Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
1.
Am J Physiol Renal Physiol ; 319(4): F571-F578, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32830537

RESUMO

(Pro)renin receptor [(P)RR] has multiple functions, but its regulation and role in the pathogenesis in glomerulonephritis (GN) are poorly defined. The aims of the present study were to determine the effects of direct renin inhibition (DRI) and demonstrate the role of (P)RR on the progression of crescentic GN. The anti-glomerular basement membrane nephritis rat model developed progressive proteinuria (83.64 ± 10.49 mg/day) and glomerular crescent formation (percent glomerular crescent: 62.1 ± 2.3%) accompanied by increased macrophage infiltration and glomerular expression of monocyte chemoattractant protein (MCP)-1, (P)RR, phospho-extracellular signal-regulated kinase (ERK)1/2, Wnt4, and active ß-catenin. Treatment with DRI ameliorated proteinuria (20.33 ± 5.88 mg/day) and markedly reduced glomerular crescent formation (20.9 ± 2.6%), induction of macrophage infiltration, (P)RR, phospho-ERK1/2, Wnt4, and active ß-catenin. Furthermore, primary cultured parietal epithelial cells stimulated by recombinant prorenin showed significant increases in cell proliferation. Notably, while the ERK1/2 inhibitor PD98059 or (P)RR-specific siRNA treatment abolished the elevation in cell proliferation, DRI treatment did not abrogate this elevation. Moreover, cultured mesangial cells showed an increase in prorenin-induced MCP-1 expression. Interestingly, (P)RR or Wnt4-specific siRNA treatment or the ß-catenin antagonist XAV939 inhibited the elevation of MCP-1 expression, whereas DRI did not. These results suggest that (P)RR regulates glomerular crescent formation via the ERK1/2 signaling and Wnt/ß-catenin pathways during the course of anti-glomerular basement membrane nephritis and that DRI mitigates the progression of crescentic GN through the reduction of (P)RR expression but not inhibition of prorenin binding to (P)RR.


Assuntos
Proliferação de Células , Glomerulonefrite/enzimologia , Células Mesangiais/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores de Superfície Celular/metabolismo , Via de Sinalização Wnt , Amidas/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Fumaratos/farmacologia , Glomerulonefrite/patologia , Glomerulonefrite/prevenção & controle , Masculino , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/patologia , Fosforilação , Ratos Endogâmicos WKY , ATPases Vacuolares Próton-Translocadoras , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt4/metabolismo , beta Catenina/metabolismo
2.
Med Sci Monit ; 26: e919399, 2020 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-32012145

RESUMO

BACKGROUND The aim of this study was to explore the effects of NADPH oxidase 5 (NOX5) in high glucose-stimulated human glomerular mesangial cells (HMCs). MATERIAL AND METHODS Cells were cultured under normal glucose (NG) or high glucose (HG) conditions. Then, NOX5 siRNA was transfected into HG-treated HMCs. A Cell Counting Kit-8 assay, colony formation assay and 5-ethynyl-20-deoxyuridine (EDU) incorporation assay were applied to measure cell proliferative ability. In addition, the levels of oxidative stress factors including reactive oxygen species (ROS), malonaldehyde (MDA), NADPH, superoxide dismutase (SOD), and glutathione peroxidase (GSH-PX), inflammatory cytokines including tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-6, IL-1ß, and monocyte chemoattractant protein-1 (MCP-1) in HMCs were detected by kits. Moreover, the expression of TLR4/NF-kappaB signaling and extracellular matrix (ECM) associated genes were evaluated by western blotting. RESULTS The results revealed that the NOX5 was overexpressed in HG-treated HMCs. Silencing of NOX5 decreased proliferation of HMCs induced by HG. And NOX5 silencing alleviated the production of MDA and NADPH accompanied by an increase of SOD and GSH-PX levels. Additionally, the contents of TNF-alpha, IL-6, IL-1ß, and MCP-1 were reduced after transfection with NOX5 siRNA. Furthermore, silencing of NOX5 deceased the expression of collagen I, collagen IV, TGF-ß1, and fibronectin induced by HG stimulation. TLR4, MyD88, and phospho-NF-kappaB p65 expression were downregulated notably in NOX5 silencing group. CONCLUSIONS Taken together, these findings demonstrated that inhibition of NOX5 attenuated HG-induced HMCs oxidative stress, inflammation, and ECM accumulation, suggesting that NOX5 may serve as a potential therapeutic target for diabetic nephropathy (DN) treatment.


Assuntos
Matriz Extracelular/metabolismo , Glucose/toxicidade , Inflamação/patologia , Células Mesangiais/enzimologia , Células Mesangiais/patologia , NADPH Oxidase 5/antagonistas & inibidores , Estresse Oxidativo , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Matriz Extracelular/efeitos dos fármacos , Inativação Gênica , Glutationa Peroxidase/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Malondialdeído/metabolismo , Células Mesangiais/efeitos dos fármacos , NADP/metabolismo , NADPH Oxidase 5/genética , NADPH Oxidase 5/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
3.
Cell Physiol Biochem ; 47(6): 2522-2533, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29991026

RESUMO

BACKGROUND/AIMS: Sphingosine 1-phosphate (S1P) is considered as a key molecule regulating various cell functions including cell growth and death. It is produced by two sphingosine kinases (SK) denoted as SK-1 and SK-2. Whereas SK-1 has been extensively studied and has been appointed a role in promoting cell growth, the function of SK-2 is controversial, and both pro-proliferative and pro-apoptotic functions have been suggested. In this study we investigated whether renal mesangial cells isolated from transgenic mice overexpressing the human Sphk2 gene (hSK2-tg) showed an altered cell response towards growth-inducing and apoptotic stimuli. METHODS: hSK2-tg mice were generated by using a Quick KnockinR strategy. Renal mesangial cells were isolated by a differential sieving method and further cultivated in vitro. Lipids were quantified by mass spectrometry. Protein expression was determined by Western blot analysis, cell proliferation was determined by 3H-thymidine incorporation, and apoptosis was determined by a DNA fragmentation ELISA. RESULTS: We show here that kidneys and mesangial cells from hSK2-tg mice express the hSK2 as well as the endogenous mouse mSK2. hSK2 and mSK2 predominantly resided in the cytosol of quiescent transgenic cells. However, S1P accumulated strongly in the nucleus and only minimally in the cytosol of transgenic cells. Functionally, hSK2-tg cells proliferated less than control cells under normal growth conditions and were also more sensitive towards stress-induced apoptosis. On the molecular level, this was reflected by reduced ERK and Akt/PKB activation, and upon staurosporine treatment, by a sensitized mitochondrial pathway as manifested by reduced anti-apoptotic Bcl-XL expression and increased cleavage of caspase-9, downstream caspase-3 and PARP-1. CONCLUSION: Altogether, these data demonstrate that SK-2 exerts an antiproliferative and apoptosis-sensitizing effect in renal mesangial cells which suggests that selective inhibitors of SK-2 may promote proliferation and reduce apoptosis and this may have impact on the outcome of proliferation-associated diseases such as mesangioproliferative glomerulonephritis.


Assuntos
Apoptose , Proliferação de Células , Sistema de Sinalização das MAP Quinases , Células Mesangiais/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Humanos , Células Mesangiais/citologia , Camundongos , Camundongos Transgênicos , Fosfotransferases (Aceptor do Grupo Álcool)/genética
4.
Biomed Pharmacother ; 102: 1077-1083, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29710524

RESUMO

Tangeretin (5, 6, 7, 8, 4'-pentamethoxyflavone), a natural compound extracted from citrus plants, has been shown to possess a variety of pharmacological activities, including anti-oxidant, anti-tumor, cytostatic and anti-diabetic properties. However, the role of tangeretin in diabetic nephropathy (DN) has not yet been investigated. This study was undertaken to elucidate the effects of tangeretin on high glucose (HG)-induced oxidative stress and extracellular matrix (ECM) accumulation in human glomerular mesangial cells (MCs) and explore the underlying mechanisms. Our results showed that tangeretin significantly inhibited HG-induced the proliferation of MCs. In addition, tangeretin dramatically reduced the levels of reactive oxygen species (ROS) and malondialdhyde (MDA), and induced SOD activity, as well as inhibited the expression of fibronectin (FN) and collagen IV in HG-stimulated MCs. Furthermore, tangeretin efficiently prevented the activation of ERK signaling pathway in HG-stimulated MCs. Taken together, these data indicated that tangeretin inhibits HG-induced cell proliferation, oxidative stress and ECM expression in glomerular MCs, at least in part, through the inactivation of ERK signaling pathway. Therefore, tangeretin may be a potential agent in the treatment of DN.


Assuntos
Matriz Extracelular/metabolismo , Flavonas/farmacologia , Glucose/toxicidade , Células Mesangiais/patologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo IV/metabolismo , Ativação Enzimática/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Fibronectinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Malondialdeído/metabolismo , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/enzimologia , Células Mesangiais/metabolismo , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
5.
Am J Physiol Renal Physiol ; 315(2): F336-F344, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29667914

RESUMO

Transforming growth factor-ß (TGFß) plays a central role in renal scarring, controlling extracellular matrix deposition by interstitial cells and mesangial cells. TGFß signals through Smad and mitogen-activated protein kinase (MAPK) pathways. To understand the role of MAPK in interstitial and mesangial cells, we genetically inactivated TGFß-activated kinase-1 ( Map3k7) using Foxd1+/cre. Embryonic kidney development was unperturbed in mutants, but spontaneous scarring of the kidney ensued during the first postnatal week, with retention of embryonic nephrogenic rests and accumulation of collagen IV in the mesangium. MAPK signaling in the mesangium of mutant mice was skewed, with depressed p38 but elevated c-Jun NH2-terminal kinase (JNK) activation at postnatal day 3. Despite normal expression of platelet-derived growth factor receptor-ß (PDGFRß) in the mesangium of mutants at birth, expression was lost concomitantly with the increase in JNK activation, and studies in isolated mesangial cells revealed that JNK negatively regulates Pdgfrß. In summary, we show that MAP3K7 balances MAPK signaling in mesangial cells, suppressing postnatal JNK activation. We propose that the balance of MAPK signaling is essential for appropriate postnatal regulation of mesangial PDGFRß expression.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Glomerulonefrite/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Células Mesangiais/enzimologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Células Cultivadas , Colágeno Tipo IV/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Fibrose , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Inativação Gênica , Predisposição Genética para Doença , Glomerulonefrite/genética , Glomerulonefrite/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases/deficiência , MAP Quinase Quinase Quinases/genética , Células Mesangiais/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Med Sci Monit ; 24: 846-854, 2018 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-29428962

