Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
Aging Cell ; 22(11): e13919, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37403257

RESUMEN

Caloric restriction is a non-pharmacological intervention known to ameliorate the metabolic defects associated with aging, including insulin resistance. The levels of miRNA expression may represent a predictive tool for aging-related alterations. In order to investigate the role of miRNAs underlying insulin resistance in adipose tissue during the early stages of aging, 3- and 12-month-old male animals fed ad libitum, and 12-month-old male animals fed with a 20% caloric restricted diet were used. In this work we demonstrate that specific miRNAs may contribute to the impaired insulin-stimulated glucose metabolism specifically in the subcutaneous white adipose tissue, through the regulation of target genes implicated in the insulin signaling cascade. Moreover, the expression of these miRNAs is modified by caloric restriction in middle-aged animals, in accordance with the improvement of the metabolic state. Overall, our work demonstrates that alterations in posttranscriptional gene expression because of miRNAs dysregulation might represent an endogenous mechanism by which insulin response in the subcutaneous fat depot is already affected at middle age. Importantly, caloric restriction could prevent this modulation, demonstrating that certain miRNAs could constitute potential biomarkers of age-related metabolic alterations.


Asunto(s)
Resistencia a la Insulina , MicroARNs , Animales , Masculino , Insulina/metabolismo , Restricción Calórica , Resistencia a la Insulina/genética , MicroARNs/genética , MicroARNs/metabolismo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo/metabolismo , Envejecimiento/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-37156296

RESUMEN

Podocytes are specialized epithelial cells that maintain the glomerular filtration barrier. These cells are susceptible to lipotoxicity in the obese state and irreversibly lost during kidney disease leading to proteinuria and renal injury. PPARγ is a nuclear receptor whose activation can be renoprotective. This study examined the role of PPARγ in the lipotoxic podocyte using a PPARγ knockout (PPARγKO) cell line and since the activation of PPARγ by Thiazolidinediones (TZD) is limited by their side effects, it explored other alternative therapies to prevent podocyte lipotoxic damage. Wild-type and PPARγKO podocytes were exposed to the fatty acid palmitic acid (PA) and treated with the TZD (Pioglitazone) and/or the Retinoid X receptor (RXR) agonist Bexarotene (BX). It revealed that podocyte PPARγ is essential for podocyte function. PPARγ deletion reduced key podocyte proteins including podocin and nephrin while increasing basal levels of oxidative and ER stress causing apoptosis and cell death. A combination therapy of low-dose TZD and BX activated both the PPARγ and RXR receptors reducing PA-induced podocyte damage. This study confirms the crucial role of PPARγ in podocyte biology and that their activation in combination therapy of TZD and BX may be beneficial in the treatment of obesity-related kidney disease.


Asunto(s)
Enfermedades Renales , Podocitos , Tiazolidinedionas , Humanos , PPAR gamma/metabolismo , Pioglitazona/farmacología , Tiazolidinedionas/metabolismo , Tiazolidinedionas/farmacología , Tiazolidinedionas/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Bexaroteno/farmacología
4.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166296, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34718120

RESUMEN

Bisphenol-A (BPA), a chemical -xenoestrogen- used in the production of the plastic lining of food and beverage containers, is present in the urine of almost the entire population. Recent studies have shown that BPA exposure is associated with podocytopathy, increased urinary albumin excretion (UAE), and hypertension. Since these changes are characteristic of early diabetic nephropathy (DN), we explored the renal effects of BPA and diabetes including the potential role of sexual dimorphism. Male and female mice were included in the following animals' groups: control mice (C), mice treated with 21.2 mg/kg of BPA in the drinking water (BPA), diabetic mice induced by streptozotocin (D), and D mice treated with BPA (D + BPA). Male mice form the D + BPA group died by the tenth week of the study due probably to hydro-electrolytic disturbances. Although BPA treated mice did not show an increase in serum creatinine, as observed in D and D + BPA groups, they displayed similar alteration to those of the D group, including increased in kidney damage biomarkers NGAL and KIM-1, UAE, hypertension, podocytopenia, apoptosis, collapsed glomeruli, as well as TGF-ß, CHOP and PCNA upregulation. UAE, collapsed glomeruli, PCNA staining, TGF-ß, NGAL and animal survival, significantly impaired in D + BPA animals. Moreover, UAE, collapsed glomeruli and animal survival also displayed a sexual dimorphism pattern. In conclusion, oral administration of BPA is capable of promoting in the kidney alterations that resemble early DN. Further translational studies are needed to clarify the potential role of BPA in renal diseases, particularly in diabetic patients.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/genética , Riñón/efectos de los fármacos , Fenoles/toxicidad , Animales , Apoptosis/efectos de los fármacos , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/inducido químicamente , Nefropatías Diabéticas/patología , Femenino , Receptor Celular 1 del Virus de la Hepatitis A/genética , Humanos , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/patología , Riñón/patología , Lipocalina 2/genética , Masculino , Ratones , Caracteres Sexuales
5.
Biomolecules ; 11(11)2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34827568

RESUMEN

Adipose tissue is an organ that shows great plasticity and is able to adapt to the conditions to which the body is subdued. It participates in the regulation of energetic homeostasis and has endocrine functions. Recent studies have shown how the parathyroid hormone-related protein (PTHrP)/Parathyroid Hormone Receptor 1 (PTH1R) axis participates in the regulation of adipogenesis, opposing the action of Peroxisome proliferator-activated receptor gamma (PPARγ). In addition to this, PTHrP is overexpressed in adipose tissue in situations of wear and tear of the body, favoring browning and lipolysis in this tissue. It is also overexpressed in adipose tissue in stressful situations but in the opposite direction, in obesity, metabolic syndrome, type 2 diabetes mellitus (T2DM) and gestational diabetes mellitus (GDM). In conclusion, the PTHrP/PTH1R axis has a main role in adipose tissue, participating in its differentiation and remodeling. PTHrP might be used in obesity treatment and its complications for its ability to reprogram adipogenesis and adipose tissue expansion, WAT browning and for the improvement of the insulin sensitivity. In addition, PTHrP could even be used as a marker of placental status and maternal adaptations to prevent future metabolic problems in mothers and children, as well as in the treatment of bone-related diseases such as osteoporosis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Femenino , Humanos , Proteína Relacionada con la Hormona Paratiroidea , Embarazo
6.
Dis Model Mech ; 14(9)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34431499

RESUMEN

Glomerulosclerosis and tubulointerstitial fibrosis are pathological features of chronic kidney disease. Transforming growth factor ß (TGFß) is a key player in the development of fibrosis. However, of the three known TGFß isoforms, only TGFß1 has an established role in fibrosis, and the pathophysiological relevance of TGFß2 and TGFß3 is unknown. Because Tgfb3 deficiency in mice results in early postnatal lethality, we analyzed the kidney phenotype of heterozygous Tgfb3-knockout mice (Tgfb3+/-) and compared it with that of matched wild-type mice. Four-month-old Tgfb3+/- mice exhibited incipient renal fibrosis with epithelial-mesenchymal transition, in addition to glomerular basement membrane thickening and podocyte foot process effacement associated with albuminuria. Also evident was insulin resistance and oxidative stress at the renal level, together with aberrant renal lipid metabolism and mitochondrial function. Omics analysis revealed toxic species, such as diacylglycerides and ceramides, and dysregulated mitochondrial metabolism in Tgfb3+/- mice. Kidneys of Tgfb3+/- mice showed morphological alterations of mitochondria and overactivation of non-canonical MAPK ERK1/2 and JNK cascades. Our study indicates that renal TGFß3 might have antifibrotic and renoprotective properties, opposing or counteracting the activity of TGFß1. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Metabolismo de los Lípidos , Factor de Crecimiento Transformador beta3/metabolismo , Animales , Fibrosis , Riñón/metabolismo , Ratones , Ratones Noqueados , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
7.
Nephron ; 145(6): 604-608, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33849028

RESUMEN

There is a strong relationship between the kidney and the heart, where if one of these organs fails, so does the other, in the so-called cardiorenal syndrome (CRS). Besides, there are also interactions with the rest of the body leading to a metabolic state that establishes a feedback loop that is perpetuated. The CRS is characterized by hemodynamic changes, activation of neuro-humoral systems, natriuretic peptides, and changes in mineral metabolism. In this scenario, the kidney and heart, connected by a dysfunctional endothelium, inevitably fail. In obesity, this syndrome is exacerbated due to the complications of adipose tissue dysfunction, in the so-called cardiorenal metabolic syndrome (CRMetS). Obesity promotes adipose tissue dysfunction because it exceeds lipid storage capacity and leads to a lipotoxic state, characterized by inflammation, hypertension, insulin resistance and dyslipidemia, oxidative stress, and hyperuricemia, among others, that affect different organs other than the adipose tissue. In addition, the pro-inflammatory gut microbiota present in obese patients releases uremic toxins, contributing to oxidative stress and inflammation, perpetuating and accelerating the progression of this pathology. In this article, we describe the contribution of obesity, the factors and mechanisms implicated in the development of the CRMetS. Despite the great knowledge about the CRS, more research is needed to characterize the CRMetS given the global obesity epidemic.


Asunto(s)
Síndrome Cardiorrenal/complicaciones , Corazón/fisiopatología , Riñón/fisiopatología , Obesidad/fisiopatología , Síndrome Cardiorrenal/fisiopatología , Humanos
8.
Sci Rep ; 11(1): 7717, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33833312

RESUMEN

When exposed to nutrient excess and insulin resistance, pancreatic ß-cells undergo adaptive changes in order to maintain glucose homeostasis. The role that growth control genes, highly expressed in early pancreas development, might exert in programming ß-cell plasticity in later life is a poorly studied area. The imprinted Igf2 (insulin-like growth factor 2) gene is highly transcribed during early life and has been identified in recent genome-wide association studies as a type 2 diabetes susceptibility gene in humans. Hence, here we investigate the long-term phenotypic metabolic consequences of conditional Igf2 deletion in pancreatic ß-cells (Igf2ßKO) in mice. We show that autocrine actions of IGF2 are not critical for ß-cell development, or for the early post-natal wave of ß-cell remodelling. Additionally, adult Igf2ßKO mice maintain glucose homeostasis when fed a chow diet. However, pregnant Igf2ßKO females become hyperglycemic and hyperinsulinemic, and their conceptuses exhibit hyperinsulinemia and placentomegalia. Insulin resistance induced by congenital leptin deficiency also renders Igf2ßKO females more hyperglycaemic compared to leptin-deficient controls. Upon high-fat diet feeding, Igf2ßKO females are less susceptible to develop insulin resistance. Based on these findings, we conclude that in female mice, autocrine actions of ß-cell IGF2 during early development determine their adaptive capacity in adult life.


Asunto(s)
Plasticidad de la Célula/fisiología , Factor II del Crecimiento Similar a la Insulina/fisiología , Células Secretoras de Insulina/citología , Animales , Femenino , Glucosa/metabolismo , Homeostasis , Insulina/sangre , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Noqueados , Embarazo
9.
Aging Cell ; 18(3): e12948, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30920127

RESUMEN

Age-related increased adiposity is an important contributory factor in the development of insulin resistance (IR) and is associated with metabolic defects. Caloric restriction (CR) is known to induce weight loss and to decrease adiposity while preventing metabolic risk factors. Here, we show that moderate 20% CR delays early deleterious effects of aging on white and brown adipose tissue (WAT and BAT, respectively) function and improves peripheral IR. To elucidate the role of CR in delaying early signs of aging, young (3 months), middle-aged (12 months), and old (20 months) mice fed al libitum and middle-aged and old mice subjected to early-onset CR were used. We show that impaired plasticity of subcutaneous WAT (scWAT) contributes to IR, which is already evident in middle-aged mice. Moreover, alteration of thyroid axis status with age is an important factor contributing to BAT dysfunction in middle-aged animals. Both defects in WAT and BAT/beige cells are ameliorated by CR. Accordingly, CR attenuated the age-related decline in scWAT function and decreased the extent of fibro-inflammation. Furthermore, CR promoted scWAT browning. In brief, our study identifies the contribution of scWAT impairment to age-associated metabolic dysfunction and identifies browning in response to food restriction, as a potential therapeutic strategy to prevent the adverse metabolic effects in middle-aged animals.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Envejecimiento/metabolismo , Restricción Calórica , Animales , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones de la Cepa 129 , Tomografía Computarizada por Tomografía de Emisión de Positrones
10.
Nephron ; 143(1): 28-32, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30625473

RESUMEN

The recent and ongoing worldwide increase in the prevalence of obesity parallels the increase in the incidence of chronic kidney disease (CKD). This association suggests an implication of lipotoxicity in the development of kidney diseases. The increased influx of lipids into the kidney can be explained in the context of the "Adipose Tissue Expandability Hypothesis". This hypothesis states that the adipose tissue has a limited expansion capability, which is different for each individual, and once this limit is reached, the adipose tissue cannot store any more lipids and will thus release them into the bloodstream. The accumulation of lipids in the kidney is known as renal lipotoxicity. Renal lipotoxicity is known to cause detrimental effects on the kidney by several mechanisms of action including reclusion of pro-inflammatory factors, oxidative and ER stress development, insulin resistance (IR), lipid metabolism deregulation or renin-angiotensin aldosterone system overactivation. Isoform peroxisome proliferator-activated receptor gamma (PPARγ) seems to play an important role in the development of this lipotoxicity as proven by several studies in -animals and cultured cells. Thus, PPARγ agonists are of -interest in the therapeutic approach to treat CKD in the context of obesity. This review aims to summarize our current knowledge of the mechanism by which lipotoxicity affects renal structure and function using in vivo and in vitro models as examples focusing on PPARγ.


Asunto(s)
Riñón/metabolismo , Metabolismo de los Lípidos , Obesidad/complicaciones , Insuficiencia Renal Crónica/etiología , Animales , Estrés del Retículo Endoplásmico , Humanos , Resistencia a la Insulina , Obesidad/metabolismo , Estrés Oxidativo , PPAR gamma/agonistas , PPAR gamma/fisiología
11.
Int J Mol Sci ; 19(7)2018 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-30012954

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear hormone receptors that control the transcription of specific genes by binding to regulatory DNA sequences. Among the three subtypes of PPARs, PPARγ modulates a broad range of physiopathological processes, including lipid metabolism, insulin sensitization, cellular differentiation, and cancer. Although predominantly expressed in adipose tissue, PPARγ expression is also found in different regions of the kidney and, upon activation, can redirect metabolism. Recent studies have highlighted important roles for PPARγ in kidney metabolism, such as lipid and glucose metabolism and renal mineral control. PPARγ is also implicated in the renin-angiotensin-aldosterone system and, consequently, in the control of systemic blood pressure. Accordingly, synthetic agonists of PPARγ have reno-protective effects both in diabetic and nondiabetic patients. This review focuses on the role of PPARγ in renal metabolism as a likely key factor in the maintenance of systemic homeostasis.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Riñón/metabolismo , Metabolismo de los Lípidos , PPAR gamma/metabolismo , Animales , Humanos , Insulina/metabolismo , Riñón/efectos de los fármacos , Riñón/fisiopatología , PPAR gamma/agonistas , Sistema Renina-Angiotensina , Tiazolidinedionas/farmacología
12.
Mol Med ; 22: 724-736, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27782293

RESUMEN

Pregnancy requires the adaptation of maternal energy metabolism including expansion and functional modifications of adipose tissue. Insulin resistance (IR), predominantly during late gestation, is a physiological metabolic adaptation that serves to support the metabolic demands of fetal growth. The molecular mechanisms underlying these adaptations are not fully understood and may contribute to gestational diabetes mellitus. Peroxisome proliferator-activated receptor gamma (PPARγ) controls adipogenesis, glucose and lipid metabolism and insulin sensitivity. The PPARγ2 isoform is mainly expressed in adipocytes and is thus likely to contribute to adipose tissue adaptation during late pregnancy. In the present study, we investigated the contribution of PPARγ2 to the metabolic adaptations occurring during the late phase of pregnancy in the context of IR. Using a model of late pregnancy in PPARγ2 knockout (KO) mice, we found that deletion of PPARγ2 exacerbated IR in association with lower serum adiponectin levels, increased body weight and enhanced lipid accumulation in liver. Lack of PPARγ2 provoked changes in the distribution of fat mass and preferentially prevented the expansion of the perigonadal depot while at the same time exacerbating inflammation. PPARγ2KO pregnant mice presented adipose tissue depot-dependent decreased expression of genes involved in lipid metabolism. Collectively, these data indicate that PPARγ2 is essential to promote healthy adipose tissue expansion and immune and metabolic functionality during pregnancy, contributing to the physiological adaptations that lead gestation to term.

13.
Biochem Biophys Res Commun ; 478(3): 1043-8, 2016 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-27553274

RESUMEN

Alpha-dystroglycanopathies are a heterogenic group of human rare diseases that have in common defects of α-dystroglycan O-glycosylation. These congenital disorders share common features as muscular dystrophy, malformations on central nervous system and more rarely altered ocular development, as well as mutations on a set of candidate genes involved on those syndromes. Severity of the syndromes is variable, appearing Walker-Warburg as the most severe where mutations at protein O-mannosyl transferases POMT1 and POMT2 genes are frequently described. When studying the lack of MmPomt1 in mouse embryonic development, as a murine model of Walker-Warburg syndrome, MmPomt1 null phenotype was lethal because Reitchert's membrane fails during embryonic development. Here, we report gene expression from Gallus gallus orthologous genes to human candidates on alpha-dystroglycanopathies POMT1, POMT2, POMGnT1, FKTN, FKRP and LARGE, making special emphasis in expression and localization of GgPomt1. Results obtained by quantitative RT-PCR, western-blot and immunochemistry revealed close gene expression patterns among human and chicken at key tissues affected during development when suffering an alpha-dystroglycanopathy, leading us to stand chicken as a useful animal model for molecular characterization of glycosyltransferases involved in the O-glycosylation of α-Dystroglycan and its role in embryonic development.


Asunto(s)
Pollos/genética , Distroglicanos/metabolismo , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Estudios de Asociación Genética , Homología de Secuencia de Aminoácido , Animales , Humanos , Inmunohistoquímica , Médula Espinal/embriología , Médula Espinal/metabolismo
14.
J Nephrol ; 29(5): 603-10, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26956132

RESUMEN

In the last few decades, rapid changes in lifestyle have led to an alarming increase in the prevalence of obesity and obesity-associated complications. Obese patients are at increased risk of developing hypertension, heart disease, insulin resistance, dyslipidemia, type 2 diabetes and kidney disease. The surplus of calories is normally stored as triglycerides in adipose tissue. However, excess lipids can also accumulate ectopically in other organs, including the kidney, contributing to their damage through toxic processes named lipotoxicity. The kidney is negatively affected by dyslipidemia, lipid accumulation and changes in circulating adipokines that bring about alterations in renal lipid metabolism and promote insulin resistance, generation of reactive oxygen species and endoplasmic reticulum stress, ultimately leading to alterations in the glomerular filtration barrier and renal failure. This review focuses on the pathogenic molecular mechanisms associated with renal lipotoxicity, and presents new insights about potential new therapeutic targets and biomarkers such as microRNAs and long non-coding RNAs, of relevance for the early detection of lipid-associated kidney disease.


Asunto(s)
Tejido Adiposo/metabolismo , Grasas de la Dieta/efectos adversos , Dislipidemias/metabolismo , Metabolismo Energético , Riñón/metabolismo , Obesidad/metabolismo , Insuficiencia Renal/metabolismo , Adipoquinas/metabolismo , Tejido Adiposo/fisiopatología , Animales , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/metabolismo , Dislipidemias/epidemiología , Dislipidemias/genética , Dislipidemias/fisiopatología , Estrés del Retículo Endoplásmico , Ingestión de Energía , Marcadores Genéticos , Tasa de Filtración Glomerular , Humanos , Resistencia a la Insulina , Riñón/fisiopatología , MicroARNs/genética , Obesidad/epidemiología , Obesidad/genética , Obesidad/fisiopatología , ARN Largo no Codificante/genética , Especies Reactivas de Oxígeno/metabolismo , Insuficiencia Renal/epidemiología , Insuficiencia Renal/genética , Insuficiencia Renal/fisiopatología , Medición de Riesgo , Factores de Riesgo , Transducción de Señal
15.
PLoS One ; 10(11): e0142291, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26545114

RESUMEN

In the last few decades a change in lifestyle has led to an alarming increase in the prevalence of obesity and obesity-associated complications. Obese patients are at increased risk of developing hypertension, heart disease, insulin resistance (IR), dyslipidemia, type 2 diabetes and renal disease. The excess calories are stored as triglycerides in adipose tissue, but also may accumulate ectopically in other organs, including the kidney, which contributes to the damage through a toxic process named lipotoxicity. Recently, the evidence suggests that renal lipid accumulation leads to glomerular damage and, more specifically, produces dysfunction in podocytes, key cells that compose and maintain the glomerular filtration barrier. Our aim was to analyze the early mechanisms underlying the development of renal disease associated with the process of lipotoxicity in podocytes. Our results show that treatment of podocytes with palmitic acid produced intracellular accumulation of lipid droplets and abnormal glucose and lipid metabolism. This was accompanied by the development of inflammation, oxidative stress and endoplasmic reticulum stress and insulin resistance. We found specific rearrangements of the actin cytoskeleton and slit diaphragm proteins (Nephrin, P-Cadherin, Vimentin) associated with this insulin resistance in palmitic-treated podocytes. We conclude that lipotoxicity accelerates glomerular disease through lipid accumulation and inflammation. Moreover, saturated fatty acids specifically promote insulin resistance by disturbing the cytoarchitecture of podocytes. These data suggest that renal lipid metabolism and cytoskeleton rearrangements may serve as a target for specific therapies aimed at slowing the progression of podocyte failure during metabolic syndrome.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Resistencia a la Insulina , Riñón/metabolismo , Metabolismo de los Lípidos , Podocitos/metabolismo , Citoesqueleto de Actina/química , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Citocalasina D/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inflamación/metabolismo , Ratones , Estrés Oxidativo , Ácido Palmítico/metabolismo , Podocitos/efectos de los fármacos
16.
Nephrol Dial Transplant ; 20(7): 1320-8, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15840669

RESUMEN

BACKGROUND: The podocyte is bathed in an angiotensin II (AngII)-rich ultrafiltrate, but the impact of AngII on podocyte pathobiology is not well known. Because podocytes play a direct role in the glomerular basement membrane (GBM) thickening of diabetes, the alpha3(IV) collagen chain was examined. Podocyte expression of alpha3(IV) collagen may involve the transforming growth factor-beta (TGF-beta) and vascular endothelial growth factor (VEGF) systems. METHODS: Cultured mouse podocytes were treated with various doses of AngII for selected periods of time, with or without inhibitors of TGF-beta and VEGF signalling, SB-431542 and SU5416, respectively. TGF-beta1 and VEGF were assayed by enzyme-linked immunosorbent assay (ELISA); alpha3(IV) collagen, TGF-beta type II receptor and phospho-Smad2 were assayed by immunoblotting. RESULTS: AngII >or=10(-10) M was found to stimulate the production of alpha3(IV) collagen significantly in as short a time as 3 h. The expression of alpha3(IV) collagen was influenced by the TGF-beta system, but AngII did not increase the podocyte's production of TGF-beta1 ligand; rather, it increased the expression of the TGF-beta type II receptor and activated the TGF-beta signalling system through Smad2. Despite the TGF-beta receptor upregulation, synergy between AngII and TGF-beta1 to boost alpha3(IV) collagen production was not observed. However, blockade of TGF-beta signalling with SB-431542 prevented AngII from stimulating alpha3(IV) collagen production. Podocyte expression of alpha3(IV) collagen was also increased by the autocrine activity of VEGF. Podocytes were stimulated to secrete VEGF by 10(-10) M or higher AngII after 48 h. Blockade of the endogenous VEGF activity by SU5416 prevented AngII-stimulated alpha3(IV) collagen production. CONCLUSIONS: AngII stimulates the podocyte to produce alpha3(IV) collagen protein via mechanisms involving TGF-beta and VEGF signalling. Alterations in alpha3(IV) collagen production may contribute to GBM thickening and perhaps proteinuria in diabetes.


Asunto(s)
Angiotensina II/farmacología , Autoantígenos/efectos de los fármacos , Colágeno Tipo IV/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Glomérulos Renales/efectos de los fármacos , Vasoconstrictores/farmacología , Angiotensina II/administración & dosificación , Animales , Autoantígenos/biosíntesis , Técnicas de Cultivo de Célula , Colágeno Tipo IV/biosíntesis , Relación Dosis-Respuesta a Droga , Células Epiteliales/metabolismo , Glomérulos Renales/citología , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Tiempo , Factor de Crecimiento Transformador beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/fisiología , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/fisiología , Vasoconstrictores/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA