Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Front Med (Lausanne) ; 8: 679518, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34336885

RESUMEN

The molecular pathogenesis of diabetic kidney disease progression is complex and remains unresolved. Rho-GAP MYO9A was recently identified as a novel podocyte protein and a candidate gene for monogenic FSGS. Myo9A involvement in diabetic kidney disease has been suggested. Here, we examined the effect of diabetic milieu on Myo9A expression in vivo and in vitro. We determined that Myo9A undergoes S-nitrosylation, a post-translational modification dependent on nitric oxide (NO) availability. Diabetic mice with nodular glomerulosclerosis and severe proteinuria associated with doxycycline-induced, podocyte-specific VEGF 164 gain-of-function showed markedly decreased glomerular Myo9A expression and S-nitrosylation, as compared to uninduced diabetic mice. Immortalized mouse podocytes exposed to high glucose revealed decreased Myo9A expression, assessed by qPCR, immunoblot and immunocytochemistry, and reduced Myo9A S-nitrosylation (SNO-Myo9A), assessed by proximity link assay and biotin switch test, functionally resulting in abnormal podocyte migration. These defects were abrogated by exposure to a NO donor and were not due to hyperosmolarity. Our data demonstrate that high-glucose induced decrease of both Myo9A expression and SNO-Myo9A is regulated by NO availability. We detected S-nitrosylation of Myo9A interacting proteins RhoA and actin, which was also altered by high glucose and NO dependent. RhoA activity inversely related to SNO-RhoA. Collectively, data suggest that dysregulation of SNO-Myo9A, SNO-RhoA and SNO-actin may contribute to the pathogenesis of advanced diabetic kidney disease and may be amenable to therapeutic targeting.

2.
Front Pharmacol ; 12: 788886, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35280251

RESUMEN

Vascular endothelial growth factor-a (VEGF-A) and nitric oxide (NO) are essential for glomerular filtration barrier homeostasis, and are dysregulated in diabetic kidney disease (DKD). While NO availability is consistently low in diabetes, both high and low VEGF-A have been reported in patients with DKD. Here we examined the effect of inducible podocyte VEGF-A knockdown (VEGFKD ) in diabetic mice and in endothelial nitric oxide synthase knockout mice (eNOS-/- ). Diabetes was induced with streptozotocin using the Animal Models of Diabetic Complications Consortium (AMDCC) protocol. Induction of podocyte VEGFKD led to diffuse glomerulosclerosis, foot process effacement, and GBM thickening in both diabetic mice with intact eNOS and in non-diabetic eNOS-/-:VEGFKD mice. VEGFKD diabetic mice developed mild proteinuria and maintained normal glomerular filtration rate (GFR), associated with extremely high NO and thiol urinary excretion. In eNOS-/-:VEGFKD (+dox) mice severe diffuse glomerulosclerosis was associated with microaneurisms, arteriolar hyalinosis, massive proteinuria, and renal failure. Collectively, data indicate that combined podocyte VEGF-A and eNOS deficiency result in diffuse glomerulosclerosis in mice; compensatory NO and thiol generation prevents severe proteinuria and GFR loss in VEGFKD diabetic mice with intact eNOS, whereas VEGFKD induction in eNOS-/-:VEGFKD mice causes massive proteinuria and renal failure mimicking DKD in the absence of diabetes. Mechanistically, we identify VEGFKD -induced abnormal S-nitrosylation of specific proteins, including ß3-integrin, laminin, and S-nitrosoglutathione reductase (GSNOR), as targetable molecular mechanisms involved in the development of advanced diffuse glomerulosclerosis and renal failure.

6.
Nephrol Dial Transplant ; 33(9): 1514-1525, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29635428

RESUMEN

Background: Vascular endothelial growth factor A (VEGF) is an essential growth factor during glomerular development and postnatal homeostasis. VEGF is secreted in high amounts by podocytes into the primary urine, back-filtered across the glomerular capillary wall to act on endothelial cells. So far it has been assumed that VEGF back-filtration is driven at a constant rate exclusively by diffusion. Methods: In the present work, glomerular VEGF back-filtration was investigated in vivo using a novel extended model based on endothelial fenestrations as surrogate marker for local VEGF concentrations. Single nephron glomerular filtration rate (SNGFR) and/or local filtration flux were manipulated by partial renal mass ablation, tubular ablation, and in transgenic mouse models of systemic or podocytic VEGF overexpression or reduction. Results: Our study shows positive correlations between VEGF back-filtration and SNGFR as well as effective filtration rate under physiological conditions along individual glomerular capillaries in rodents and humans. Conclusion: Our results suggest that an additional force drives VEGF back-filtration, potentially regulated by SNGFR.


Asunto(s)
Capilares/fisiopatología , Tasa de Filtración Glomerular/fisiología , Glomérulos Renales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Modelos Animales de Enfermedad , Glomérulos Renales/fisiopatología , Ratones , Ratones Noqueados , Nefrectomía
9.
Nefrologia ; 35(2): 131-8, 2015.
Artículo en Inglés, Español | MEDLINE | ID: mdl-26300505

RESUMEN

The prevalence of diabetes mellitus increased during the last century and it is estimated that 45% of the patients are not diagnosed. In South America the prevalence of diabetes and chronic kidney disease (CKD) increased, with a great disparity among the countries with respect to access to dialysis. In Ecuador it is one of the main causes of mortality, principally in the provinces located on the coast of the Pacific Ocean. The greatest single cause of beginning dialysis is diabetic nephropathy (DN). Even using the best therapeutic options for DN, the residual risk of proteinuria and of terminal CKD remains high. In this review we indicate the importance of the problem globally and in our region. We analyse relevant cellular and molecular studies that illustrate the crucial significance of glomerular events in DN development and evolution and in insulin resistance. We include basic anatomical, pathophysiological and clinical concepts, with special attention to the role of angiogenic factors such as the vascular endothelial growth factor (VEGF-A) and their relationship to the insulin receptor, endothelial isoform of nitric oxide synthase (eNOS) and angiopoietins. We also propose various pathways that have therapeutic potential in our opinion. Greater in-depth study of VEGF-A and angiopoietins, the state of glomerular VEGF resistance, the relationship of VEGF receptor 2/nephrin, VEGF/insulin receptors/nephrin and the relationship of VEGF/eNOS-NO at glomerular level could provide solutions to the pressing world problem of DN and generate new treatment alternatives.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología , Angiopoyetinas/fisiología , Nefropatías Diabéticas/epidemiología , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Salud Global , Humanos , Resistencia a la Insulina , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Proteínas de la Membrana/fisiología , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Receptor de Insulina/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Transducción de Señal
10.
Nefrología (Madr.) ; 35(2): 131-138, mar.-abr. 2015. ilus
Artículo en Español | IBECS | ID: ibc-139278

RESUMEN

La prevalencia de diabetes mellitus aumentó en el último siglo y se estima que el 45%de los pacientes, no estarían diagnosticados. En Sudamérica la prevalencia de diabetes y de enfermedad renal crónica (ERC) incrementó, existiendo gran disparidad entre los países respecto al acceso a diálisis. En Ecuador es una de las principales causas de mortalidad, principalmente en las provincias ubicadas en la costa del océano Pacífico. La mayor causa aislada de ingreso a diálisis es la nefropatía diabética (ND). Aun utilizando las mejores opciones terapéuticas para la ND, el riesgo residual de proteinuria y de ERC terminal permanece elevado. En esta revisión describimos la importancia del problema en el mundo y en nuestra región Analizamos estudios moleculares y celulares relevantes que indican la crucial importancia de eventos glomerulares en el desarrollo y en la evolución de la ND y en la insulinorresistencia. Incluimos conceptos anatómicos, fisiopatológicos y clínicos básicos, desarrollando especial énfasis en el rol de factores angiogénicos como el factor de crecimiento vascular endotelial(VEGF-A) y su relación con el receptor de insulina, la sintasa endotelial de óxido nítrico-óxidonítrico (eNOS) y las angiopoietinas. En el transcurso del texto proponemos diversas vías, que a nuestro entender tienen potencial terapéutico. Profundizar en el estudio del VEGF-A y la sangiopoietinas, el estado de VEGF resistencia glomerular, la relación del receptor 2 de VEGF/nefrina, VEGF/receptores de insulina/nefrina, la relación VEGF/eNOS-ON a nivel glomerular podría aportar soluciones al acuciante problema de la ND en el mundo y generar nuevas alternativas de tratamiento (AU)


The prevalence of diabetes mellitus increased during the last century and it is estimated that45% of the patients are not diagnosed. In South America the prevalence of diabetes and chronic kidney disease (CKD) increased, with a great disparity among the countries with respect to access to dialysis. In Ecuador it is one of the main causes of mortality, principally in the provinces located on the coast of the Pacific Ocean. The greatest single cause of beginning dialysis is diabetic nephropathy (DN). Even using the best therapeutic options for DN, the residual risk of proteinuria and of terminal CKD remains high. In this review we indicate the importance of the problem globally and in our region. We analyse relevant cellular and molecular studies that illustrate the crucial significance of glomerular events in DN development and evolution and in insulin resistance. We include basic anatomical, pathophysiological and clinical concepts, with special attention to the role of angiogenic factors such as the vascular endothelial growth factor (VEGF-A) and their relationship to the insulin receptor, endothelial isoform of nitric oxide synthase (eNOS) and angiopoietins. We also propose various pathways that have therapeutic potential in our opinion. Greater in-depth study of VEGF-A and angiopoietins, the state of glomerular VEGF resistance, the relationship of VEGF receptor 2/nephrin, VEGF/insulin receptors/nephrin and the relationship of VEGF/eNOS-NO at glomerular level could provide solutions to the pressing world problem of DN and generate new treatment alternatives (AU)


Asunto(s)
Humanos , Nefropatías Diabéticas/fisiopatología , Receptores de Factores de Crecimiento Endotelial Vascular/fisiología , Receptor de Insulina/fisiología , Angiopoyetinas/fisiología , Podocitos/fisiología , Resistencia a la Insulina/fisiología , Óxido Nítrico/fisiología , Tasa de Filtración Glomerular/fisiología
11.
Diabetes ; 64(5): 1743-59, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25475434

RESUMEN

The onset of diabetic nephropathy (DN) is highlighted by glomerular filtration barrier abnormalities. Identifying pathogenic factors and targetable pathways driving DN is crucial to developing novel therapies and improving the disease outcome. Semaphorin3a (sema3a) is a guidance protein secreted by podocytes. Excess sema3a disrupts the glomerular filtration barrier. Here, using immunohistochemistry, we show increased podocyte SEMA3A in renal biopsies from patients with advanced DN. Using inducible, podocyte-specific Sema3a gain-of-function (Sema3a(+)) mice made diabetic with streptozotocin, we demonstrate that sema3a is pathogenic in DN. Diabetic Sema3a(+) mice develop massive proteinuria, renal insufficiency, and extensive nodular glomerulosclerosis, mimicking advanced DN in humans. In diabetic mice, Sema3a(+) exacerbates laminin and collagen IV accumulation in Kimmelstiel-Wilson-like glomerular nodules and causes diffuse podocyte foot process effacement and F-actin collapse via nephrin, αvß3 integrin, and MICAL1 interactions with plexinA1. MICAL1 knockdown and sema3a inhibition render podocytes not susceptible to sema3a-induced shape changes, indicating that MICAL1 mediates sema3a-induced podocyte F-actin collapse. Moreover, sema3a binding inhibition or podocyte-specific plexinA1 deletion markedly ameliorates albuminuria and abrogates renal insufficiency and the diabetic nodular glomerulosclerosis phenotype of diabetic Sema3a(+) mice. Collectively, these findings indicate that excess sema3a promotes severe diabetic nephropathy and identifies novel potential therapeutic targets for DN.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Podocitos/metabolismo , Semaforina-3A/metabolismo , Actinas/metabolismo , Animales , Cromonas , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Diabetes Mellitus Experimental , Nefropatías Diabéticas/patología , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Laminina/genética , Laminina/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteínas de Microfilamentos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteinuria/etiología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Insuficiencia Renal , Semaforina-3A/sangre , Semaforina-3A/genética , Semaforina-3A/orina , Proteínas WT1/genética , Proteínas WT1/metabolismo , Xantonas
12.
J Am Soc Nephrol ; 25(8): 1814-24, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24578128

RESUMEN

VEGF-A and nitric oxide are essential for glomerular filtration barrier homeostasis and are dysregulated in diabetic nephropathy. Here, we examined the effect of excess podocyte VEGF-A on the renal phenotype of endothelial nitric oxide synthase (eNOS) knockout mice. Podocyte-specific VEGF(164) gain of function in eNOS(-/-) mice resulted in nodular glomerulosclerosis, mesangiolysis, microaneurysms, and arteriolar hyalinosis associated with massive proteinuria and renal failure in the absence of diabetic milieu or hypertension. In contrast, podocyte-specific VEGF(164) gain of function in wild-type mice resulted in less pronounced albuminuria and increased creatinine clearance. Transmission electron microscopy revealed glomerular basement membrane thickening and podocyte effacement in eNOS(-/-) mice with podocyte-specific VEGF(164) gain of function. Furthermore, glomerular nodules overexpressed collagen IV and laminin extensively. Biotin-switch and proximity ligation assays demonstrated that podocyte-specific VEGF(164) gain of function decreased glomerular S-nitrosylation of laminin in eNOS(-/-) mice. In addition, treatment with VEGF-A decreased S-nitrosylated laminin in cultured podocytes. Collectively, these data indicate that excess glomerular VEGF-A and eNOS deficiency is necessary and sufficient to induce Kimmelstiel-Wilson-like nodular glomerulosclerosis in mice through a process that involves deposition of laminin and collagen IV and de-nitrosylation of laminin.


Asunto(s)
Nefropatías Diabéticas/etiología , Óxido Nítrico Sintasa de Tipo III/fisiología , Podocitos/metabolismo , Proteinuria/etiología , Insuficiencia Renal/etiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Técnicas de Cultivo de Célula , Colágeno Tipo IV/metabolismo , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Membrana Basal Glomerular/metabolismo , Membrana Basal Glomerular/patología , Laminina/metabolismo , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo III/genética , Proteinuria/metabolismo , Proteinuria/patología , Insuficiencia Renal/metabolismo , Insuficiencia Renal/patología
13.
Am J Pathol ; 183(4): 1156-1168, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23954273

RESUMEN

Semaphorin-3A (Sema3a), a guidance protein secreted by podocytes, is essential for normal kidney patterning and glomerular filtration barrier development. Here, we report that podocyte-specific Sema3a gain-of-function in adult mice leads to proteinuric glomerular disease involving the three layers of the glomerular filtration barrier. Reversibility of the glomerular phenotype upon removal of the transgene induction provided proof-of-principle of the cause-and-effect relationship between podocyte Sema3a excess and glomerular disease. Mechanistically, excess Sema3a induces dysregulation of nephrin, matrix metalloproteinase 9, and αvß3 integrin in vivo. Sema3a cell-autonomously disrupts podocyte shape. We identified a novel direct interaction between the Sema3a signaling receptor plexinA1 and nephrin, linking extracellular Sema3a signals to the slit-diaphragm signaling complex. We conclude that Sema3a functions as an extracellular negative regulator of the structure and function of the glomerular filtration barrier in the adult kidney. Our findings demonstrate a crosstalk between Sema3a and nephrin signaling pathways that is functionally relevant both in vivo and in vitro.


Asunto(s)
Enfermedades Renales/patología , Glomérulos Renales/patología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Podocitos/metabolismo , Podocitos/patología , Receptores de Superficie Celular/metabolismo , Semaforina-3A/metabolismo , Animales , Forma de la Célula , Regulación hacia Abajo , Membrana Basal Glomerular/metabolismo , Membrana Basal Glomerular/patología , Membrana Basal Glomerular/ultraestructura , Integrina alfaVbeta3/metabolismo , Enfermedades Renales/metabolismo , Glomérulos Renales/metabolismo , Glomérulos Renales/ultraestructura , Ratones , Podocitos/ultraestructura , Unión Proteica , Proteinuria/metabolismo , Proteinuria/patología , Transducción de Señal
14.
Semin Nephrol ; 32(4): 385-93, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22958493

RESUMEN

Vascular endothelial growth factor-A (VEGF-A) is a protein secreted by podocytes that is necessary for survival of endothelial cells, podocytes, and mesangial cells. VEGF-A regulates slit-diaphragm signaling and podocyte shape via VEGF-receptor 2-nephrin-nck-actin interactions. Chronic hyperglycemia-induced excess podocyte VEGF-A and low endothelial nitric oxide drive the development and the progression of diabetic nephropathy. The abnormal cross-talk between VEGF-A and nitric oxide pathways is fueled by the diabetic milieu, resulting in increased oxidative stress. Recent findings on these pathogenic molecular mechanisms provide new potential targets for therapy for diabetic renal disease.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Podocitos/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Supervivencia Celular , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Óxidos de Nitrógeno/metabolismo , Podocitos/patología , Sistema Renina-Angiotensina
15.
PLoS One ; 7(7): e40589, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22808199

RESUMEN

Podocyte or endothelial cell VEGF-A knockout causes thrombotic microangiopathy in adult mice. To study the mechanism involved in acute and local injury caused by low podocyte VEGF-A we developed an inducible, podocyte-specific VEGF-A knockdown mouse, and we generated an immortalized podocyte cell line (VEGF(KD)) that downregulates VEGF-A upon doxycycline exposure. Tet-O-siVEGF:podocin-rtTA mice express VEGF shRNA in podocytes in a doxycycline-regulated manner, decreasing VEGF-A mRNA and VEGF-A protein levels in isolated glomeruli to ~20% of non-induced controls and urine VEGF-A to ~30% of control values a week after doxycycline induction. Induced tet-O-siVEGF:podocin-rtTA mice developed acute renal failure and proteinuria, associated with mesangiolysis and microaneurisms. Glomerular ultrastructure revealed endothelial cell swelling, GBM lamination and podocyte effacement. VEGF knockdown decreased podocyte fibronectin and glomerular endothelial alpha(V)beta(3) integrin in vivo. VEGF receptor-2 (VEGFR2) interacts with beta(3) integrin and neuropilin-1 in the kidney in vivo and in VEGF(KD) podocytes. Podocyte VEGF knockdown disrupts alpha(V)beta(3) integrin activation in glomeruli, detected by WOW1-Fab. VEGF silencing in cultured VEGF(KD) podocytes downregulates fibronectin and disrupts alpha(V)beta(3) integrin activation cell-autonomously. Collectively, these studies indicate that podocyte VEGF-A regulates alpha(V)beta(3) integrin signaling in the glomerulus, and that podocyte VEGF knockdown disrupts alpha(V)beta(3) integrin activity via decreased VEGFR2 signaling, thereby damaging the three layers of the glomerular filtration barrier, causing proteinuria and acute renal failure.


Asunto(s)
Técnicas de Silenciamiento del Gen , Integrina alfaVbeta3/metabolismo , Podocitos/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Doxiciclina/farmacología , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Fibronectinas/metabolismo , Ratones , Modelos Animales , Neuropilina-1/metabolismo , Fenotipo , Podocitos/efectos de los fármacos , Podocitos/patología , Unión Proteica/efectos de los fármacos , Proteinuria/complicaciones , Proteinuria/metabolismo , Proteinuria/patología , Proteinuria/fisiopatología , ARN Interferente Pequeño/metabolismo , Insuficiencia Renal/complicaciones , Insuficiencia Renal/metabolismo , Insuficiencia Renal/patología , Insuficiencia Renal/fisiopatología , Transducción de Señal/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
J Biol Chem ; 286(46): 39933-44, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21937443

RESUMEN

The transmembrane protein nephrin is an essential component of slit diaphragms, the specialized cell junctions that link podocyte foot processes. Podocytes are epithelial cells that surround the glomerular capillaries in the kidney and are necessary for the organ-filtering function. Nephrin signaling complex transduces extracellular cues to the podocyte cytoskeleton and regulates podocyte shape and function. Vascular endothelial growth factor A (VEGF-A) is a required growth factor produced and secreted by podocytes. Accumulating evidence suggests a cross-talk between VEGF-A and nephrin signaling pathways. We previously showed that in vivo nephrin associates with VEGF receptor-2 (VEGFR2), the signaling receptor for VEGF-A. In the present work, we characterized the interaction between nephrin and VEGFR2 in cultured cells and in vitro. We demonstrate that nephrin-VEGFR2 interaction is direct using mass spectrometry, immunoprecipitation, GST-binding assays, and blot overlay experiments. This interaction occurs through VEGFR2 and nephrin cytoplasmic domains. Nephrin-VEGFR2 interaction is modulated by tyrosine phosphorylation of both cytoplasmic domains. Furthermore, the nephrin-VEGFR2 complex involves Nck and actin. VEGF-A signaling via this complex results in decreased cell size. We provide evidence that this multiprotein interaction occurs in cultured podocytes. We propose that the nephrin-VEGFR2 complex acts as a key mediator to transduce local VEGF-A signals to the podocyte actin cytoskeleton, regulating the foot process structure and glomerular filter integrity.


Asunto(s)
Actinas/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/metabolismo , Podocitos/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Actinas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células COS , Chlorocebus aethiops , Espectrometría de Masas , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Complejos Multiproteicos/genética , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Fosforilación/fisiología , Podocitos/citología , Estructura Terciaria de Proteína , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
17.
Am J Pathol ; 177(5): 2225-33, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20829436

RESUMEN

The tight regulation of vascular endothelial growth factor-A (VEGF-A) signaling is required for both the development and maintenance of the glomerular filtration barrier, but the pathogenic role of excessive amounts of VEGF-A detected in multiple renal diseases remains poorly defined. We generated inducible transgenic mice that overexpress podocyte VEGF164 at any chosen stage of development. In this study, we report the phenotypes that result from podocyte VEGF164 excess during organogenesis and after birth. On doxycycline induction, podocin-rtTA:tet-O-VEGF164 mice express twofold higher kidney VEGF164 levels than single transgenic mice, localized to podocytes. Podocyte VEGF164 overexpression during organogenesis resulted in albuminuria at birth and was associated with glomerulomegaly, uniform podocyte effacement, very few and wide foot processes joined by occluding junctions, almost complete absence of slit diaphragms, and swollen endothelial cells with few fenestrae as revealed by transmission electron microscopy. Podocyte VEGF164 overexpression after birth caused massive albuminuria in 70% of 2-week-old mice, glomerulomegaly, and minimal changes on light microscopy. Transmission electron microscopy showed podocyte effacement and fusion and morphologically normal endothelial cells. Podocyte VEGF164 overexpression induced nephrin down-regulation without podocyte loss. VEGF164-induced abnormalities were reversible on removal of doxycycline and were unresponsive to methylprednisolone. Collectively, the data suggest that moderate podocyte VEGF164 overexpression during organogenesis results in congenital nephrotic syndrome, whereas VEGF164 overexpression after birth induces a steroid-resistant minimal change like-disease in mice.


Asunto(s)
Riñón , Síndrome Nefrótico/metabolismo , Podocitos/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Humanos , Riñón/crecimiento & desarrollo , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Transgénicos , Nefrosis Lipoidea/genética , Nefrosis Lipoidea/metabolismo , Nefrosis Lipoidea/patología , Síndrome Nefrótico/genética , Síndrome Nefrótico/patología , Podocitos/patología , Podocitos/ultraestructura , Proteinuria/patología , Factor A de Crecimiento Endotelial Vascular/genética
18.
Kidney Int ; 77(11): 989-99, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20375978

RESUMEN

We sought to examine the pathogenic role of excessive VEGF-A expression in podocytes, since it has been reported that diabetic nephropathy and other glomerular diseases are associated with increased VEGF-A expression. The induction of podocyte-specific VEGF164 overexpression in adult transgenic mice led to proteinuria, glomerulomegaly, glomerular basement membrane thickening, mesangial expansion, loss of slit diaphragms, and podocyte effacement. When doxycycline-mediated VEGF164 was stopped, these abnormalities reversed. These findings were associated with reversible downregulation of metalloproteinase 9 and nephrin expression. Using transmission electron microscopy, we established that VEGF-A receptor-2 (VEGFR2) was expressed in podocytes and glomerular endothelial cells. We also found that VEGF164 induced VEGFR2 phosphorylation in podocytes. Further, we were able to co-immunoprecipitate VEGFR2 and nephrin using whole kidney lysates, confirming interaction in vivo. This implies that autocrine and paracrine VEGF-A signaling through VEGFR2 occurs in podocytes and may mediate the glomerular phenotype caused by VEGF164 overexpression. Thus, we suggest that podocyte VEGF164 overexpression in adult mice is sufficient to induce glomerular filtration barrier structural and functional abnormalities similar to those present in murine diabetic nephropathy.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Enfermedades Renales/metabolismo , Podocitos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factores de Edad , Animales , Comunicación Autocrina , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Genotipo , Membrana Basal Glomerular/patología , Enfermedades Renales/genética , Enfermedades Renales/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Comunicación Paracrina , Fenotipo , Fosforilación , Podocitos/patología , Unión Proteica , Proteinuria/genética , Proteinuria/metabolismo , Transducción de Señal , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
19.
Development ; 136(23): 3979-89, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19906865

RESUMEN

Semaphorin3a (Sema3a), a chemorepellant guidance protein, plays crucial roles in neural, cardiac and peripheral vascular patterning. Sema3a is expressed in the developing nephron, mature podocytes and collecting tubules. Sema3a acts as a negative regulator of ureteric bud branching, but its function in glomerular development has not been examined. Here we tested the hypothesis that Sema3a regulates glomerular vascular development using loss- and gain-of-function mouse models. Sema3a deletion resulted in defects in renal vascular patterning, excess endothelial cells within glomerular capillaries, effaced podocytes with extremely wide foot processes and albuminuria. Podocyte Sema3a overexpression during organogenesis resulted in glomerular hypoplasia, characterized by glomerular endothelial cell apoptosis, delayed and abnormal podocyte foot process development, a complete absence of slit diaphragms and congenital proteinuria. Nephrin, WT1 and VEGFR2 were downregulated in Sema3a-overexpressing kidneys. We conclude that Sema3a is an essential negative regulator of endothelial cell survival in developing glomeruli and plays a crucial role in podocyte differentiation in vivo. Hence, a tight regulation of Sema3a dosage is required for the establishment of a normal glomerular filtration barrier.


Asunto(s)
Diferenciación Celular/genética , Células Endoteliales/fisiología , Glomérulos Renales/crecimiento & desarrollo , Podocitos/fisiología , Semaforina-3A/fisiología , Animales , Apoptosis/fisiología , Recuento de Células , Núcleo Celular/metabolismo , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Técnica del Anticuerpo Fluorescente Directa , Colorantes Fluorescentes/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Griffonia/química , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Indoles/metabolismo , Glomérulos Renales/ultraestructura , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Lectinas de Plantas/metabolismo , Podocitos/citología , Podocitos/metabolismo , Podocitos/ultraestructura , Ratas , Proteínas Recombinantes/metabolismo , Semaforina-3A/genética , Semaforina-3A/metabolismo
20.
Nephron Clin Pract ; 95(1): c9-14, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14520016

RESUMEN

BACKGROUND: Malnutrition is widely prevalent in dialysis. Malnourished patients present depletion of somatic protein stores and a decrease in lean body mass (LBM) that can be measured by different techniques. AIMS: (1) To assess the reliability of lean mass measurements obtained by creatinine kinetics (CrK) in a group of stable peritoneal dialysis (PD) patients, using dual-energy x-ray absorptiometry (DEXA) measurements as the reference method; (2) to establish the reproducibility of LBM estimated by CrK in individual patients analyzing repeated measurement in the short term, and (3) to correlate measurements of LBM with laboratory determinations that assess nutritional status. METHODS: We performed a cross-sectional evaluation of LBM by DEXA and CrK in 39 PD patients. In 14 patients we performed repeated measurements of LBM by CrK in the short term. RESULTS: No significant difference was found in mean lean mass values estimated by both methods: mean DEXA LBM was 41.7 kg, 36.1 +/- 4.5 kg in females and 52.7 +/- 6.4 kg in males and mean CrK LBM was 41.08 kg, 37.5 +/- 6.1 kg in females and 48.1 +/- 8.4 kg in males. A good correlation was found between both techniques (r = 0.71; p < 0.003). The mean difference between the two methods was 0.638 +/- 6.95 kg (95% confidence limits: -12.98 and +14.26). A wide scatter of the differences between both methods was seen throughout the range of measurements of LBM. When LBM by CrK was repeatedly (2-3 times) measured in a period of 3-4 months in 14 patients, it had a coefficient of variation (CV) of 15.39% (range 2.89-42.88%), while body weight CV in the same period was 0.69% (range 0-1.9%). CONCLUSIONS: CrK is an unsatisfactory method for the assessment of LBM in PD patients.


Asunto(s)
Composición Corporal , Creatinina/metabolismo , Estado Nutricional , Diálisis Peritoneal , Absorciometría de Fotón , Índice de Masa Corporal , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...