RESUMO

BACKGROUND To investigate the protective effect of ursolic acid (UA) on high glucose (HG)-induced human glomerular mesangial cell injury and to determine whether UA inhibits cell proliferation and reactive oxygen species (ROS) production by suppressing PI3K/Akt/mTOR pathway activation. MATERIAL AND METHODS Human mesangial cells were cultured with normal glucose (NG group), high glucose (HG group), mannitol (mannitol hypertonic control group), or high glucose with different concentrations (0.5, 1.0, and 2.0 mmol/L) of UA (HG+UA groups). Cell proliferation and intracellular ROS levels were assessed by methyl thiazolyl tetrazolium (MTT) and dichloro-dihydro-fluorescein diacetate (DCFH-DA) flow cytometry assays, respectively. Western blotting was used to detect mesangial cell expression of PI3K/Akt/mTOR pathway components, including Akt, p-Akt, mTOR, and p-mTOR, and proteins related to cell injury, including TGF-ß1 and fibronectin (FN). mRNA expression of TGF-ß1 and FN were evaluated using real-time quantitative polymerase chain reaction (PCR). RESULTS Abnormal proliferation was observed in human glomerular mesangial cells at 48 h after treatment with HG, and UA suppressed the HG-induced proliferation of mesangial cells in a dose-dependent manner. UA inhibited ROS generation and oxidative stress in mesangial cells and mitigated mesangial cell injury. Treatment with UA reduced Akt and mTOR phosphorylation levels in mesangial cells exposed to HG (p<0.05 vs. HG) and downregulated protein and mRNA expression of TGF-ß1 and FN in these cells (p<0.05 vs. HG). CONCLUSIONS UA attenuated mesangial cell proliferation and ROS generation by inhibiting HG-mediated PI3K/Akt/mTOR pathway activation, thereby ameliorating mesangial cell damage.


Assuntos
Glucose/toxicidade , Células Mesangiais/enzimologia , Células Mesangiais/patologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Triterpenos/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Fibronectinas/genética , Fibronectinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células Mesangiais/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Ácido Ursólico
7.
Am J Physiol Renal Physiol ; 314(4): F630-F642, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357434

RESUMO

The development of nephritis is a leading cause of morbidity and mortality in lupus patients. Although the general pathophysiological progression of lupus nephritis is known, the molecular mediators and mechanisms are incompletely understood. Previously, we demonstrated that the glycosphingolipid (GSL) catabolic pathway is elevated in the kidneys of MRL/lpr lupus mice and human lupus patients with nephritis. Specifically, the activity of neuraminidase (NEU) and expression of Neu1, an enzyme in the GSL catabolic pathway is significantly increased. To better understand the role and mechanisms by which this pathway contributes to the progression of LN, we analyzed the expression and effects of NEU activity on the function of MRL/lpr lupus-prone mesangial cells (MCs). We demonstrate that NEU1 and NEU3 promote IL-6 production in MES13 MCs. Neu1 expression, NEU activity, and IL-6 production are significantly increased in stimulated primary MRL/lpr lupus-prone MCs, and blocking NEU activity inhibits IL-6 production. NEU1 and NEU3 expression overlaps IgG deposits in MCs in vitro and in renal sections from nephritic MRL/lpr mice. Together, our results suggest that NEU activity mediates IL-6 production in lupus-prone MCs possibly through an IgG-receptor complex signaling pathway.


Assuntos
Interleucina-6/metabolismo , Nefrite Lúpica/enzimologia , Células Mesangiais/enzimologia , Neuraminidase/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Modelos Animais de Doenças , Feminino , Inibidores de Glicosídeo Hidrolases/farmacologia , Imunoglobulina G/farmacologia , Nefrite Lúpica/sangue , Nefrite Lúpica/tratamento farmacológico , Nefrite Lúpica/patologia , Masculino , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/patologia , Camundongos Endogâmicos MRL lpr , Neuraminidase/antagonistas & inibidores , Neuraminidase/genética , Receptores de IgG/metabolismo , Transdução de Sinais , Regulação para Cima
8.
Am J Physiol Cell Physiol ; 314(2): C242-C253, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29141920

RESUMO

Injury of mesangial cells (MC) is a prominent feature of glomerulonephritis. Activated MC secrete inflammatory mediators that induce cell apoptosis. Parathyroid hormone-related peptide (PTHrP) is a locally active cytokine that enhances cell survival and is upregulated by proinflammatory factors in many cell types. The aim of this study was to analyze the regulation of PTHrP expression by inflammatory cytokines and to evaluate whether PTHrP itself acts as a proinflammatory and/or survival factor on male murine MC in primary culture. Our results showed that IL-1ß (10 ng/ml) and TNF-α (10 ng/ml) rapidly and transiently upregulated PTHrP expression in MC. The effects of IL-1ß were both transcriptional and posttranscriptional, with stabilization of the PTHrP mRNA by human antigen R (HuR). Proteome profiler arrays showed that PTHrP itself enhanced cytokines within 2 h in cell lysates, mainly IL-17, IL-16, IL-1α, and IL-6. PTHrP also stimulated sustained expression (2-4 h) of chemokines, mainly regulated upon activation normal T cell expressed and secreted (RANTES)/C-C motif chemokine 5 (CCL5) and macrophage inflammatory protein-2 (MIP-2)/C-X-C motif chemokine 2 (CXCL2), thymus and activation-regulated chemokine (TARC)/CCL17, and interferon-inducible T cell α-chemoattractant (I-TAC)/CXCL11. Moreover, PTHrP markedly enhanced cyclooxygenase-2 (COX-2) expression and elicited its autoinduction through the activation of the NF-κB pathway. PTHrP induced MC survival via the COX-2 products, and PTHrP overexpression in MC blunted the apoptotic effects of IL-1ß and TNF-α. Altogether, these findings suggest that PTHrP functions as a booster of glomerular inflammatory processes and may be a negative feedback loop preserving MC survival.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Glomerulonefrite/enzimologia , Mediadores da Inflamação/metabolismo , Interleucina-1beta/farmacologia , Células Mesangiais/efeitos dos fármacos , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células Cultivadas , Glomerulonefrite/genética , Glomerulonefrite/patologia , Masculino , Células Mesangiais/enzimologia , Células Mesangiais/patologia , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo/genética , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Regulação para Cima
9.
Artigo em Inglês | MEDLINE | ID: mdl-29154978

RESUMO

7-Ketocholesterol (7-KCHO) is a highly proinflammatory oxysterol and plays an important role in the pathophysiology of diabetic nephropathy (DN). Lipoxygenases (LOXs) and cyclooxygenases (COXs) are also involved in the development of DN. The aim of this study was to clarify the effects of 7-KCHO on mRNA expression of LOXs and COXs as well as pro-inflammatory cytokines in human mesangial cells (HMC). We evaluated cell viability by WST-8 assay and measured mRNA expression by reverse transcription-polymerase chain reaction. Intracellular reactive oxygen species (ROS) production was evaluated by flow cytometry. Although 7-KCHO did not affect cell viability of HMC, 7-KCHO stimulated significant increases in mRNA expression of 12-LOX, COX-2 and pro-inflammatory cytokines. 7-KCHO also induced an increase in ROS production, while N-acetylcysteine partially suppressed the increase. The 12-LOX and COX-2 inhibitors also suppressed mRNA expression of cytokines. These findings may contribute to the elucidation of the molecular mechanism of the pathophysiology of DN.


Assuntos
Nefropatias Diabéticas/patologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Cetocolesteróis/farmacologia , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/enzimologia , Espécies Reativas de Oxigênio/metabolismo , Araquidonato 12-Lipoxigenase/genética , Araquidonato 12-Lipoxigenase/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Inibidores Enzimáticos/farmacologia , Humanos , Interleucina-1beta/genética , Interleucina-6/genética , Células Mesangiais/metabolismo , Células Mesangiais/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
10.
J Am Soc Nephrol ; 28(10): 2879-2885, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28701517

RESUMO

Human glomerular diseases can be caused by several different diseases, many of which include mesangial expansion and/or proliferation followed by glomerulosclerosis. However, molecular mechanisms underlying the pathologic mesangial changes remain poorly understood. Here, we investigated the role of the mammalian target of rapamycin complex 1 (mTORC1)-S6 kinase pathway in mesangial expansion and/or proliferation by ablating an upstream negative regulator, tuberous sclerosis complex 1 (TSC1), using tamoxifen-induced Foxd1-Cre mice [Foxd1ER(+) TSC1 mice]. Foxd1ER(+) TSC1 mice showed mesangial expansion with increased production of collagen IV, collagen I, and α-smooth muscle actin in glomeruli, but did not exhibit significant mesangial proliferation or albuminuria. Furthermore, rapamycin treatment of Foxd1ER(+) TSC1 mice suppressed mesangial expansion. Among biopsy specimens from patients with glomerular diseases, analysis of phosphorylated ribosomal protein S6 revealed mesangial cell mTORC1 activation in IgA nephropathy and in lupus mesangial proliferative nephritis but not in the early phase of diabetic nephropathy. In summary, mesangial cell mTORC1 activation can cause mesangial expansion and has clinical relevance for human glomerular diseases. This report also confirms that the tamoxifen-induced mesangium-specific Cre-loxP system is useful for studies designed to clarify the role of the mesangium in glomerular diseases in adults.


Assuntos
Nefropatias/enzimologia , Células Mesangiais/enzimologia , Complexos Multiproteicos/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Feminino , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Transgênicos
11.
Int J Mol Sci ; 18(7)2017 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-28671608

RESUMO

Lipotoxicity-induced mesangial cell apoptosis is implicated in the exacerbation of diabetic nephropathy (DN). Protein arginine methyltransferases (PRMTs) have been known to regulate a variety of biological functions. Recently, it was reported that PRMT1 expression is increased in proximal tubule cells under diabetic conditions. However, their roles in mesangial cells remain unexplored. Thus, we examined the pathophysiological roles of PRMTs in mesangial cell apoptosis. Treatment with palmitate, which mimics cellular lipotoxicity, induced mesangial cell apoptosis via protein kinase RNA-like endoplasmic reticulum kinase (PERK) and ATF6-mediated endoplasmic reticulum (ER) stress signaling. Palmitate treatment increased PRMT1 expression and activity in mesangial cells as well. Moreover, palmitate-induced ER stress activation and mesangial cell apoptosis was diminished by PRMT1 knockdown. In the mice study, high fat diet-induced glomerular apoptosis was attenuated in PRMT1 haploinsufficient mice. Together, these results provide evidence that lipotoxicity-induced PRMT1 expression promotes ER stress-mediated mesangial cell apoptosis. Strategies to regulate PRMT1 expression or activity could be used to prevent the exacerbation of DN.


Assuntos
Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Lipídeos/toxicidade , Células Mesangiais/enzimologia , Células Mesangiais/patologia , Proteína-Arginina N-Metiltransferases/metabolismo , Fator 6 Ativador da Transcrição/metabolismo , Animais , Dieta Hiperlipídica , Técnicas de Silenciamento de Genes , Haploinsuficiência/efeitos dos fármacos , Células Mesangiais/efeitos dos fármacos , Camundongos , Palmitatos/toxicidade , Ratos , Transdução de Sinais/efeitos dos fármacos , eIF-2 Quinase/metabolismo
12.
Am J Physiol Renal Physiol ; 313(2): F423-F429, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28490526

RESUMO

The goal of this study was to address the role of ATP-citrate lyase (ACL), an enzyme that converts citrate to acetyl-CoA, in high glucose (HG)-induced histone acetylation and profibrotic gene expression. Our recent ChIP-Seq studies have demonstrated that HG induces genome-wide histone hyperacetylation in mesangial cells (MCs). Here, we showed that exposure of MCs to HG markedly increased histone acetylation at the H3K9/14 and H3K18 marks and induced the expression of potent profibrotic factors TGF-ß1, TGF-ß3, and connective tissue growth factor (CTGF). The induction of these profibrotic factors was further enhanced by histone deacetylase inhibitor but suppressed by histone acetyl-transferase inhibitor, confirming the importance of histone acetylation in this regulation. Interestingly, HG not only upregulated ACL expression but also promoted ACL nuclear translocation, evidenced by increased ACL concentration and activity in the nuclear extracts. Consistent with this observation, transfection of MCs with a plasmid-carrying green fluorescent protein (GFP)-ACL fusion protein led to GFP nuclear accumulation when cultured in HG condition. Silencing ACL with siRNAs alleviated HG-induced histone hyperacetylation, as well as upregulation of TGF-ß1, TGF-ß3, CTGF, and extracellular matrix (ECM) proteins fibronectin and collagen type IV, whereas ACL overexpression further enhanced HG induction of histone acetylation, as well as these profibrotic factors and ECM proteins. Collectively, these observations demonstrate that HG promotes ACL expression and translocation into the nucleus, where ACL converts citrate to acetyl-CoA to provide the substrate for histone acetylation, leading to upregulation of fibrogenic genes. Therefore, ACL plays a critical role in epigenetic regulation of diabetic renal fibrosis.


Assuntos
ATP Citrato (pro-S)-Liase/metabolismo , Nefropatias Diabéticas/enzimologia , Epigênese Genética/efeitos dos fármacos , Glucose/toxicidade , Histonas/genética , Células Mesangiais/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , ATP Citrato (pro-S)-Liase/genética , Acetilcoenzima A/metabolismo , Acetilação , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular Transformada , Ácido Cítrico/metabolismo , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrose , Células Mesangiais/enzimologia , Células Mesangiais/patologia , Camundongos , Interferência de RNA , Fatores de Tempo , Transfecção , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta3/genética , Fator de Crescimento Transformador beta3/metabolismo , Regulação para Cima
13.
J Mol Endocrinol ; 59(1): R1-R10, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28420715

RESUMO

Diabetic nephropathy is one of the most frequent, devastating and costly complications of diabetes. The available therapeutic approaches are limited. Dipeptidyl peptidase type 4 (DPP-4) inhibitors represent a new class of glucose-lowering drugs that might also have reno-protective properties. DPP-4 exists in two forms: a plasma membrane-bound form and a soluble form, and can exert many biological actions mainly through its peptidase activity and interaction with extracellular matrix components. The kidneys have the highest DPP-4 expression level in mammalians. DPP-4 expression and urinary activity are up-regulated in diabetic nephropathy, highlighting its role as a potential target to manage diabetic nephropathy. Preclinical animal studies and some clinical data suggest that DPP-4 inhibitors decrease the progression of diabetic nephropathy in a blood pressure- and glucose-independent manner. Many studies reported that these reno-protective effects could be due to increased half-life of DPP-4 substrates such as glucagon-like peptide-1 (GLP-1) and stromal derived factor-1 alpha (SDF-1a). However, the underlying mechanisms are far from being completely understood and clearly need further investigations.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Dipeptidil Peptidase 4/genética , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Hipoglicemiantes/uso terapêutico , Células Mesangiais/efeitos dos fármacos , Podócitos/efeitos dos fármacos , Substâncias Protetoras/uso terapêutico , Animais , Quimiocina CXCL12/agonistas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Ensaios Clínicos como Assunto , Nefropatias Diabéticas/enzimologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Dipeptidil Peptidase 4/metabolismo , Avaliação Pré-Clínica de Medicamentos , Matriz Extracelular , Regulação da Expressão Gênica , Peptídeo 1 Semelhante ao Glucagon/agonistas , Peptídeo 1 Semelhante ao Glucagon/genética , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Células Mesangiais/enzimologia , Células Mesangiais/patologia , Podócitos/enzimologia , Podócitos/patologia
14.
J Clin Invest ; 127(3): 912-928, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28165339

RESUMO

Steroid-resistant nephrotic syndrome (SRNS) causes 15% of chronic kidney disease cases. A mutation in 1 of over 40 monogenic genes can be detected in approximately 30% of individuals with SRNS whose symptoms manifest before 25 years of age. However, in many patients, the genetic etiology remains unknown. Here, we have performed whole exome sequencing to identify recessive causes of SRNS. In 7 families with SRNS and facultative ichthyosis, adrenal insufficiency, immunodeficiency, and neurological defects, we identified 9 different recessive mutations in SGPL1, which encodes sphingosine-1-phosphate (S1P) lyase. All mutations resulted in reduced or absent SGPL1 protein and/or enzyme activity. Overexpression of cDNA representing SGPL1 mutations resulted in subcellular mislocalization of SGPL1. Furthermore, expression of WT human SGPL1 rescued growth of SGPL1-deficient dpl1Δ yeast strains, whereas expression of disease-associated variants did not. Immunofluorescence revealed SGPL1 expression in mouse podocytes and mesangial cells. Knockdown of Sgpl1 in rat mesangial cells inhibited cell migration, which was partially rescued by VPC23109, an S1P receptor antagonist. In Drosophila, Sply mutants, which lack SGPL1, displayed a phenotype reminiscent of nephrotic syndrome in nephrocytes. WT Sply, but not the disease-associated variants, rescued this phenotype. Together, these results indicate that SGPL1 mutations cause a syndromic form of SRNS.


Assuntos
Aldeído Liases , Movimento Celular/genética , Ictiose Lamelar , Células Mesangiais/enzimologia , Mutação , Síndrome Nefrótica , Aldeído Liases/genética , Aldeído Liases/metabolismo , Animais , Linhagem Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Feminino , Humanos , Ictiose Lamelar/enzimologia , Ictiose Lamelar/genética , Ictiose Lamelar/patologia , Masculino , Células Mesangiais/patologia , Camundongos , Camundongos Knockout , Síndrome Nefrótica/enzimologia , Síndrome Nefrótica/genética , Síndrome Nefrótica/patologia , Transporte Proteico/genética , Ratos
15.
Oncotarget ; 8(6): 10185-10198, 2017 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-28052039

RESUMO

Hyperuricemia is not only the main feature of gout but also a cause of gout-related organ injuries including glomerular hypertrophy and sclerosis. Uric acid (UA) has been proven to directly cause mesangial cell (MC) proliferation with elusive mechanisms. The present study was undertaken to examined the role of inflammatory cascade of COX-2/mPGES-1/PGE2 in UA-induced MC proliferation. In the dose- and time-dependent experiments, UA increased cell proliferation shown by the increased total cell number, DNA synthesis rate, and the number of cells in S and G2 phases in parallel with the upregulation of cyclin A2 and cyclin D1. Interestingly, UA-induced cell proliferation was accompanied with the upregulation of COX-2 and mPGES-1 at both mRNA and protein levels. Strikingly, inhibition of COX-2 via a specific COX-2 inhibitor NS-398 markedly blocked UA-induced MC proliferation. Meanwhile, UA-induced PGE2 production was almost entirely abolished. Furthermore, inhibiting mPGES-1 by a siRNA approach in MCs also ameliorated UA-induced MC proliferation in line with a significant blockade of PGE2 secretion. More importantly, in gout patients, we observed a significant elevation of urinary PGE2 excretion compared with healthy controls, indicating a translational potential of this study to the clinic. In conclusion, our findings indicated that COX-2/mPGES-1/PGE2 cascade activation mediated UA-induced MC proliferation. This study offered new insights into the understanding and the intervention of UA-related glomerular injury.


Assuntos
Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Gota/enzimologia , Hiperuricemia/enzimologia , Células Mesangiais/efeitos dos fármacos , Prostaglandina-E Sintases/metabolismo , Ácido Úrico/farmacologia , Idoso , Animais , Estudos de Casos e Controles , Linhagem Celular , Ciclina A2/metabolismo , Ciclina D1/metabolismo , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/urina , Relação Dose-Resposta a Droga , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Gota/genética , Gota/patologia , Gota/urina , Humanos , Hiperuricemia/genética , Hiperuricemia/patologia , Hiperuricemia/urina , Masculino , Células Mesangiais/enzimologia , Células Mesangiais/patologia , Camundongos , Pessoa de Meia-Idade , Prostaglandina-E Sintases/genética , Interferência de RNA , Pontos de Checagem da Fase S do Ciclo Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção
16.
Br J Nutr ; 116(9): 1491-1501, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27753426

RESUMO

Low-protein diet plus ketoacids (LPD+KA) has been reported to decrease proteinuria in patients with chronic kidney diseases (CKD). However, the mechanisms have not been clarified. As over-activation of intrarenal renin-angiotensin system (RAS) has been shown to play a key role in the progression of CKD, the current study was performed to investigate the direct effects of LPD+KA on intrarenal RAS, independently of renal haemodynamics. In this study, 3/4 subtotal renal ablated rats were fed 18 % normal-protein diet (Nx-NPD), 6 % low-protein diet (Nx-LPD) or 5 % low-protein diet plus 1 % ketoacids (Nx-LPD+KA) for 12 weeks. Sham-operated rats fed NPD served as controls. The level of proteinuria and expression of renin, angiotensin II (AngII) and its type 1 receptors (AT1R) in the renal cortex were markedly higher in Nx-NPD group than in the sham group. LPD+KA significantly decreased the proteinuria and inhibited intrarenal RAS activation. To exclude renal haemodynamic impact on intrarenal RAS, the serum samples derived from the different groups were added to the culture medium of mesangial cells. It showed that the serum from Nx-NPD directly induced higher expression of AngII, AT1R, fibronectin and transforming growth factor-ß1 in the mesangial cells than in the control group. Nx-LPD+KA serum significantly inhibited these abnormalities. Then, proteomics and biochemical detection suggested that the mechanisms underlying these beneficial effects of LPD+KA might be amelioration of the nutritional metabolic disorders and oxidative stress. In conclusion, LPD+KA could directly inhibit the intrarenal RAS activation, independently of renal haemodynamics, thus attenuating the proteinuria in CKD rats.


Assuntos
Dieta com Restrição de Proteínas , Suplementos Nutricionais , Modelos Animais de Doenças , Cetoácidos/uso terapêutico , Rim/metabolismo , Sistema Renina-Angiotensina , Uremia/dietoterapia , Angiotensina II/química , Angiotensina II/genética , Angiotensina II/metabolismo , Animais , Linhagem Celular , Regulação para Baixo , Regulação da Expressão Gênica , Resistência à Insulina , Rim/fisiopatologia , Masculino , Células Mesangiais/enzimologia , Células Mesangiais/metabolismo , Nefrectomia/efeitos adversos , Estresse Oxidativo , Proteinúria/etiologia , Proteinúria/prevenção & controle , Proteômica/métodos , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/química , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Renina/antagonistas & inibidores , Renina/genética , Renina/metabolismo , Uremia/etiologia , Uremia/metabolismo , Uremia/fisiopatologia
17.
Biomed Pharmacother ; 80: 338-342, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27133074

RESUMO

Glomerular mesangial cells (MCs) proliferation and extracellular matrix (ECM) accumulation have been recognized as major pathogenic events in the progression of diabetic nephropathy. Betulinic acid (BA), (3ß-hydroxy-lup-20(29)-en-28-oic acid), is a naturally occurring pentacyclic lupane group triterpenoid, and it has been shown to possess glucose-lowering property. However, the role of BA on MC proliferation and ECM accumulation in diabetic condition remains unclear. So, in the present study, we investigated the role of BA on cell proliferation and ECM accumulation in rat glomerular MCs cultured under high glucose (HG) condition. In the current study, we demonstrated that BA suppressed HG-induced MC proliferation, arrested HG-induced cell-cycle progression, reversed HG-inhibited expression of p21(Waf1/Cip1) and p27(Kip1). It also suppressed HG-induced fibronectin (FN) expression in MCs. Furthermore, BA inhibited HG-induced phosphorylation of ERK1/2 and p38MAPK in MCs. In conclusion, our present study demonstrated that BA inhibited HG-induced cell proliferation and FN expression in MCs via inhibiting ERK1/2 and p38MAPK pathways. Thus, BA may serve as a potential drug for the treatment of diabetic nephropathy.


Assuntos
Fibronectinas/metabolismo , Glucose/farmacologia , Células Mesangiais/citologia , Células Mesangiais/metabolismo , Triterpenos/farmacologia , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/enzimologia , Camundongos , Triterpenos Pentacíclicos , Fosforilação/efeitos dos fármacos , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Ácido Betulínico
18.
Bioorg Med Chem ; 24(13): 3029-3034, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27234891

RESUMO

The upregulation of PGE2 by mesangial cells has been observed under chronic inflammation condition. In the present work, renal mesangial cells were stimulated to trigger a huge increase of PGE2 synthesis and were treated in the absence or presence of known PLA2 inhibitors. A variety of synthetic inhibitors, mainly developed in our labs, which are known to selectively inhibit each of GIVA cPLA2, GVIA iPLA2, and GIIA/GV sPLA2, were used as tools in this study. Synthetic sPLA2 inhibitors, such as GK115 (an amide derivative based on the non-natural amino acid (R)-γ-norleucine) as well as GK126 and GK241 (2-oxoamides based on the natural (S)-α-amino acid leucine and valine, respectively) presented an interesting effect on the suppression of PGE2 formation.


Assuntos
Dinoprostona/metabolismo , Células Mesangiais/efeitos dos fármacos , Inibidores de Fosfolipase A2/farmacologia , Fosfolipases A2 Secretórias/antagonistas & inibidores , Animais , Células Cultivadas , Células Mesangiais/enzimologia , Modelos Biológicos , Estrutura Molecular , Inibidores de Fosfolipase A2/química , Ratos
19.
Mol Med Rep ; 13(6): 5269-75, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27122182

RESUMO

Increasing evidence has demonstrated that the activation of heme oxygenase (HO)­1 reduces autophagy stimulated by oxidative stress injury, in which the supraphysiological production of reactive oxygen species (ROS) is detected. However, the potential mechanism underlying this effect remains unclear. The present study aimed to investigate the function of HO­1 activation in the regulation of autophagy in glomerular mesangial cells subjected to H2O2­induced oxidative stress injury. The results demonstrated that the HO­1 agonist, hemin, reduces the LC3 protein level, which was enhanced by H2O2 treatment. Furthermore, hemin­activated HO­1 may function as a regulator of oxidative stress­induced autophagy in a dose­dependent manner. Pharmacological activation of c­Jun N­terminal kinase (JNK) inhibited the effect of hemin, indicating that the JNK signaling pathway is associated with the mechanism of HO­1 in impeding excessive autophagy. In addition to successfully alleviating H2O2­induced oxidative stress and cellular apoptosis, hemin­activated HO­1 may provide cytoprotection against rapamycin, a specific autophagy agonist. The present result suggested the inhibitory action of HO­1 in the avoidance of a potentially enhanced linkage between autophagy and apoptosis, particularly in the setting of excessive ROS. Therefore, enhancing the intracellular activity of HO­1 may assist the crosstalk between oxidative stress, autophagy and apoptosis, and represent a novel therapeutic strategy for renal ischemic disease.


Assuntos
Autofagia/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Peróxido de Hidrogênio/farmacocinética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células Mesangiais/enzimologia , Estresse Oxidativo/efeitos dos fármacos , Linhagem Celular , Humanos , MAP Quinase Quinase 4/metabolismo , Células Mesangiais/patologia
20.
Acta Histochem ; 118(2): 183-93, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26797190

RESUMO

Oxidative stress contributes substantially to the pathophysiology of diabetic nephropathy (DN). Consumption of an antioxidant-fortified (AO) diet from an early age prevents or delays later development of DN in the Zucker rat female with type 2 diabetes. We hypothesize this is due to effects on mesangial matrix and renal nitric oxide synthase (NOS) distribution and to sex-specific differences in NOS responses in the diabetic kidney. Total glomerular tuft area (GTA) and PAS-positive tuft area (PTA), endothelial (e), neuronal (n) and inducible (i) NOS were quantified in males and females on AO or regular (REG) diet at 6 and 20 weeks of age. eNOS was observed in glomeruli and tubules. nNOS predominantly localized to tubular epithelium in both cortex and medulla. iNOS was expressed in proximal and distal tubules and collecting ducts. Sex, diabetes duration and AO diet affected the distribution of the three isoforms. GTA and PTA increased with duration of hyperglycemia and showed a negative correlation with renal levels of all NOS isoforms. AO diet in both genders was associated with less PAS-positive staining and less mesangial expansion than the REG diet, an early increase in cortical iNOS in males, and sex-specific changes in cortical eNOS at 20 weeks. These effects of AO diet may contribute to sex-specific preservation of renal function in females.


Assuntos
Antioxidantes/administração & dosagem , Diabetes Mellitus Tipo 2/enzimologia , Nefropatias Diabéticas/enzimologia , Células Mesangiais/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Administração Oral , Animais , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/dietoterapia , Nefropatias Diabéticas/dietoterapia , Nefropatias Diabéticas/etiologia , Dieta , Feminino , Isoenzimas/metabolismo , Glomérulos Renais/enzimologia , Glomérulos Renais/patologia , Masculino , Ratos Zucker , Caracteres Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA