Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Kidney Int ; 100(1): 90-106, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33675847

RESUMO

The strong predictive value of proteinuria in chronic glomerulopathies is firmly established as well as the pathogenic role of angiotensin II promoting progression of glomerular disease with an altered glomerular filtration barrier, podocyte injury and scarring of glomeruli. Here we found that chronic angiotensin II-induced hypertension inhibited autophagy flux in mouse glomeruli. Deletion of Atg5 (a gene encoding a protein involved autophagy) specifically in the podocyte resulted in accelerated angiotensin II-induced podocytopathy, accentuated albuminuria and glomerulosclerosis. This indicates that autophagy is a key protective mechanism in the podocyte in this condition. Angiotensin-II induced calpain activity in podocytes inhibits autophagy flux. Podocytes from mice with transgenic expression of the endogenous calpain inhibitor calpastatin displayed higher podocyte autophagy at baseline that was resistant to angiotensin II-dependent inhibition. Also, sustained autophagy with calpastatin limited podocyte damage and albuminuria. These findings suggest that hypertension has pathogenic effects on the glomerular structure and function, in part through activation of calpains leading to blockade of podocyte autophagy. These findings uncover an original mechanism whereby angiotensin II-mediated hypertension inhibits autophagy via calcium-induced recruitment of calpain with pathogenic consequences in case of imbalance by calpastatin activity. Thus, preventing a calpain-mediated decrease in autophagy may be a promising new therapeutic strategy for nephropathies associated with high renin-angiotensin system activity.


Assuntos
Podócitos , Angiotensina II/toxicidade , Animais , Autofagia , Proteínas de Ligação ao Cálcio , Glomérulos Renais , Camundongos
2.
Nephrol Dial Transplant ; 36(4): 609-617, 2021 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-33241414

RESUMO

BACKGROUND: The recent success achieved with the use of B cell-depleting agents in some patients with minimal change nephrotic syndrome (MCNS) suggests an unexpected role for B lymphocytes in the pathogenesis of this immune-mediated glomerular disease. Nevertheless, no extensive B-cell phenotyping analysis has ever been performed in untreated adult patients soon after MCNS diagnosis. METHODS: We investigated the distribution of the different B-cell subpopulations in 22 untreated adult patients with biopsy-proven MCNS [MCNS relapse (MCNS-Rel)]. We compared these data with those for 24 healthy controls, 13 MCNS patients in remission (with no specific treatment) and 19 patients with idiopathic membranous nephropathy (IMN). RESULTS: Patients with MCNS-Rel or IMN had higher proteinuria and lower serum albumin and gammaglobulin levels (P < 0.0001 for all comparisons) than MCNS patients in remission. Plasmablasts were the only B-cell subsets present at significantly higher levels in MCNS-Rel patients than in the patients of the other three groups (P < 0.05 for all comparisons). The lower albumin levels and higher proteinuria levels were positively correlated with the percentage of circulating plasmablasts (Spearman test's ρ = -0.54, P = 0.01 and ρ = 0.65, P = 0.002, respectively). Similarly, the increase of immunoglobulin M (IgM) and the decrease of IgG levels were significantly associated with the percentage of plasmablasts in MCNS-Rel patients (Spearman's ρ = 0.36, P = 0.01 and Spearman's ρ = -0.60, P = 0.01, respectively). Increased production of interleukin (IL)-21, IL-6 and B-cell activating factor (BAFF) in the serum of MCNS-Rel patients was found significantly correlated with the percentage of plasmablasts (ρ = 0.72, P = 0.0002, ρ = 0.49, P = 0.04 and ρ = 0.62, P = 0.009, respectively). CONCLUSIONS: An increase in the proportion of circulating plasmablasts seems to be a hallmark of untreated MCNS in adult patients. Further studies are required to more precisely determine the phenotype and functions of these cells.


Assuntos
Fator Ativador de Células B/sangue , Glomerulonefrite Membranosa/diagnóstico , Nefrose Lipoide/diagnóstico , Síndrome Nefrótica/diagnóstico , Plasmócitos/metabolismo , Adulto , Estudos de Casos e Controles , Feminino , Glomerulonefrite Membranosa/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Nefrose Lipoide/sangue , Síndrome Nefrótica/sangue , Recidiva
3.
J Am Soc Nephrol ; 29(3): 1049-1062, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29321143

RESUMO

Generation of de novo donor-specific antibodies (dnDSAs) after renal transplant is recognized as the leading cause of late transplant failure. Hence, the optimal immunosuppressive strategies to limit dnDSA development need to be defined. Recent clinical trials using the novel costimulatory blockade agent CTLA4-Ig (Belatacept) have shown that kidney transplant recipients (KTRs) treated with Belatacept have better graft survival and function and a lower proportion of dnDSAs than control-treated KTRs. Mechanisms involved in the control of humoral responses by Belatacept remain to be investigated. Here, we analyzed the effect of Belatacept on different steps of the B cell-mediated response in humans. In vitro, Belatacept reduced plasmablast differentiation, Ig production, and the expression of the major transcription factor involved in plasma cell function, Blimp-1, in a T cell-independent manner. Moreover, Belatacept induced activation of the STAT3 transcription factor in stimulated B cells and reduced the expression of CD86. Additionally, Belatacept blocked CD28-mediated activation of T follicular helper cells (Tfhs) in an autologous Tfh-memory B cells model. We then validated these observations in KTRs treated with Belatacept, who had a reduced proportion of blood effector B cells and activated Tfh (PD1+ICOS+) compared with control-treated KTRs. Our in vitro and in vivo results suggest that Belatacept modulates B cell function directly and at the level of B cell-Tfh interaction. These mechanisms likely account for the optimal control of humoral responses observed in KTRs treated with Belatacept.


Assuntos
Abatacepte/farmacologia , Linfócitos B/efeitos dos fármacos , Comunicação Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Imunossupressores/farmacologia , Abatacepte/uso terapêutico , Idoso , Aloenxertos/imunologia , Linfócitos B/fisiologia , Antígeno B7-2/metabolismo , Antígenos CD28/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Feminino , Humanos , Imunoglobulina G/biossíntese , Imunossupressores/uso terapêutico , Transplante de Rim , Masculino , Pessoa de Meia-Idade , Plasmócitos/fisiologia , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/fisiologia
4.
J Autoimmun ; 88: 91-102, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29056249

RESUMO

Minimal-change nephrotic syndrome (MCNS) is an immune-mediated glomerular disease. We have analyzed the modifications on T-cell subsets in twenty-three patients who were highly steroid/calcineurin inhibitor and/or mycophenolate mofetil-dependent for frequently relapsing nephrotic syndrome (FRNS) and who were enrolled in a multicenter, double-blind, randomized, placebo vs Rituximab-controlled trial. Patients with FRNS entered the trial at remission and were randomly assigned to receive either Rituximab or placebo. In both groups, patient blood samples were analyzed at inclusion and then monthly until six months post-perfusion. Disclosure of patient's allocation code occurred in relapse or at the end of the trial. All patients under placebo displaying relapse were subsequently treated with Rituximab. Despite the significant decrease of immunosuppressive drugs, remission was maintained in all patients included in the Rituximab group, except one (n = 9/10). On the other hand, relapses occurred within a few weeks (means ≈ 7.3 weeks) in all patients receiving placebo (n = 13). At inclusion, before rituximab therapy, the frequency of different T-cell subsets were highly similar in both groups, except for CD8+ and invariant TCRVα24 T-cell subsets, which were significantly increased in patients of the Placebo group ((p = 0,0414 and p = 0.0428, respectively). Despite the significant decrease of immunosuppressive drugs, remission was maintained in all patients included in the Rituximab group (n = 10), except one. Relapses were associated with a significant decrease in CD4+CD25highFoxP3high Tregulatory cells (p = 0.0005) and IL2 expression (p = 0.0032), while CMIP abundance was significantly increased (p = 0.03). Remissions after Rituximab therapy were associated in both groups with significant decrease in the frequency of CD4+CD45RO+CXCR5+, invariant natural killer T-cells (INKT) and CD4-CD8- (double-negative, DN) T-cells expressing the invariant Vα24 chain (DN-TCR Vα24) T-cells, suggesting that MCNS involves a disorder of innate and adaptive immune response, which can be stabilized by Rituximab treatment.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células T Matadoras Naturais/imunologia , Nefrose Lipoide/tratamento farmacológico , Rituximab/uso terapêutico , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Imunidade Adaptativa , Adolescente , Antígenos CD20/imunologia , Criança , Pré-Escolar , Método Duplo-Cego , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Imunidade Inata , Masculino , Placebos , Receptores de Antígenos de Linfócitos T/metabolismo , Resultado do Tratamento
5.
J Am Soc Nephrol ; 28(5): 1491-1505, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27895156

RESUMO

Transient receptor potential channel C6 (TRPC6) gain-of-function mutations and increased TRPC6 expression in podocytes induce glomerular injury and proteinuria. Sildenafil reduces TRPC6 expression and activity in nonrenal cell types, although the mechanism is unknown. Peroxisome proliferator-activated receptor γ (PPAR-γ) is a downstream target of sildenafil in the cyclic guanosine monophosphate (cGMP)-activated protein kinase G (PKG) axis. PPAR-γ agonists, like pioglitazone, appear antiproteinuric. We hypothesized that sildenafil inhibits TRPC6 expression in podocytes through PPAR-γ-dependent mechanisms, thereby counteracting podocyte injury and proteinuria. Treatment with sildenafil, the cGMP derivative 8-bromoguanosine 3',5'-cyclic monophosphate sodium salt (8-Br-cGMP), or pioglitazone dose-dependently downregulated podocyte injury-induced TRPC6 expression in vitro Knockdown or application of antagonists of PKG or PPAR-γ enhanced TRPC6 expression in podocytes and counteracted effects of sildenafil and 8-Br-cGMP. We observed similar effects on TRPC6 promoter activity and TRPC6-dependent calcium influx. Chromatin immunoprecipitation showed PPAR-γ binding to the TRPC6 promoter. Sildenafil or pioglitazone treatment prevented proteinuria and the increased TRPC6 expression in rats with adriamycin-induced nephropathy and mice with hyperglycemia-induced renal injury. Rats receiving PPAR-γ antagonists displayed proteinuria and increased podocyte TRPC6 expression, as did podocyte-specific PPAR-γ knockout mice, which were more sensitive to adriamycin and not protected by sildenafil. Thus, sildenafil ameliorates podocyte injury and prevents proteinuria through cGMP- and PKG-dependent binding of PPAR-γ to the TRPC6 promoter, which inhibits TRPC6 promoter activity, expression, and activity. Because sildenafil is approved for clinical use, our results suggest that additional clinical study of its antiproteinuric effect in glomerular disease is warranted.


Assuntos
PPAR gama/fisiologia , Podócitos/efeitos dos fármacos , Proteinúria/prevenção & controle , Citrato de Sildenafila/uso terapêutico , Canais de Cátion TRPC/antagonistas & inibidores , Animais , Regulação para Baixo , Camundongos , Ratos , Canal de Cátion TRPC6
6.
Biochim Biophys Acta ; 1861(12 Pt A): 2000-2010, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27725263

RESUMO

Because the protective effect of oleate against palmitate-induced insulin resistance may be lessened in skeletal muscle once cell metabolism is overloaded by fatty acids (FAs), we examined the impact of varying amounts of oleate on palmitate metabolic channeling and insulin signaling in C2C12 myotubes. Cells were exposed to 0.5mM of palmitate and to increasing doses of oleate (0.05, 0.25 and 0.5mM). Impacts of FA treatments on radio-labelled FA fluxes, on cellular content in diacylglycerols (DAG), triacylglycerols (TAG), ceramides, acylcarnitines, on PKCθ, MAPKs (ERK1/2, p38) and NF-ΚB activation, and on insulin-dependent Akt phosphorylation were examined. Low dose of oleate (0.05mM) was sufficient to improve palmitate complete oxidation to CO2 (+29%, P<0.05) and to alter the cellular acylcarnitine profile. Insulin-induced Akt phosphorylation was 48% higher in that condition vs. palmitate alone (p<0.01). Although DAG and ceramide contents were significantly decreased with 0.05mM of oleate vs. palmitate alone (-47 and -28%, respectively, p<0.01), 0.25mM of oleate was required to decrease p38 MAPK and PKCθ phosphorylation, thus further improving the insulin signaling (+32%, p<0.05). By contrast, increasing oleate concentration from 0.25 to 0.5mM, thus increasing total amount of FA from 0.75 to 1mM, deteriorated the insulin signaling pathway (-30%, p<0.01). This was observed despite low contents in DAG and ceramides, and enhanced palmitate incorporation into TAG (+27%, p<0.05). This was associated with increased incomplete FA ß-oxidation and impairment of acylcarnitine profile. In conclusion, these combined data place mitochondrial ß-oxidation at the center of the regulation of muscle insulin sensitivity, besides p38 MAPK and PKCθ.


Assuntos
Insulina/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Ácido Oleico/farmacologia , Palmitatos/metabolismo , Transdução de Sinais/fisiologia , Animais , Carnitina/análogos & derivados , Carnitina/metabolismo , Linhagem Celular , Ceramidas/metabolismo , Diglicerídeos/metabolismo , Ácidos Graxos/metabolismo , Resistência à Insulina/fisiologia , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , NF-kappa B/metabolismo , Oxirredução/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Triglicerídeos/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
J Am Soc Nephrol ; 27(1): 172-88, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25999406

RESUMO

Necrotizing and crescentic rapidly progressive GN (RPGN) is a life-threatening syndrome characterized by a rapid loss of renal function. Evidence suggests that podocyte expression of the transcription factor peroxisome proliferator-activated receptor γ (PPARγ) may prevent podocyte injury, but the function of glomerular PPARγ in acute, severe inflammatory GN is unknown. Here, we observed marked loss of PPARγ abundance and transcriptional activity in glomerular podocytes in experimental RPGN. Blunted expression of PPARγ in podocyte nuclei was also found in kidneys from patients diagnosed with crescentic GN. Podocyte-specific Pparγ gene targeting accentuated glomerular damage, with increased urinary loss of albumin and severe kidney failure. Furthermore, a PPARγ gain-of-function approach achieved by systemic administration of thiazolidinedione (TZD) failed to prevent severe RPGN in mice with podocyte-specific Pparγ gene deficiency. In nuclear factor erythroid 2-related factor 2 (NRF2)-deficient mice, loss of podocyte PPARγ was observed at baseline. NRF2 deficiency markedly aggravated the course of RPGN, an effect that was partially prevented by TZD administration. Furthermore, delayed administration of TZD, initiated after the onset of RPGN, still alleviated the severity of experimental RPGN. These findings establish a requirement for the NRF2-PPARγ cascade in podocytes, and we suggest that these transcription factors have a role in augmenting the tolerance of glomeruli to severe immune-complex mediated injury. The NRF2-PPARγ pathway may be a therapeutic target for RPGN.


Assuntos
Glomerulonefrite/etiologia , Fator 2 Relacionado a NF-E2/fisiologia , PPAR gama/biossíntese , Podócitos/metabolismo , Animais , Masculino , Camundongos
8.
Gut ; 65(11): 1882-1894, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26338827

RESUMO

OBJECTIVE: Previous studies suggested that microRNA-21 may be upregulated in the liver in non-alcoholic steatohepatitis (NASH), but its role in the development of this disease remains unknown. This study aimed to determine the role of microRNA-21 in NASH. DESIGN: We inhibited or suppressed microRNA-21 in different mouse models of NASH: (a) low-density lipoprotein receptor-deficient (Ldlr-/-) mice fed a high-fat diet and treated with antagomir-21 or antagomir control; (b) microRNA-21-deficient and wild-type mice fed a methionine-choline-deficient (MCD) diet; (c) peroxisome proliferation-activator receptor α (PPARα)-deficient mice fed an MCD diet and treated with antagomir-21 or antagomir control. We assessed features of NASH and determined liver microRNA-21 levels and cell localisation. MicroRNA-21 levels were also quantified in the liver of patients with NASH, bland steatosis or normal liver and localisation was determined. RESULTS: Inhibiting or suppressing liver microRNA-21 expression reduced liver cell injury, inflammation and fibrogenesis without affecting liver lipid accumulation in Ldlr-/- fed a high-fat diet and in wild-type mice fed an MCD diet. Liver microRNA-21 was overexpressed, primarily in biliary and inflammatory cells, in mouse models as well as in patients with NASH, but not in patients with bland steatosis. PPARα, a known microRNA-21 target, implicated in NASH, was decreased in the liver of mice with NASH and restored following microRNA-21 inhibition or suppression. The effect of antagomir-21 was lost in PPARα-deficient mice. CONCLUSIONS: MicroRNA-21 inhibition or suppression decreases liver injury, inflammation and fibrosis, by restoring PPARα expression. Antagomir-21 might be a future therapeutic strategy for NASH.


Assuntos
MicroRNAs/metabolismo , Hepatopatia Gordurosa não Alcoólica , Oligonucleotídeos , PPAR alfa/metabolismo , Animais , Dieta Hiperlipídica , Perfilação da Expressão Gênica/métodos , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Metabolismo dos Lipídeos , Lipoproteínas LDL/metabolismo , Camundongos , MicroRNAs/antagonistas & inibidores , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , Oligonucleotídeos/metabolismo , Oligonucleotídeos/farmacologia , PPAR alfa/antagonistas & inibidores
9.
Kidney Int ; 90(6): 1298-1311, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27650733

RESUMO

The WT1 (Wilm's tumor suppressor) gene is expressed throughout life in podocytes and is essential for the functional integrity of the glomerular filtration barrier. We have previously shown that CMIP (C-Maf inducing protein) is overproduced in podocyte diseases and alters intracellular signaling. Here we isolated the proximal region of the human CMIP promoter and showed by chromatin immunoprecipitation assays and electrophoretic-mobility shift that Wilm's tumor protein (WT1) bound to 2 WT1 response elements, located at positions -290/-274 and -57/-41 relative to transcription start site. Unlike the human CMIP gene, only one Wt1 response element was identified in the mouse Cmip proximal promoter located at position -217/-206. Luciferase reporter assays indicated that WT1 dose-dependently inhibited the transcriptional induction of the CMIP promoter. Transfection of decoy oligonucleotides mimicking the WT1 response elements prevented the inhibition of WT1 on CMIP promoter activity. Furthermore, WT1 silencing promoted Cmip expression. In line with these findings, the abundance of Cmip was early and significantly increased at the transcript and protein level in podocytes displaying a primary defect in Wt1, including Denys-Drash syndrome and Frasier syndrome. Thus, WT1 is a major repressor of the CMIP gene in physiological situations, while conditional deletion of CMIP in the developing kidney did not affect the development of mature glomeruli.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Podócitos/metabolismo , Proteínas WT1/metabolismo , Animais , Sequência de Bases , Síndrome de Denys-Drash/metabolismo , Feminino , Síndrome de Frasier/metabolismo , Regulação da Expressão Gênica , Humanos , Rim/embriologia , Masculino , Camundongos , Regiões Promotoras Genéticas
10.
FASEB J ; 29(6): 2473-83, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25713059

RESUMO

Adult skeletal muscle is a dynamic, remarkably plastic tissue, which allows myofibers to switch from fast/glycolytic to slow/oxidative types and to increase mitochondrial fatty acid oxidation (mFAO) capacity and vascularization in response to exercise training. mFAO is the main muscle energy source during endurance exercise, with carnitine palmitoyltransferase 1 (CPT1) being the key regulatory enzyme. Whether increasing muscle mFAO affects skeletal muscle physiology in adulthood actually remains unknown. To investigate this, we used in vivo electrotransfer technology to express in mouse tibialis anterior (TA), a fast/glycolytic muscle, a mutated CPT1 form (CPT1mt) that is active but insensitive to malonyl-CoA, its physiologic inhibitor. In young (2-mo-old) adult mice, muscle CPT1mt expression enhanced mFAO (+40%), but also increased the percentage of oxidative fibers (+28%), glycogen content, and capillary-to-fiber density (+45%). This CPT1mt-induced muscle remodeling, which mimicked exercise-induced oxidative phenotype, led to a greater resistance to muscle fatigue. In the context of aging, characterized by sarcopenia and reduced oxidative capacity, CPT1mt expression in TAs from aged (20-mo-old) mice partially reversed aging-associated sarcopenia and fiber-type transition, and increased muscle capillarity. These findings provide evidence that mFAO regulates muscle phenotype and may be a potential target to combat age-related decline in muscle function.


Assuntos
Carnitina O-Palmitoiltransferase/metabolismo , Ácidos Graxos/metabolismo , Mitocôndrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Fatores Etários , Animais , Western Blotting , Carnitina O-Palmitoiltransferase/genética , Expressão Gênica , Glicogênio/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias Musculares/fisiologia , Fadiga Muscular/genética , Fadiga Muscular/fisiologia , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/fisiologia , Mutação , Oxirredução , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sarcopenia/genética , Sarcopenia/fisiopatologia , Transfecção
11.
J Am Soc Nephrol ; 25(5): 1050-62, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24722437

RESUMO

The endothelin system has emerged as a novel target for the treatment of diabetic nephropathy. Endothelin-1 promotes mesangial cell proliferation and sclerosis. However, no direct pathogenic effect of endothelin-1 on podocytes has been shown in vivo and endothelin-1 signaling in podocytes has not been investigated. This study investigated endothelin effects in podocytes during experimental diabetic nephropathy. Stimulation of primary mouse podocytes with endothelin-1 elicited rapid calcium transients mediated by endothelin type A receptors (ETARs) and endothelin type B receptors (ETBRs). We then generated mice with a podocyte-specific double deletion of ETAR and ETBR (NPHS2-Cre×Ednra(lox/lox)×Ednrb(lox/lox) [Pod-ETRKO]). In vitro, treatment with endothelin-1 increased total ß-catenin and phospho-NF-κB expression in wild-type glomeruli, but this effect was attenuated in Pod-ETRKO glomeruli. After streptozotocin injection to induce diabetes, wild-type mice developed mild diabetic nephropathy with microalbuminuria, mesangial matrix expansion, glomerular basement membrane thickening, and podocyte loss, whereas Pod-ETRKO mice presented less albuminuria and were completely protected from glomerulosclerosis and podocyte loss, even when uninephrectomized. Moreover, glomeruli from normal and diabetic Pod-ETRKO mice expressed substantially less total ß-catenin and phospho-NF-κB compared with glomeruli from counterpart wild-type mice. This evidence suggests that endothelin-1 drives development of glomerulosclerosis and podocyte loss through direct activation of endothelin receptors and NF-κB and ß-catenin pathways in podocytes. Notably, both the expression and function of the ETBR subtype were found to be important. Furthermore, these results indicate that activation of the endothelin-1 pathways selectively in podocytes mediates pathophysiologic crosstalk that influences mesangial architecture and sclerosis.


Assuntos
Nefropatias Diabéticas/etiologia , Endotelina-1/fisiologia , Podócitos/metabolismo , Podócitos/patologia , Animais , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Regulação para Baixo/genética , Regulação para Baixo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Receptor de Endotelina A/genética , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/genética , Receptor de Endotelina B/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , beta Catenina/metabolismo
12.
Curr Opin Nephrol Hypertens ; 21(4): 417-27, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22660552

RESUMO

PURPOSE OF REVIEW: Podocytes are highly specialized epithelial cells that line the urinary surface of the glomerular capillary tuft. Dysfunction or death of podocytes impacts glomerular permeability and filtration. Here, we discuss the recent findings about the role of specific cell signaling pathways in glomerular diseases with an emphasis on the molecules in the podocyte that represent candidate therapeutic targets. RECENT FINDINGS: A number of local endogenous factors that retard the progression of diabetic nephropathy have recently been identified and include angiopoietin-1 (Angpt1), Smad7 and nuclear factor (erythroid-derived 2)-like 2 (Nrf2). Calcium-dependent regulation of podocyte actin dynamics involving transient receptor potential canonical (TRPC) channels and the Rho and Rac small GTPases has been shown to play important functions in glomerular health and disease. A central role for mammalian target of rapamycin (mTOR) activation in the development of diabetic nephropathy and regulation of autophagic flux in podocytes during aging has been demonstrated. Discovery of a circulating factor (suPAR) that can modulate outside-in beta3 integrin signaling in recurrent focal segmental glomerulosclerosis provides exciting therapeutic possibilities. Another secreted factor, the hyposialylated form of angiopoietin-like-protein 4 (ANGPTL4) was found to favor albuminuria in rats and in minimal change disease. Therapeutic sialylation of ANGPTL4 could limit albuminuria. Finally, neutralization of de novo paracrine activation of glomerular epithelial cells by heparin-binding epidermal growth factor (EGF)-like growth factor or EGF receptor antagonists could limit crescent formation and renal failure in immune-mediated vasculitis. SUMMARY: Here, we review the recent developments in our understanding of signaling pathways required for podocyte function in health and disease. Manipulation of these pathways provides an attractive therapeutic option for glomerular diseases.


Assuntos
Nefropatias/tratamento farmacológico , Glomérulos Renais/efeitos dos fármacos , Terapia de Alvo Molecular , Transdução de Sinais/efeitos dos fármacos , Animais , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Barreira de Filtração Glomerular/efeitos dos fármacos , Barreira de Filtração Glomerular/metabolismo , Humanos , Nefropatias/metabolismo , Nefropatias/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , Podócitos/efeitos dos fármacos , Podócitos/metabolismo
13.
Nephrol Dial Transplant ; 27(4): 1297-304, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22467748

RESUMO

Glomerular kidney diseases are of major public health importance because of their strong impact on the quality of life of patients and of their costly management. A relatively neglected area of study is the local factors that influence development of glomerular demolition. The involvement of a glomerular factor has been now demonstrated in glomerulonephritis with cell proliferation such as crescentic rapidly progressive glomerulonephritis (RPGN). Various unrelated immune disorders promote RPGN, such as antibodies directed against the glomerular basement membrane, deposition of immune complexes or antibodies directed against neutrophils. Despite the heterogeneity of these causing diseases, RPGNs share similar histopathological features, which suggest involvement of common final pathways. De novo expression of heparin-binding epidermal growth factor-like growth factor (HB-EGF) in glomerular epithelial cells is found specifically in human glomerulonephritis with proliferation of these cells and dedifferentiation of podocytes. A receptor for HB-EGF, the EGF receptor (EGFR), is expressed by parietal epithelial cells and podocytes. Furthermore, in a mouse model of RPGN, HB-EGF deficiency or conditional targeting of the Egfr alleles in podocytes markedly alleviated RPGN, renal failure and death. This indicates that the HB-EGF/EGFR pathway plays a pivotal role in RPGN and opens therapeutic perspectives as EGFR inhibitors are clinically available.


Assuntos
Fator de Crescimento Epidérmico/antagonistas & inibidores , Receptores ErbB/antagonistas & inibidores , Glomerulonefrite/tratamento farmacológico , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/fisiopatologia , Animais , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Humanos , Camundongos
14.
J Biol Chem ; 285(47): 36818-27, 2010 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-20837491

RESUMO

The mechanisms underlying the protective effect of monounsaturated fatty acids (e.g. oleate) against the lipotoxic action of saturated fatty acids (e.g. palmitate) in skeletal muscle cells remain poorly understood. This study aimed to examine the role of mitochondrial long-chain fatty acid (LCFA) oxidation in mediating oleate's protective effect against palmitate-induced lipotoxicity. CPT1 (carnitine palmitoyltransferase 1), which is the key regulatory enzyme of mitochondrial LCFA oxidation, is inhibited by malonyl-CoA, an intermediate of lipogenesis. We showed that expression of a mutant form of CPT1 (CPT1mt), which is active but insensitive to malonyl-CoA inhibition, in C2C12 myotubes led to increased LCFA oxidation flux even in the presence of high concentrations of glucose and insulin. Furthermore, similar to preincubation with oleate, CPT1mt expression protected muscle cells from palmitate-induced apoptosis and insulin resistance by decreasing the content of deleterious palmitate derivates (i.e. diacylglycerols and ceramides). Oleate preincubation exerted its protective effect by two mechanisms: (i) in contrast to CPT1mt expression, oleate preincubation increased the channeling of palmitate toward triglycerides, as a result of enhanced diacylglycerol acyltransferase 2 expression, and (ii) oleate preincubation promoted palmitate oxidation through increasing CPT1 expression and modulating the activities of acetyl-CoA carboxylase and AMP-activated protein kinase. In conclusion, we demonstrated that targeting mitochondrial LCFA oxidation via CPT1mt expression leads to the same protective effect as oleate preincubation, providing strong evidence that redirecting palmitate metabolism toward oxidation is sufficient to protect against palmitate-induced lipotoxicity.


Assuntos
Apoptose , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Ácido Oleico/química , Palmitatos/farmacologia , Animais , Western Blotting , Carnitina O-Palmitoiltransferase/metabolismo , Células Cultivadas , Imunofluorescência , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Ácido Oleico/metabolismo , Oxirredução , Consumo de Oxigênio , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Diagnostics (Basel) ; 11(4)2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33916159

RESUMO

The use of inhibitors of vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor 2 (VEGFR2) signaling for the treatment of cancer has increased over the last decade. This signaling pathway plays a fundamental role in angiogenesis and also in kidney physiology. The emergence of anti-angiogenic therapies has led to adverse nephrotoxic effects, despite improving the outcomes of patients. In this review, we will present the different anti-angiogenic therapies targeting the VEGFR pathway in association with the incidence of renal manifestations during their use. In addition, we will discuss, in detail, the pathophysiological mechanisms of frequent renal diseases such as hypertension, proteinuria, renal dysfunction, and electrolyte disorders. Finally, we will outline the cellular damage described following these therapies.

16.
Rev Prat ; 71(2): 198-205, 2021 Feb.
Artigo em Francês | MEDLINE | ID: mdl-34160984

RESUMO

"Targeted therapies and pathophysiological mechanisms of proteinuria Targeted therapy represents a promising therapeutic approach for patients with diverse cancers and has enabled significant development in medical oncology. This new class of anticancer drugs includes antibodies, fusion-proteins and receptor tyrosine kinase inhibitors among others. Depending on their molecular targeting, side effects can affect multiple organs, especially the kidney. Antiangiogenic agents inhibit the VEGF/VEGFR pathway resulting in reduction of nitric oxide production and alteration of podocytes function, which causes hypertension and proteinuria. EGFR inhibitors are responsible of electrolytic disorders. Hereby, we synthetized the current knowledge on renal toxicities on main molecular targeted therapies. Toxicities management is mainly based on clinical and biological monitoring, which can lead to drug withdrawing or dose adaptation."


"Thérapies ciblées et mécanismes physiopathologiques de la protéinurie Les thérapies ciblées occupent désormais une place majeure en oncologie médicale. Elles existent sous plusieurs formes d'administration, parentérale ou per os, et comportent entre autres des anticorps, des protéines de fusion et des inhibiteurs de récepteur des tyrosine kinases. Les effets indésirables liés à ces thérapies dépendent généralement de leur classe thérapeutique. Les antiangiogéniques, qui altèrent les podocytes et la production de monoxyde d'azote via l'inhibition de la voie VEGF/VEGFR, entraînent hypertension artérielle et protéinurie et sont parfois à l'origine de microangiopathies thrombotiques. Les inhibiteurs de l'EGFR entraînent principalement des troubles hydroélectrolytiques par inhibition de transporteurs rénaux. D'autres toxicités sont plus rares, comme les inhibiteurs de BRAF, parfois à l'origine d'insuffisances rénales aiguës immuno-allergiques. La prise en charge de ces toxicités repose généralement sur la surveillance clinique et biologique et peut conduire à la suspension des traitements ou à leur adaptation posologique."


Assuntos
Antineoplásicos , Insuficiência Renal , Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/efeitos adversos , Humanos , Rim , Oncologia , Terapia de Alvo Molecular/efeitos adversos
17.
Clin Transl Med ; 11(7): e460, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34323419

RESUMO

BACKGROUND: The Wilms tumor 1 suppressor gene, WT1, is expressed throughout life in podocytes and is essential for their function. Downregulation of WT1 has been reported in podocyte diseases but the underlying mechanisms remain unclear. Podocyte injury is the hallmark of idiopathic nephrotic syndrome (INS), the most frequent glomerular disease in children and young adults. An increase in the abundance of Cmaf-inducing protein (CMIP) has been found to alter podocyte function, but it is not known whether CMIP affects WT1 expression. METHODS: Transcriptional and post-transcriptional regulation of WT1in the presence of CMIP was studied using transient transfection, mouse models, and siRNA handling. RESULTS: We showed that overproduction of CMIP in the podocyte was consistently associated with a downregulation of WT1 according to two mechanisms. We found that CMIP prevented the NF-kB-mediated transcriptional activation of WT1. We demonstrated that CMIP interacts directly with WT1 through its leucine-rich repeat domain. Overexpression of CMIP in the M15 cell line induced a downregulation of WT1, which was prevented by lactacystin, a potent proteasome inhibitor. We showed that CMIP exhibits an E3 ligase activity and targets WT1 to proteasome degradation. Intravenous injection of Cmip-siRNA specifically prevented the repression of Wt1 in lipopolysaccharides-induced proteinuria in mice. CONCLUSIONS: These data suggest that CMIP is a repressor of WT1 and might be a critical player in the pathophysiology of some podocyte diseases. Because WT1 is required for podocyte integrity, CMIP could be considered a therapeutic target in podocyte diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Síndrome Nefrótica/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas WT1/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Regulação para Baixo/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/metabolismo , Síndrome Nefrótica/metabolismo , Podócitos/citologia , Podócitos/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Ligação Proteica , Proteinúria/patologia , Proteinúria/prevenção & controle , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/metabolismo , Ativação Transcricional , Proteínas WT1/genética
18.
Cell Mol Immunol ; 17(10): 1026-1041, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31395948

RESUMO

Upon their interaction with cognate antigen, T cells integrate different extracellular and intracellular signals involving basal and induced protein-protein interactions, as well as the binding of proteins to lipids, which can lead to either cell activation or inhibition. Here, we show that the selective T cell expression of CMIP, a new adapter protein, by targeted transgenesis drives T cells toward a naïve phenotype. We found that CMIP inhibits activation of the Src kinases Fyn and Lck after CD3/CD28 costimulation and the subsequent localization of Fyn and Lck to LRs. Video microscopy analysis showed that CMIP blocks the recruitment of LAT and the lipid raft marker cholera toxin B at the site of TCR engagement. Proteomic analysis identified several protein clusters differentially modulated by CMIP and, notably, Cofilin-1, which is inactivated in CMIP-expressing T cells. Moreover, transgenic T cells exhibited the downregulation of GM3 synthase, a key enzyme involved in the biosynthesis of gangliosides. These results suggest that CMIP negatively impacts proximal signaling and cytoskeletal rearrangement and defines a new mechanism for the negative regulation of T cells that could be a therapeutic target.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transdução de Sinais , Linfócitos T/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Antígenos CD28/metabolismo , Complexo CD3/metabolismo , Polaridade Celular , Citocinas/metabolismo , Ativação Enzimática , Glicoesfingolipídeos/metabolismo , Humanos , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Microdomínios da Membrana/metabolismo , Camundongos Transgênicos , Fenótipo , Proteômica , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Quinases da Família src/metabolismo
19.
Bio Protoc ; 9(8): e3210, 2019 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-33655004

RESUMO

In glomerular diseases, podocytes are one type of cells involved in dysfunction of glomerular filtration. In these conditions, podocyte proteins expression change. Therefore, immunofluorescent staining of podocytes can be performed to visualize podocyte localization of proteins of interest. In this protocol, we detail a method to stain podocytes with a specific marker WT-1 (Wilms Tumor-1) for "in situ" podocyte analysis by immunofluorescent microscopy, more informative technique than other techniques (as Western blot).

20.
Nephrol Ther ; 15 Suppl 1: S1-S5, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30981386

RESUMO

Necrotizing and crescentic rapidly progressive glomerulonephritis or crescentic glomerulonephritis is one of the severest forms of acquired glomerular diseases with significant mortality. Risk of end-stage renal failure at 5 years is near 30%, with a number of patients developing chronic kidney disease. Currently, autoimmune crescentic glomerulonephritides are treated with broad-spectrum immunosuppression inducing remission of the injury in the majority of patients. However, treatment is associated with significant side effects and by the time remission is achieved the majority of patients have developed renal tissue damage and significant impairment of their kidney function with a steep slope of deterioration within the first weeks following initiation of immunosuppression. It is therefore important to develop complementary strategies that would be immediately active on the common process of destructive epithelial processes. We have worked to identify the major cellular pathways contributing to glomerular destruction in this context by a systematic comparison of patient tissues and experimental models. Our studies demonstrate the pivotal role of local intra- and intercellular communications in orchestrating the global glomerular tolerance to a severe rapidly progressive glomerulonephritis model with excellent anatomoclinical correlative expressions in kidney biopsies of individuals diagnosed with crescentic glomerulonephritis, irrespectively of the causal immune disorder. We hope that such approaches deciphering mechanisms of cellular adaptation that underlie kidney damage control in response to vasculitides, integrating both stress and damage responses, will delineate novel complementary therapies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA