age- and niche-associated proliferative block, neurospheres from aged S7ΔEx1 mice regained the hyperproliferative property. Further, pSmad2 antibody staining intensity was elevated in the DG and SVZ of 12-month old transgenic compared to WT mice, indicating increased intracellular TGF-beta signaling in the aged S7ΔEx1 mice. In summary, this points toward differential effects of S7ΔEx1 on neurogenesis: (i) a hyperproliferation in young animals caused by a cell autonomous mechanism, and (ii) a TGF-beta dependent modulation of neurogenesis in aged S7ΔEx1 animals that abrogates the cell-intrinsic hyperproliferative properties and results in reduced proliferation, increased stem cell quiescence, and enhanced survival of newly generated cells.


Assuntos
Envelhecimento/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Proliferação de Células , Sobrevivência Celular , Proteína Duplacortina , Masculino , Camundongos
18.
Carcinogenesis ; 35(7): 1536-46, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24480808

RESUMO

Transforming growth factor-beta (TGF-ß) is deeply involved in colorectal cancer development and the disruption of the TGF-ß signaling in dysplastic cells is required for tumor to grow. Nevertheless, tumor cells express TGF-ß to escape the immune surveillance mediated by T cells. T-cell expression of Smad7, an intracellular inhibitor of the TGF-ß signaling, protects against colitis-associated colorectal cancer. However, whether Smad7 in T cells might influence colorectal cancer growth independently of chronic inflammation and which T-cell subset is involved in this process is unknown. To address this issue, T-cell-specific Smad7 transgenic mice and wild-type (WT) littermates were subcutaneously transplanted with syngenic MC38 colon carcinoma cells. Smad7Tg mice were resistant to tumor development compared with WT mice and protection was dependent on CD4(+) T cells. Smad7 expression in T cells increased the number of tumor-infiltrating Tbet/ROR-γ-t double-positive CD4 T cells characterized by the expression of tumor necrosis factor-alpha (TNF-α) and interferon-gamma but lower IL17A. The low expression of IL17A caused by the Smad7 expression in tumor-infiltrating CD4(+) T cells enabled the TNF-α-mediated killing of cancer cells both in vitro and in vivo, thus indicating that the Smad7-mediated plastic effect on T-cell phenotype induces protection against colorectal cancer.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Colorretais/patologia , Interleucina-17/fisiologia , Linfócitos do Interstício Tumoral/imunologia , Proteína Smad7/fisiologia , Células Th17/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Interferon gama/metabolismo , Linfócitos do Interstício Tumoral/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Th17/patologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/genética
19.
J Bone Miner Res ; 29(3): 716-24, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23959527

RESUMO

Craniosynostosis, the premature fusion of one or more sutures between the calvarial bones, is a common birth defect. Mutations in genes encoding receptors for the transforming growth factor-beta (TGFß) family of signaling molecules have been associated with craniosynostosis, but how TGFß signaling is regulated during suture development is not known. In the present study, we found that expression of Smad2 and Smad3, intracellular mediators of canonical TGFß signaling, gradually increases during early postnatal suture development in rat in both the coronal suture (CS), which remains patent throughout life, and the posterior frontal suture (PFS), which undergoes programmed closure by postnatal day 22. The amounts of phosphorylated Smad2 and Smad3 proteins showed a similar gradual increase in the PFS and CS, but in the CS, Smad2/3 activation was suppressed after neonatal day 10. The suppression of Smad2/3 activation in the CS correlated with upregulation of Smad7 expression. We demonstrate that siRNA-mediated knockdown of Smad7 caused increased phosphorylation of Smad2 and Smad3 and induced osseous obliteration of the CS from postnatal days 10 to 22. The Smad7 siRNA-induced CS closure was associated with significantly increased levels of Fgf10 and phosphorylated ERK1/2 in the suture mesenchyme. Moreover, addition of the Erk1/2 inhibitor U0126 partially blocked Smad7-siRNA-induced CS closure. These findings suggest that canonical TGFß signaling induces suture closure at least in part through activation of FGF and ERK signaling and that Smad7 plays an important role in maintaining suture patency by suppressing canonical TGFß signaling during suture development.


Assuntos
Suturas Cranianas/fisiologia , Transdução de Sinais/fisiologia , Proteína Smad7/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Sequência de Bases , Suturas Cranianas/metabolismo , Primers do DNA , Técnicas de Silenciamento de Genes , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Proteína Smad7/genética , Proteína Smad7/metabolismo
20.
Toxicol Lett ; 224(2): 175-85, 2014 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-24211420

RESUMO

Conversion of hepatic stellate cells (HSCs) into hepatic myofibroblasts is a necessary event during the development of liver fibrosis. DNA methyltransferase 1 (DNMT1), which catalyzes DNA methylation and subsequently leads to the transcriptional repression of profibrotic genes, is selectively induced in myofibroblasts from diseased livers. Treatment of HSC with the DNA methylation inhibitor, 5-aza-2'-deoxycytidine (5-azadC), prevented TGF-ß1-induced proliferation and alpha-smooth muscle actin (α-SMA) and collagen expression. 5-AzadC also rescued TGF-ß1-induced suppression of Smad7 expression which occurs during HSC activation. Similarly, silencing the expression of the DNMT1 gene ameliorated the suppression of Smad7 expression by TGF-ß1. In addition, DNMT1 inhibition, by 5-azadC or DNMT1 silencing, prevented the phosphorylation of Smad2 and Smad3. These studies suggest that epigenetic repression of Smad7 promotes the phosphorylation of Smad2 and Smad3 that may be an important molecular mechanism for perpetuated HSC activation and liver fibrosis.


Assuntos
DNA (Citosina-5-)-Metiltransferases/fisiologia , Células Estreladas do Fígado/fisiologia , Cirrose Hepática Experimental/etiologia , Proteína Smad7/fisiologia , Animais , DNA (Citosina-5-)-Metiltransferase 1 , Epigênese Genética , Masculino , Fosforilação , Ratos , Ratos Sprague-Dawley , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Proteína Smad7/genética , Fator de Crescimento Transformador beta1/farmacologia , Cicatrização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...