Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
Mol Ther Nucleic Acids ; 33: 180-190, 2023 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-37449045

RESUMO

Transforming growth factor ß (TGF-ß)/Smad3 plays a vital role in hypertensive cardiac fibrosis. The long non-coding RNA (lncRNA) Erbb4-IR is a novel Smad3-dependent lncRNA that mediates kidney fibrosis. However, the role of Erbb4-IR in hypertensive heart disease remains unexplored and was investigated in the present study by ultrasound-microbubble-mediated silencing of cardiac Erbb4-IR in hypertensive mice induced by angiotensin II. We found that chronic angiotensin II infusion induced hypertension and upregulated cardiac Erbb4-IR, which was associated with cardiac dysfunction, including a decrease in left ventricle ejection fraction (LVEF) and LV fractional shortening (LVFS) and an increase in LV mass. Knockdown of cardiac Erbb4-IR by Erbb4-IR short hairpin RNA (shRNA) gene transfer effectively improved the angiotensin II-induced deterioration of cardiac function, although blood pressure was not altered. Furthermore, silencing cardiac Erbb4-IR also inhibited angiotensin II-induced progressive cardiac fibrosis, as evidenced by reduced collagen I and III, alpha-smooth muscle actin (α-SMA), and fibronectin accumulation. Mechanistically, improved hypertensive cardiac injury by specifically silencing cardiac Erbb4-IR was associated with increased myocardial Smad7 and miR-29b, revealing that Erbb4-IR may target Smad7 and miR-29b to mediate angiotensin II-induced hypertensive cardiac fibrosis. In conclusion, Erbb4-IR is pathogenic in angiotensin II (Ang II)-induced cardiac remodeling, and targeting Erbb4-IR may be a novel therapy for hypertensive cardiovascular diseases.

3.
Int J Biol Sci ; 19(2): 521-536, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36632461

RESUMO

Neuropeptide Y (NPY) is produced by the nerve system and may contribute to the progression of CKD. The present study found the new protective role for NPY in AKI in both patients and animal models. Interestingly, NPY was constitutively expressed in blood and resident kidney macrophages by co-expressing NPY and CD68+ markers, which was lost in patients and mice with AKI-induced by cisplatin. Unexpectedly, NPY was renoprotective in AKI as mice lacking NPY developed worse renal necroinflammation and renal dysfunction in cisplatin and ischemic-induced AKI. Importantly, NPY was also a therapeutic agent for AKI because treatment with exogenous NPY dose-dependently inhibited cisplatin-induced AKI. Mechanistically, NPY protected kidney from AKI by inactivating M1 macrophages via the Y1R-NF-κB-Mincle-dependent mechanism as deleting or silencing NPY decreased Y1R but increased NF-κB-Mincle-mediated M1macrophage activation and renal necroinflammation, which were reversed by addition of NPY or by silencing Mincle but promoted by blocking Y1R with BIBP 3226. Thus, NPY is renoprotective and may be a novel therapeutic agent for AKI. NPY may act via Y1R to protect kidney from AKI by blocking NF-κB-Mincle-mediated M1 macrophage activation and renal necroinflammation.


Assuntos
Injúria Renal Aguda , NF-kappa B , Neuropeptídeo Y , Receptores de Neuropeptídeo Y , Animais , Camundongos , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/induzido quimicamente , Cisplatino/farmacologia , Rim/efeitos dos fármacos , Rim/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Neuropeptídeo Y/metabolismo , Neuropeptídeo Y/farmacologia , Neuropeptídeo Y/uso terapêutico , Receptores de Neuropeptídeo Y/metabolismo
4.
J Nat Med ; 77(1): 73-86, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36115008

RESUMO

Excessive intrahepatocellular lipid accumulation or steatosis is caused by abnormal lipid metabolism and a common character of nonalcoholic fatty liver disease (NAFLD), which may progress into cirrhosis and hepatocellular cancer. Andrographolide (Andro) is the primary active ingredient extracted from Andrographis paniculata, showing a protective role against dietary steatosis with the mechanism not fully understood. In this study, we showed that administration of Andro (50, 100, and 200 mg/kg/day for 8 weeks, respectively) attenuated obesity and metabolic syndrome in high-fat diet (HFD)-fed mice with improved glucose tolerance, insulin sensitivity, and reduced hyperinsulinemia, hyperglycemia, and hyperlipidemia. HFD-fed mice presented hepatic steatosis, which was significantly prevented by Andro. In vitro, Andro decreased the intracellular lipid droplets in oleic acid-treated LO2 cells. The selected RT-PCR array revealed a robust expression suppression of the fatty acid transport proteins (FATPs) by Andro treatment. Most importantly, we found that Andro consistently reduced the expression of FATP2 in both the oleic acid-treated LO2 cells and liver tissues of HFD-fed mice. Overexpression of FATP2 abolished the lipid-lowering effect of Andro in oleic acid-treated LO2 cells. Andro treatment also reduced the fatty acid uptake in oleic acid-treated LO2 cells, which was blunted by FATP2 overexpression. Collectively, our findings reveal a novel mechanism underlying the anti-steatosis effect of Andro by suppressing FATP2-mediated fatty acid uptake, suggesting the potential therapeutic application of Andro in the treatment of NAFLD.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Animais , Camundongos , Coenzima A Ligases/metabolismo , Coenzima A Ligases/farmacologia , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Ácidos Graxos/uso terapêutico , Metabolismo dos Lipídeos , Fígado , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Ácido Oleico/metabolismo , Ácido Oleico/farmacologia , Ácido Oleico/uso terapêutico
5.
Front Physiol ; 13: 937878, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091385

RESUMO

Renal fibrosis is a common feature of all types of chronic kidney disease (CKD) and is tightly regulated by the TGF-ß/Smad3 pathway. Let-7i-5p belongs to the let-7 microRNA family with diverse biological functions. It has been reported that let-7i-5p suppresses fibrotic disease in the heart, lungs, and blood vessels, while the role of let-7i-5p in renal fibrosis remains limited. In this study, we aimed to investigate the role of let-7i-5p in renal fibrosis in a mouse model of unilateral ureteral obstruction (UUO) and TGF-ß1-stimulated renal tubular cell line TCMK1. The RNA-targeting CRISPR/Cas13d system was used to knock down let-7i-5p. Renal injury and fibrosis were determined by histological analysis, RT-PCR, Western blot, and immunostaining. Our results have shown that in the kidneys after UUO, the expression of let-7i-5p was significantly increased along with notable tubular injury and interstitial fibrosis. Electroporation of let-7i-targeting Cas13d plasmid efficiently knocked down let-7i-5p in kidneys after UUO with reduced tubular injury, fibrotic area, and expression of fibrotic marker genes α-SMA, fibronectin, and Col1a1. In TGF-ß1-stimulated TCMK1 cells, knockdown of let-7i-5p by Cas13d plasmid transfection also blunted the expression of fibrotic marker genes. Most importantly, the genomic locus of let-7i showed enriched binding of Smad3 as revealed by chromatin immunoprecipitation. In TCMK1 cells, the overexpression of Smad3 can directly induce the expression of let-7i-5p. However, the deletion of Smad3 abolished TGF-ß1-stimulated let-7i-5p expression. Collectively, these findings suggest that let-7i-5p is a Smad3-dependent microRNA that plays a pathogenic role in renal fibrosis. Let-7i-5p could be a promising target for the treatment of CKD-associated renal fibrosis.

6.
Biomolecules ; 12(3)2022 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-35327565

RESUMO

Beta (ß) cell dysfunction or loss is the common pathological feature in all types of diabetes mellitus (diabetes). Resolving the underlying mechanism may facilitate the treatment of diabetes by preserving the ß cell population and function. It is known that TGF-ß signaling plays diverse roles in ß cell development, function, proliferation, apoptosis, and dedifferentiation. Inhibition of TGF-ß signaling expands ß cell lineage in the development. However, deletion of Tgfbr1 has no influence on insulin demand-induced but abolishes inflammation-induced ß cell proliferation. Among canonical TGF-ß signaling, Smad3 but not Smad2 is the predominant repressor of ß cell proliferation in response to systemic insulin demand. Deletion of Smad3 simultaneously improves ß cell function, apoptosis, and systemic insulin resistance with the consequence of eliminated overt diabetes in diabetic mouse models, revealing Smad3 as a key mediator and ideal therapeutic target for type-2 diabetes. However, Smad7 shows controversial effects on ß cell proliferation and glucose homeostasis in animal studies. On the other hand, overexpression of Tgfb1 prevents ß cells from autoimmune destruction without influence on ß cell function. All these findings reveal the diverse regulatory roles of TGF-ß signaling in ß cell biology.


Assuntos
Diabetes Mellitus , Células Secretoras de Insulina , Insulinas , Animais , Proliferação de Células , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Células Secretoras de Insulina/metabolismo , Insulinas/metabolismo , Camundongos , Fator de Crescimento Transformador beta/metabolismo
7.
Theranostics ; 12(1): 379-395, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34987651

RESUMO

Rationale: Poor ß cell proliferation is one of the detrimental factors hindering islet cell replacement therapy for patients with diabetes. Smad3 is an important transcriptional factor of TGF-ß signaling and has been shown to promote diabetes by inhibiting ß cell proliferation. Therefore, we hypothesize that Smad3-deficient islets may be a novel cell replacement therapy for diabetes. Methods: We examined this hypothesis in streptozocin-induced type-1 diabetic mice and type-2 diabetic db/db mice by transplanting Smad3 knockout (KO) and wild type (WT) islets under the renal capsule, respectively. The effects of Smad3KO versus WT islet replacement therapy on diabetes and diabetic kidney injury were examined. In addition, RNA-seq was applied to identify the downstream target gene underlying Smad3-regulated ß cell proliferation in Smad3KO-db/db versus Smad3WT-db/db mouse islets. Results: Compared to Smad3WT islet therapy, treatment with Smad3KO islets produced a much better therapeutic effect on both type-1 and type-2 diabetes by significantly lowering serum levels of blood glucose and HbA1c and protected against diabetic kidney injuries by preventing an increase in serum creatinine and the development of proteinuria, mesangial matrix expansion, and fibrosis. These were associated with a significant increase in grafted ß cell proliferation and blood insulin levels, resulting in improved glucose intolerance. Mechanistically, RNA-seq revealed that compared with Smad3WT-db/db mouse islets, deletion of Smad3 from db/db mouse islets markedly upregulated E2F3, a pivotal regulator of cell cycle G1/S entry. Further studies found that Smad3 could bind to the promoter of E2F3, and thus inhibit ß cell proliferation via an E2F3-dependent mechanism as silencing E2F3 abrogated the proliferative effect on Smad3KO ß cells. Conclusion: Smad3-deficient islet replacement therapy can significantly improve both type-1 and type-2 diabetes and protect against diabetic kidney injury, which is mediated by a novel mechanism of E2F3-dependent ß cell proliferation.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Fator de Transcrição E2F3/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína Smad3/metabolismo , Animais , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
8.
Int J Mol Sci ; 22(15)2021 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-34360646

RESUMO

Diabetic nephropathy (DN) is one of the most common complications in diabetes mellitus and the leading cause of end-stage renal disease. TGF-ß is a pleiotropic cytokine and has been recognized as a key mediator of DN. However, anti-TGF-ß treatment for DN remains controversial due to the diverse role of TGF-ß1 in DN. Thus, understanding the regulatory role and mechanisms of TGF-ß in the pathogenesis of DN is the initial step towards the development of anti-TGF-ß treatment for DN. In this review, we first discuss the diverse roles and signaling mechanisms of TGF-ß in DN by focusing on the latent versus active TGF-ß1, the TGF-ß receptors, and the downstream individual Smad signaling molecules including Smad2, Smad3, Smad4, and Smad7. Then, we dissect the regulatory mechanisms of TGF-ß/Smad signaling in the development of DN by emphasizing Smad-dependent non-coding RNAs including microRNAs and long-non-coding RNAs. Finally, the potential therapeutic strategies for DN by targeting TGF-ß signaling with various therapeutic approaches are discussed.


Assuntos
Nefropatias Diabéticas/patologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Nefropatias Diabéticas/metabolismo , Humanos , Transdução de Sinais
9.
Artigo em Inglês | MEDLINE | ID: mdl-34257680

RESUMO

Chronic kidney disease (CKD) has become a global health issue, and there is increasing evidence showing the beneficial roles of traditional Chinese medicine (TCM) in CKD treatment. Here, we studied the renoprotective role of Mahuang decoction, a famous TCM prescription, in a rat CKD model induced with the combination of doxorubicin and adenine. Our data showed that intragastric administration of Mahuang decoction inhibited the loss of bodyweight and attenuated proteinuria, serum creatinine, and blood urea nitrogen in CKD rats. Kidney histological analysis revealed decreased tubulointerstitial injury and fibrosis in CKD rats treated with Mahuang decoction accompanied with suppressed expression of TGF-ß1 and phosphorylated NF-κB/P65 (p-P65) as indicated by immunohistochemistry. ELISA analysis demonstrated reduced serum levels of proinflammatory cytokines TNFα and IL-6. Most importantly, intestinal microbiota analysis by 16s rRNA-seq showed that Mahuang decoction restored the impaired richness and diversity of intestinal microflora and recovered the disrupted microbial community through reducing the abundance of deleterious microbes and promoting the expansion of beneficial microbes in CKD rats. Collectively, our findings demonstrated that Mahuang decoction mitigated kidney functional and structural impairment in CKD rats which were associated with the restoration of dysbiosis of intestinal microbiota, implying its potential in clinical CKD treatment.

10.
J Cell Mol Med ; 25(10): 4860-4869, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33733577

RESUMO

Diabetic cardiomyopathy (DCM) is a common diabetic complication characterized by diastolic relaxation abnormalities, myocardial fibrosis and chronic heart failure. Although TGF-ß/Smad3 signalling has been shown to play a critical role in chronic heart disease, the role and mechanisms of Smad3 in DCM remain unclear. We reported here the potential role of Smad3 in the development of DCM by genetically deleting the Smad3 gene from db/db mice. At the age of 32 weeks, Smad3WT-db/db mice developed moderate to severe DCM as demonstrated by a marked increase in the left ventricular (LV) mass, a significant fall in the LV ejection fraction (EF) and LV fractional shortening (FS), and progressive myocardial fibrosis and inflammation. In contrast, db/db mice lacking Smad3 (Smad3KO-db/db) were protected against the development of DCM with normal cardiac function and undetectable myocardial inflammation and fibrosis. Interestingly, db/db mice with deleting one copy of Smad3 (Smad3 ± db/db) did not show any cardioprotective effects. Mechanistically, we found that deletion of Smad3 from db/db mice largely protected cardiac Smad7 from Smurf2-mediated ubiquitin proteasome degradation, thereby inducing IBα to suppress NF-kB-driven cardiac inflammation. In addition, deletion of Smad3 also altered Smad3-dependent miRNAs by up-regulating cardiac miR-29b while suppressing miR-21 to exhibit the cardioprotective effect on Smad3KO-db/db mice. In conclusion, results from this study reveal that Smad3 is a key mediator in the pathogenesis of DCM. Targeting Smad3 may be a novel therapy for DCM.


Assuntos
Diabetes Mellitus Experimental/complicações , Cardiomiopatias Diabéticas/prevenção & controle , Fibrose/prevenção & controle , Inflamação/prevenção & controle , Proteína Smad3/fisiologia , Animais , Cardiomiopatias Diabéticas/etiologia , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/patologia , Fibrose/etiologia , Fibrose/metabolismo , Fibrose/patologia , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Fator de Crescimento Transformador beta
11.
Theranostics ; 11(6): 2845-2859, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33456576

RESUMO

Rationale: Transforming Growth Factor-beta (TGF-ß) /Smad3 signaling has been shown to play important roles in fibrotic and inflammatory diseases, but its role in beta cell function and type 2 diabetes is unknown. Methods: The role of Smad3 in beta cell function under type 2 diabetes condition was investigated by genetically deleting Smad3 from db/db mice. Phenotypic changes of pancreatic islets and beta cell function were compared between Smad3 knockout db/db (Smad3KO-db/db) mice and Smad3 wild-type db/db (Smad3WT-db/db) mice, and other littermate controls. Islet-specific RNA-sequencing was performed to identify Smad3-dependent differentially expressed genes associated with type 2 diabetes. In vitro beta cell proliferation assay and insulin secretion assay were carried out to validate the mechanism by which Smad3 regulates beta cell proliferation and function. Results: The results showed that Smad3 deficiency completely protected against diabetes-associated beta cell loss and dysfunction in db/db mice. By islet-specific RNA-sequencing, we identified 8160 Smad3-dependent differentially expressed genes associated with type 2 diabetes, where Smad3 deficiency markedly prevented the down-regulation of those genes. Mechanistically, Smad3 deficiency preserved the expression of beta cell development mediator Pax6 in islet, thereby enhancing beta cell proliferation and function in db/db mice in vivo and in Min6 cells in vitro. Conclusions: Taken together, we discovered a pathogenic role of Smad3 in beta cell loss and dysfunction via targeting the protective Pax6. Thus, Smad3 may represent as a novel therapeutic target for type 2 diabetes prevention and treatment.


Assuntos
Proliferação de Células/fisiologia , Diabetes Mellitus Experimental/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Fator de Transcrição PAX6/metabolismo , Proteína Smad3/metabolismo , Animais , Diabetes Mellitus Tipo 2/metabolismo , Nefropatias Diabéticas/metabolismo , Regulação para Baixo/fisiologia , Feminino , Glucose/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo
12.
J Asian Nat Prod Res ; 23(6): 584-595, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33233968

RESUMO

Decursin, a coumarin compound isolated from Angelica gigas has been shown to possess multiple anti-tumor activities. But it's still little known about the effects associated with cervical cancer. To explore the anti-tumor role of decursin and gain insights into its underlying mechanisms, we analyzed proliferation in parallel with apoptosis and migration in HeLa cells. Our findings implied that decursin can provoke apoptosis, and inhibit cell proliferation, migration in HeLa cells. More importantly, decursin also inhibited the tumor growth in vivo. The mechanisms may be associated with the regulation of Akt activation, with implications for novel therapeutic strategies on cervical cancer.[Formula: see text].


Assuntos
Benzopiranos , Butiratos , Transdução de Sinais , Neoplasias do Colo do Útero , Apoptose , Benzopiranos/farmacologia , Butiratos/farmacologia , Proliferação de Células , Feminino , Células HeLa , Humanos , Estrutura Molecular , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt
13.
Phytomedicine ; 52: 284-294, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30599909

RESUMO

BACKGROUND: Acute kidney injury (AKI) is a common kidney disease with a high risk of death and can develop into chronic kidney disease (CKD) and renal failure eventually. Curcumin, an herbal supplement, has been reported exhibiting a renoprotective role in AKI. However, the underlying mechanism is largely unclear. PURPOSE: Recent research showed that Mincle (Macrophage-inducible C-type lectin) maintained M1 macrophage polarization, which plays a key role in kidney injury of AKI through up-regulating the expression and secretion of inflammatory cytokines. Here, we investigated the effects of Curcumin on Mincle expression and macrophage polarization in vitro using lipopolysaccharide (LPS) induced macrophage inflammatory cell model and in vivo using a cisplatin induced murine AKI (cis-AKI) model. METHODS: Cell activation, inflammatory cytokines expression and secretion, protein levels, macrophage polarization and renal pathology were analyzed. RESULTS: Our results showed that Curcumin markedly reduced the mRNA expression and secretion of IL-1ß, IL-6, TNFα and MCP-1 in LPS stimulated RAW264.7 cell and the supernatant. The same results were found in Curcumin treated cis-AKI kidney and blood. The data also demonstrated that Curcumin remarkably down-regulated mRNA expression and protein level of Mincle in cis-AKI kidney and also reduced expression of iNOS (M1 macrophage marker) as well as inhibited the activation of Syk and NF-kB. Interestingly, although Mincle deletion in RAW264.7 cell largely decreased the LPS-induced protein level of iNOS, Curcumin cannot further reduce expression of iNOS without Mincle, indicating that Curcumin inhibits M1 macrophage with a Mincle-dependent pattern. Furthermore, flow cytometry results showed that Curcumin significantly decreased the iNOS positive macrophages and increased the CD206 (M2 macrophage marker) positive macrophages in vivo and in vitro. CONCLUSION: Our findings prove that Curcumin protects kidney from cisplatin induced AKI through inhibiting Mincle maintained M1 macrophage phenotype, that may provide a specific renoprotection mechanism for Curcumin to develop it as a new therapeutic candidate for AKI.


Assuntos
Cisplatino/efeitos adversos , Curcumina/farmacologia , Rim/efeitos dos fármacos , Lectinas Tipo C/metabolismo , Macrófagos/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Nefrite/tratamento farmacológico , Injúria Renal Aguda/patologia , Animais , Citocinas/metabolismo , Regulação para Baixo , Rim/patologia , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Fenótipo , Células RAW 264.7 , Regulação para Cima
14.
Ren Fail ; 38(9): 1398-1404, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27764998

RESUMO

Increasing evidences have shown that Helicobacter pylori (Hp) is a pathogen closely related to extra-gastric disorders. Our previous in vitro studies had demonstrated that Hp infection, at least via cytotoxin-associated gene A protein (CagA), might play an important role in the pathogenesis of IgA nephropathy (IgAN) by stimulating proliferation and ectopic synthesis of aberrantly glycosylated IgA1 of B cells. However, the relevant clinical evidence of IgAN resulted from Hp infection remain to be elucidated. This study aimed to investigate the risk incidence of IgAN caused by Hp infection. 22 primary IgAN, 20 non-IgA nephropathy (n-IgAN), and 30 healthy controls were included in this study. We found that the rate of IgG anti-Hp seropositivity was significantly improved in IgAN, but the current Hp infection was similar in all groups. The production and underglycosylation of IgA1 tended to increase in IgAN patients with IgG anti-Hp seropositivity. A tendency toward increased the risk of clinical prognosis was seen in IgAN with Hp infection. Hp antigen and CagA were only deposited in renal tubules, and enhanced antigen deposition in response to Hp was observed in IgAN. Our study suggested that Hp infection might have a pathogenic role in IgAN through giving rise to strongly mucosal immune response, and based on damage of renal tubular.


Assuntos
Glomerulonefrite por IGA/etiologia , Infecções por Helicobacter/complicações , Helicobacter pylori/isolamento & purificação , Túbulos Renais/patologia , Adulto , Anticorpos Antibacterianos/análise , Antígenos de Bactérias/análise , Biópsia , Ensaio de Imunoadsorção Enzimática , Feminino , Glomerulonefrite por IGA/diagnóstico , Glomerulonefrite por IGA/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/imunologia , Humanos , Imunoglobulina A/imunologia , Imunoglobulina A/metabolismo , Imuno-Histoquímica , Túbulos Renais/metabolismo , Masculino , Pessoa de Meia-Idade
15.
Ren Fail ; 38(3): 458-64, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26837331

RESUMO

Cytotoxin-associated antigen A (CagA), a major virulence factor of Helicobacter pylori (Hp), is associated with the pathogenesis of peptic ulcer and gastric cancer. Recent researches demonstrated that Hp exists in palatine tonsil in all studied IgA nephropathy (IgAN) patients, most of which were CagA-positive, suggesting that CagA may be a causative pathogenic factor of IgAN. However, the underlying molecular mechanisms and signaling pathway are still largely unclear. In the present study, CCK8 assay, enzyme-linked immunosorbent assay, and immunohistochemistry were performed to investigate the effect of CagA on cell proliferation and extracellular matrix secretion in rat glomerular mesangial cells. RT-PCR and western blotting were used to reveal the potential signaling pathway. Rat glomerular mesangial cells were treated with recombinant CagA protein for 72 h, in a dose- and time-dependent manner. We found that CagA promoted cell proliferation and extracellular matrix secretion by inhibiting signaling pathway of apoptosis. Taken together, these findings suggested that CagA induced cellular injury in glomerular mesangium by proliferation and secretion of extracellular matrix, and may play an important role in pathogenesis of IgAN.


Assuntos
Antígenos de Bactérias/farmacologia , Apoptose , Proteínas de Bactérias/farmacologia , Proliferação de Células , Matriz Extracelular/metabolismo , Mesângio Glomerular/citologia , Transdução de Sinais , Animais , Técnicas de Cultura de Células , Linhagem Celular , Glomerulonefrite por IGA/fisiopatologia , Ratos
16.
Int J Mol Med ; 36(2): 518-26, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26063203

RESUMO

Aldosterone is a steroid hormone secreted from the adrenal cortex, which regulates blood pressure. Higher concentrations of aldosterone can cause several diseases, including hypertension, diabetic nephropathy and chronic kidney disease. Previous reports have demonstrated that aldosterone has a pathogenic role in renal injury via reactive oxygen species (ROS), which involves the regulation of autophagy. However, whether aldosterone can induce autophagy in renal tubular cells remains to be elucidated. In the present study, elevated autophagy was observed in rat renal tubular NRK-52E cells exposed to aldosterone, which was demonstrated by the increased number of autophagosomes, conversion of LC3-I to LC3-II and the expression of Beclin-1. The enhanced autophagy was accompanied by increased production of intracellular ROS, which was reversed by N-acetylcysteine, a specific inhibitor of ROS signaling. Furthermore, treatment with ginsenoside Rg1 reduced the aldosterone-induced autophagy and production of ROS, possibly through reducing the phosphorylation of AMPK and preserving mTOR activity. These findings demonstrated that aldosterone promoted ROS generation and increased autophagy in the NRK-52E cells. Ginsenoside Rg1 effectively relieved aldosterone-induced oxidative stress and abnormal autophagy, suggesting that Rg1 may be used as a potential therapeutic drug to inhibit the renal injury, which is induced by aldosterone.


Assuntos
Aldosterona/metabolismo , Antioxidantes/farmacologia , Autofagia/efeitos dos fármacos , Ginsenosídeos/farmacologia , Túbulos Renais/citologia , Túbulos Renais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Medicamentos de Ervas Chinesas/farmacologia , Túbulos Renais/metabolismo , Túbulos Renais/patologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo
17.
ScientificWorldJournal ; 2014: 682189, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25133251

RESUMO

BACKGROUND: Ex vivo culture of intact embryonic kidney has become a powerful system for studying renal development. However, few methods have been available for gene manipulation and have impeded the identification and investigation of genes in this developmental process. RESULTS: Here we systemically compared eight different serotypes of pseudotyped self-complementary adenoassociated viruses (scAAVs) transduction in cultured embryonic kidney with a modified culture procedure. We demonstrated that scAAV was highly effective in delivering genes into and expressing in compacted tissues. scAAV serotypes 2 and 8 exhibited higher efficiency of transduction compared to others. Expression kinetics assay revealed that scAAV can be used for gene manipulation at the study of UB branching and nephrogenesis. Repressing WT1 in cultured kidney using shRNA impairs tubule formation. We for the first time employed and validated scAAV as a gene delivery tool in cultured kidney. CONCLUSIONS: These findings are expected to expedite the use of the ex vivo embryonic kidney cultures for kidney development research. For other ex vivo cultured organ models, scAAV could also be a promising tool for organogenesis study.


Assuntos
Dependovirus/genética , Rim/metabolismo , Transdução Genética/métodos , Animais , Células HEK293 , Humanos , Rim/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Cultura de Órgãos/métodos
18.
Proteomics ; 12(15-16): 2556-70, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22718539

RESUMO

Autosomal recessive polycystic kidney disease (ARPKD), characterized by ectatic collecting duct, is an infantile form of PKD occurring in 1 in 20 000 births. Despite having been studied for many years, little is known about the underlying mechanisms. In the current study, we employed, for the first time, a MS-based comparative proteomics approach to investigate the differently expressed proteins between kidney tissue samples of four ARPKD and five control individuals. Thirty two differently expressed proteins were identified and six of the identified protein encoding genes performed on an independent group (three ARPKD subjects, four control subjects) were verified by semi-quantitative RT-PCR, and part of them were further validated by Western blot and immunohistochemistry. Moreover, similar alteration tendency was detected after downregulation of PKHD1 by small interfering RNA in HEK293T cell. Interestingly, most of the identified proteins are associated with mitochondria. This implies that mitochondria may be implicated in ARPKD. Furthermore, the String software was utilized to investigate the biological association network, which is based on known and predicted protein interactions. In conclusion, our findings depicted a global understanding of ARPKD progression and provided a promising resource of targeting protein, and shed some light further investigation of ARPKD.


Assuntos
Proteínas Mitocondriais/metabolismo , Rim Policístico Autossômico Recessivo/metabolismo , Proteômica/métodos , Western Blotting , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Redes Reguladoras de Genes/genética , Células HEK293 , Humanos , Imuno-Histoquímica , Lactente , Masculino , Proteínas Mitocondriais/química , Rim Policístico Autossômico Recessivo/genética , RNA Interferente Pequeno/metabolismo , Receptores de Superfície Celular/metabolismo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Espectrometria de Massas por Ionização por Electrospray
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...