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










Base de datos
Intervalo de año de publicación
1.
Clin Transl Med ; 11(7): e460, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34323419

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Síndrome Nefrótico/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas WT1/metabolismo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Regulación hacia Abajo/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , FN-kappa B/metabolismo , Síndrome Nefrótico/metabolismo , Podocitos/citología , Podocitos/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Unión Proteica , Proteinuria/patología , Proteinuria/prevención & control , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/metabolismo , Activación Transcripcional , Proteínas WT1/genética
2.
Rev Prat ; 71(2): 198-205, 2021 Feb.
Artículo en Francés | MEDLINE | ID: mdl-34160984

RESUMEN

"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."


Asunto(s)
Antineoplásicos , Insuficiencia Renal , Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/efectos adversos , Humanos , Riñón , Oncología Médica , Terapia Molecular Dirigida/efectos adversos
3.
Diagnostics (Basel) ; 11(4)2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33916159

RESUMEN

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.

4.
Kidney Int ; 100(1): 90-106, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33675847

RESUMEN

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.


Asunto(s)
Podocitos , Angiotensina II/toxicidad , Animales , Autofagia , Proteínas de Unión al Calcio , Glomérulos Renales , Ratones
5.
Nephrol Dial Transplant ; 36(4): 609-617, 2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33241414

RESUMEN

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.


Asunto(s)
Factor Activador de Células B/sangre , Glomerulonefritis Membranosa/diagnóstico , Nefrosis Lipoidea/diagnóstico , Síndrome Nefrótico/diagnóstico , Células Plasmáticas/metabolismo , Adulto , Estudios de Casos y Controles , Femenino , Glomerulonefritis Membranosa/sangre , Humanos , Masculino , Persona de Mediana Edad , Nefrosis Lipoidea/sangre , Síndrome Nefrótico/sangre , Recurrencia
8.
Cell Mol Immunol ; 17(10): 1026-1041, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-31395948

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos CD28/metabolismo , Complejo CD3/metabolismo , Polaridad Celular , Citocinas/metabolismo , Activación Enzimática , Glicoesfingolípidos/metabolismo , Humanos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Microdominios de Membrana/metabolismo , Ratones Transgénicos , Fenotipo , Proteómica , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Familia-src Quinasas/metabolismo
9.
J Clin Med ; 8(9)2019 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-31470591

RESUMEN

Systemic lupus erythematosus (SLE) is characterized by a broad spectrum of renal lesions. In lupus glomerulonephritis, histological classifications are based on immune-complex (IC) deposits and hypercellularity lesions (mesangial and/or endocapillary) in the glomeruli. However, there is compelling evidence to suggest that glomerular epithelial cells, and podocytes in particular, are also involved in glomerular injury in patients with SLE. Podocytes now appear to be not only subject to collateral damage due to glomerular capillary lesions secondary to IC and inflammatory processes, but they are also a potential direct target in lupus nephritis. Improvements in our understanding of podocyte injury could improve the classification of lupus glomerulonephritis. Indeed, podocyte injury may be prominent in two major presentations: lupus podocytopathy and glomerular crescent formation, in which glomerular parietal epithelial cells play also a key role. We review here the contribution of podocyte impairment to different presentations of lupus nephritis, focusing on the podocyte signaling pathways involved in these lesions.

10.
Nat Commun ; 10(1): 3303, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31341160

RESUMEN

The mechanisms driving the development of extracapillary lesions in focal segmental glomerulosclerosis (FSGS) and crescentic glomerulonephritis (CGN) remain poorly understood. A key question is how parietal epithelial cells (PECs) invade glomerular capillaries, thereby promoting injury and kidney failure. Here we show that expression of the tetraspanin CD9 increases markedly in PECs in mouse models of CGN and FSGS, and in kidneys from individuals diagnosed with these diseases. Cd9 gene targeting in PECs prevents glomerular damage in CGN and FSGS mouse models. Mechanistically, CD9 deficiency prevents the oriented migration of PECs into the glomerular tuft and their acquisition of CD44 and ß1 integrin expression. These findings highlight a critical role for de novo expression of CD9 as a common pathogenic switch driving the PEC phenotype in CGN and FSGS, while offering a potential therapeutic avenue to treat these conditions.


Asunto(s)
Enfermedades Renales/patología , Tetraspanina 29/fisiología , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Femenino , Glomerulonefritis/genética , Glomerulonefritis/metabolismo , Glomerulonefritis/patología , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Enfermedades Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Tetraspanina 29/genética , Tetraspanina 29/metabolismo
11.
Nephrol Ther ; 15 Suppl 1: S1-S5, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30981386

RESUMEN

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.

12.
Bio Protoc ; 9(8): e3210, 2019 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-33655004

RESUMEN

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).

13.
J Am Soc Nephrol ; 29(3): 1049-1062, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29321143

RESUMEN

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.


Asunto(s)
Abatacept/farmacología , Linfocitos B/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Inmunosupresores/farmacología , Abatacept/uso terapéutico , Anciano , Aloinjertos/inmunología , Linfocitos B/fisiología , Antígeno B7-2/metabolismo , Antígenos CD28/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Femenino , Humanos , Inmunoglobulina G/biosíntesis , Inmunosupresores/uso terapéutico , Trasplante de Riñón , Masculino , Persona de Mediana Edad , Células Plasmáticas/fisiología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/fisiología
14.
J Autoimmun ; 88: 91-102, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29056249

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células T Asesinas Naturales/inmunología , Nefrosis Lipoidea/tratamiento farmacológico , Rituximab/uso terapéutico , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Inmunidad Adaptativa , Adolescente , Antígenos CD20/inmunología , Niño , Preescolar , Método Doble Ciego , Femenino , Factores de Transcripción Forkhead/metabolismo , Humanos , Inmunidad Innata , Masculino , Placebos , Receptores de Antígenos de Linfocitos T/metabolismo , Resultado del Tratamiento
15.
Nat Commun ; 8(1): 1829, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29184126

RESUMEN

Crescentic rapidly progressive glomerulonephritis (RPGN) represents the most aggressive form of acquired glomerular disease. While most therapeutic approaches involve potentially toxic immunosuppressive strategies, the pathophysiology remains incompletely understood. Podocytes are glomerular epithelial cells that are normally growth-arrested because of the expression of cyclin-dependent kinase (CDK) inhibitors. An exception is in RPGN where podocytes undergo a deregulation of their differentiated phenotype and proliferate. Here we demonstrate that microRNA-92a (miR-92a) is enriched in podocytes of patients and mice with RPGN. The CDK inhibitor p57Kip2 is a major target of miR-92a that constitutively safeguards podocyte cell cycle quiescence. Podocyte-specific deletion of miR-92a in mice de-repressed the expression of p57Kip2 and prevented glomerular injury in RPGN. Administration of an anti-miR-92a after disease initiation prevented albuminuria and kidney failure, indicating miR-92a inhibition as a potential therapeutic strategy for RPGN. We demonstrate that miRNA induction in epithelial cells can break glomerular tolerance to immune injury.


Asunto(s)
Glomerulonefritis/tratamiento farmacológico , Glomerulonefritis/patología , MicroARNs/antagonistas & inhibidores , Podocitos/citología , Adolescente , Adulto , Anciano , Animales , Antagomirs/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/farmacología , Quinasas Ciclina-Dependientes/metabolismo , Femenino , Eliminación de Gen , Perfilación de la Expresión Génica , Glomerulonefritis/genética , Glomerulonefritis/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Adulto Joven
16.
J Am Soc Nephrol ; 28(5): 1491-1505, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27895156

RESUMEN

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.


Asunto(s)
PPAR gamma/fisiología , Podocitos/efectos de los fármacos , Proteinuria/prevención & control , Citrato de Sildenafil/uso terapéutico , Canales Catiónicos TRPC/antagonistas & inhibidores , Animales , Regulación hacia Abajo , Ratones , Ratas , Canal Catiónico TRPC6
17.
J Am Coll Cardiol ; 68(25): 2776-2793, 2016 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-28007141

RESUMEN

BACKGROUND: Innate immune responses activated through myeloid cells contribute to the initiation, progression, and complications of atherosclerosis in experimental models. However, the critical upstream pathways that link innate immune activation to foam cell formation are still poorly identified. OBJECTIVES: This study sought to investigate the hypothesis that activation of the triggering receptor expressed on myeloid cells (TREM-1) plays a determinant role in macrophage atherogenic responses. METHODS: After genetically invalidating Trem-1 in chimeric Ldlr-/-Trem-1-/- mice and double knockout ApoE-/-Trem-1-/- mice, we pharmacologically inhibited Trem-1 using LR12 peptide. RESULTS: Ldlr-/- mice reconstituted with bone marrow deficient for Trem-1 (Trem-1-/-) showed a strong reduction of atherosclerotic plaque size in both the aortic sinus and the thoracoabdominal aorta, and were less inflammatory compared to plaques of Trem-1+/+ chimeric mice. Genetic invalidation of Trem-1 led to alteration of monocyte recruitment into atherosclerotic lesions and inhibited toll-like receptor 4 (TLR 4)-initiated proinflammatory macrophage responses. We identified a critical role for Trem-1 in the upregulation of cluster of differentiation 36 (CD36), thereby promoting the formation of inflammatory foam cells. Genetic invalidation of Trem-1 in ApoE-/-/Trem-1-/- mice or pharmacological blockade of Trem-1 in ApoE-/- mice using LR-12 peptide also significantly reduced the development of atherosclerosis throughout the vascular tree, and lessened plaque inflammation. TREM-1 was expressed in human atherosclerotic lesions, mainly in lipid-rich areas with significantly higher levels of expression in atheromatous than in fibrous plaques. CONCLUSIONS: We identified TREM-1 as a major upstream proatherogenic receptor. We propose that TREM-1 activation orchestrates monocyte/macrophage proinflammatory responses and foam cell formation through coordinated and combined activation of CD36 and TLR4. Blockade of TREM-1 signaling may constitute an attractive novel and double-hit approach for the treatment of atherosclerosis.


Asunto(s)
Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/terapia , Terapia Genética/métodos , Inmunidad Innata , Ácidos Láuricos/farmacología , Glicoproteínas de Membrana/biosíntesis , Placa Aterosclerótica/terapia , Receptores Inmunológicos/biosíntesis , Rodaminas/farmacología , Animales , Apoptosis , Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/inmunología , Enfermedades de las Arterias Carótidas/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligopéptidos , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/metabolismo , Receptores Inmunológicos/antagonistas & inhibidores , Receptor Activador Expresado en Células Mieloides 1
18.
Biochim Biophys Acta ; 1861(12 Pt A): 2000-2010, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27725263

RESUMEN

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.


Asunto(s)
Insulina/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Ácido Oléico/farmacología , Palmitatos/metabolismo , Transducción de Señal/fisiología , Animales , Carnitina/análogos & derivados , Carnitina/metabolismo , Línea Celular , Ceramidas/metabolismo , Diglicéridos/metabolismo , Ácidos Grasos/metabolismo , Resistencia a la Insulina/fisiología , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , FN-kappa B/metabolismo , Oxidación-Reducción/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Triglicéridos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Kidney Int ; 90(6): 1298-1311, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27650733

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Podocitos/metabolismo , Proteínas WT1/metabolismo , Animales , Secuencia de Bases , Síndrome de Denys-Drash/metabolismo , Femenino , Síndrome de Frasier/metabolismo , Regulación de la Expresión Génica , Humanos , Riñón/embriología , Masculino , Ratones , Regiones Promotoras Genéticas
20.
Gut ; 65(11): 1882-1894, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26338827

RESUMEN

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.


Asunto(s)
MicroARNs/metabolismo , Enfermedad del Hígado Graso no Alcohólico , Oligonucleótidos , PPAR alfa/metabolismo , Animales , Dieta Alta en Grasa , Perfilación de la Expresión Génica/métodos , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Metabolismo de los Lípidos , Lipoproteínas LDL/metabolismo , Ratones , MicroARNs/antagonistas & inhibidores , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Oligonucleótidos/metabolismo , Oligonucleótidos/farmacología , PPAR alfa/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA