Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Kidney Int ; 99(5): 1102-1117, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33412162

RESUMO

Focal segmental glomerulosclerosis (FSGS) is a podocytopathy leading to kidney failure, whose molecular cause frequently remains unresolved. Here, we describe a rare MYO9A loss of function nonsense heterozygous mutation (p.Arg701∗) as a possible contributor to disease in a sibling pair with familial FSGS/proteinuria. MYO9A variants of uncertain significance were identified by whole exome sequencing in a cohort of 94 biopsy proven patients with FSGS. MYO9A is an unconventional myosin with a Rho-GAP domain that controls epithelial cell junction assembly, crosslinks and bundles actin and deactivates the small GTPase protein encoded by the RHOA gene. RhoA activity is associated with cytoskeleton regulation of actin stress fiber formation and actomyosin contractility. Myo9A was detected in mouse and human podocytes in vitro and in vivo. Knockin mice carrying the p.Arg701∗MYO9A (Myo9AR701X) generated by gene editing developed proteinuria, podocyte effacement and FSGS. Kidneys and podocytes from Myo9AR701X/+ mutant mice revealed Myo9A haploinsufficiency, increased RhoA activity, decreased Myo9A-actin-calmodulin interaction, impaired podocyte attachment and migration. Our results indicate that Myo9A is a novel component of the podocyte cytoskeletal apparatus that regulates RhoA activity and podocyte function. Thus, Myo9AR701X/+ knock-in mice recapitulate the proband FSGS phenotype, demonstrate that p.R701X Myo9A is an FSGS-causing mutation in mice and suggest that heterozygous loss-of-function MYO9A mutations may cause a novel form of human autosomal dominant FSGS. Hence, identification of MYO9A pathogenic variants in additional individuals with familial or sporadic FSGS is needed to ascertain the gene contribution to disease.


Assuntos
Glomerulosclerose Segmentar e Focal , Miosinas/genética , Podócitos , Animais , Proteínas Ativadoras de GTPase/genética , Glomerulosclerose Segmentar e Focal/genética , Humanos , Camundongos , Miosinas/metabolismo , Fenótipo
2.
Pediatr Nephrol ; 35(6): 959-968, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31049720

RESUMO

The synchronized advent of high-throughput next-generation sequencing technology and knowledge of the human genome has rendered exponential contributions to our understanding of the pathophysiology of glomerular kidney diseases. A genetic diagnosis can now be made or confirmed in about two-thirds of the suspected inherited glomerular diseases. Next-generation sequencing is adept at identifying single nucleotide variations and small insertions or deletions that constitute majority of the disease-causing mutations. Description of the complete mutation spectrum in syndromic glomerulopathies may require the use of both sequencing and cytogenetic methods to detect large structural DNA variation in addition to single nucleotide changes. The enthusiastic application of genetic and genomic knowledge to inherited glomerular diseases has uncovered anticipated and unforeseen challenges mainly related to the biological interpretation of variants of uncertain significance and the limited benefit on clinical management for the individual patient when a diagnosis is obtained. To attain the ultimate goal of transforming clinical decision-making based on accurate genetic diagnosis using genomic information, these challenges need to be addressed. Till then, the glory of genomic medicine stands the test of time in this gilded age of genomic advancements.


Assuntos
Sequenciamento do Exoma , Falência Renal Crônica/genética , Insuficiência Renal Crônica/genética , Adolescente , Fatores Etários , Síndrome Brânquio-Otorrenal/diagnóstico por imagem , Síndrome Brânquio-Otorrenal/genética , Síndrome Brânquio-Otorrenal/patologia , Criança , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Falência Renal Crônica/diagnóstico , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Polimorfismo de Nucleotídeo Único
3.
Nephrol Dial Transplant ; 33(9): 1514-1525, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29635428

RESUMO

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.


Assuntos
Capilares/fisiopatologia , Taxa de Filtração Glomerular/fisiologia , Glomérulos Renais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Modelos Animais de Doenças , Glomérulos Renais/fisiopatologia , Camundongos , Camundongos Knockout , Nefrectomia
4.
J Am Soc Nephrol ; 27(4): 1055-65, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26303067

RESUMO

Podocytes are terminally differentiated epithelial cells that reside along the glomerular filtration barrier. Evidence suggests that after podocyte injury, endoplasmic reticulum stress response is activated, but the molecular mechanisms involved are incompletely defined. In a mouse model, we confirmed that podocyte injury induces endoplasmic reticulum stress response and upregulated unfolded protein response pathways, which have been shown to mitigate damage by preventing the accumulation of misfolded proteins in the endoplasmic reticulum. Furthermore, simultaneous podocyte-specific genetic inactivation of X-box binding protein-1 (Xbp1), a transcription factor activated during endoplasmic reticulum stress and critically involved in the untranslated protein response, and Sec63, a heat shock protein-40 chaperone required for protein folding in the endoplasmic reticulum, resulted in progressive albuminuria, foot process effacement, and histology consistent with ESRD. Finally, loss of both Sec63 and Xbp1 induced apoptosis in podocytes, which associated with activation of the JNK pathway. Collectively, our results indicate that an intact Xbp1 pathway operating to mitigate stress in the endoplasmic reticulum is essential for the maintenance of a normal glomerular filtration barrier.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Podócitos/fisiologia , Fatores de Transcrição/fisiologia , Animais , Células Cultivadas , Camundongos , Fatores de Transcrição de Fator Regulador X , Proteína 1 de Ligação a X-Box
5.
Am J Kidney Dis ; 65(3): 513-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25542409

RESUMO

Hyponatremia is an electrolyte abnormality that occurs in infancy due to a variety of inherited and acquired disorders. Infants with hyponatremia can present with neurologic symptoms such as vomiting, weakness, and seizures. Common causes of hyponatremia in the infant population are excess ingestion or administration of hypotonic fluids and excessive gastrointestinal salt loss. Hyponatremia in infancy also can be a sign of less common disorders, such as mineralocorticoid deficiency or resistance, and disregulation of arginine vasopressin with impaired free-water removal. Treatment of infants with hyponatremia is dependent on the severity of symptoms and the cause of hyponatremia. In nephrogenic syndrome of inappropriate antidiuresis (NSIAD), fluid retention is due to a gain-of-function mutation in the arginine vasopressin receptor 2 (AVPR2) gene leading to low arginine vasopressin levels. We describe the case of an infant with hyponatremia due to NSIAD, whose mother also has a known mutation in the AVPR2 gene. We report the approach to the treatment of hyponatremia and its unique challenges in infancy.


Assuntos
Hiponatremia/diagnóstico , Hiponatremia/terapia , Humanos , Hiponatremia/sangue , Lactente , Masculino , Sódio/sangue , Resultado do Tratamento , Ureia/administração & dosagem
6.
J Am Soc Nephrol ; 25(8): 1814-24, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24578128

RESUMO

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.


Assuntos
Nefropatias Diabéticas/etiologia , Óxido Nítrico Sintase Tipo III/fisiologia , Podócitos/metabolismo , Proteinúria/etiologia , Insuficiência Renal/etiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Técnicas de Cultura de Células , Colágeno Tipo IV/metabolismo , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Membrana Basal Glomerular/metabolismo , Membrana Basal Glomerular/patologia , Laminina/metabolismo , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo III/genética , Proteinúria/metabolismo , Proteinúria/patologia , Insuficiência Renal/metabolismo , Insuficiência Renal/patologia
7.
Am J Pathol ; 183(4): 1156-1168, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23954273

RESUMO

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.


Assuntos
Nefropatias/patologia , Glomérulos Renais/patologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Podócitos/metabolismo , Podócitos/patologia , Receptores de Superfície Celular/metabolismo , Semaforina-3A/metabolismo , Animais , Forma Celular , Regulação para Baixo , Membrana Basal Glomerular/metabolismo , Membrana Basal Glomerular/patologia , Membrana Basal Glomerular/ultraestrutura , Integrina alfaVbeta3/metabolismo , Nefropatias/metabolismo , Glomérulos Renais/metabolismo , Glomérulos Renais/ultraestrutura , Camundongos , Podócitos/ultraestrutura , Ligação Proteica , Proteinúria/metabolismo , Proteinúria/patologia , Transdução de Sinais
8.
Pediatr Nephrol ; 29(4): 751-5, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24464477

RESUMO

Semaphorin3a (sema3a), a member of class 3 semaphorins, is a guidance protein that regulates angiogenesis, branching morphogenesis, axon growth, and cell migration, and has pleiotropic roles on organogenesis, immune response, and cancer. Sema3a is secreted by podocytes and is required for normal kidney patterning and glomerular filtration barrier development. We recently discovered that after completion of kidney development, Sema3a gain-of-function in podocytes leads to proteinuric glomerular disease in mice. Excess sema3a causes foot process effacement, glomerular basement lamination, and endothelial damage in vivo, and disrupts cell autonomously podocyte shape by down-regulating nephrin and inhibiting αvß3 integrin. We identified a novel direct interaction between nephrin and plexinA1, the sema3a signaling receptor. Nephrin-plexinA1 interaction links the slit-diaphragm signaling complex to extracellular sema3a signals. Hence, sema3a functions as an extracellular negative regulator of the structure and function of the glomerular filtration barrier.


Assuntos
Rim/embriologia , Podócitos/metabolismo , Podócitos/patologia , Semaforina-3A/metabolismo , Animais , Humanos , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Organogênese , Semaforina-3A/genética , Transdução de Sinais
9.
Clin Nephrol ; 82(3): 205-10, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23391318

RESUMO

Chronic kidney disease is common in pediatric patients following hematopoietic stem cell transplant. Its etiology is likely multifactorial and depends both on pre-conditioning regimens as well as immunosuppressive therapy and posttransplant prophylactic medications. Graft vs. host disease (GVHD) is a common sequela of hematopoietic stem cell transplant and has been associated with the nephrotic syndrome (NS). Here we report a case of a pediatric patient who developed proteinuria and renal insufficiency after stem cell transplant. A kidney biopsy showed chronic interstitial nephritis and extensive foot process effacement, which are likely sequelae of GVHD. Moreover we show decreased CD4 and CD3 lymphocyte counts in the interstitial infiltrate, suggesting that abnormal lymphocyte response might play a role in podocyte injury following GVHD. This case illustrates the importance of the kidney biopsy in the assessment of stem cell transplant-mediated renal failure.


Assuntos
Doença Enxerto-Hospedeiro/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Nefrite Intersticial/etiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/cirurgia , Proteinúria/etiologia , Insuficiência Renal/etiologia , Biópsia , Criança , Evolução Fatal , Doença Enxerto-Hospedeiro/diagnóstico , Humanos , Rim/patologia , Masculino , Nefrite Intersticial/diagnóstico , Valor Preditivo dos Testes , Proteinúria/diagnóstico , Recidiva , Insuficiência Renal/diagnóstico
11.
J Biol Chem ; 286(46): 39933-44, 2011 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21937443

RESUMO

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.


Assuntos
Actinas/metabolismo , Proteínas de Membrana/metabolismo , Complexos Multiproteicos/metabolismo , Podócitos/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Actinas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células COS , Chlorocebus aethiops , Espectrometria de Massas , Proteínas de Membrana/genética , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos/genética , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Fosforilação/fisiologia , Podócitos/citologia , Estrutura Terciária de Proteína , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética
12.
Development ; 136(23): 3979-89, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19906865

RESUMO

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.


Assuntos
Diferenciação Celular/genética , Células Endoteliais/fisiologia , Glomérulos Renais/crescimento & desenvolvimento , Podócitos/fisiologia , Semaforina-3A/fisiologia , Animais , Apoptose/fisiologia , Contagem de Células , Núcleo Celular/metabolismo , Técnicas de Cocultura , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Técnica Direta de Fluorescência para Anticorpo , Corantes Fluorescentes/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Griffonia/química , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Indóis/metabolismo , Glomérulos Renais/ultraestrutura , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Lectinas de Plantas/metabolismo , Podócitos/citologia , Podócitos/metabolismo , Podócitos/ultraestrutura , Ratos , Proteínas Recombinantes/metabolismo , Semaforina-3A/genética , Semaforina-3A/metabolismo
13.
Am J Pathol ; 177(5): 2225-33, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20829436

RESUMO

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.


Assuntos
Rim , Síndrome Nefrótica/metabolismo , Podócitos/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Humanos , Rim/crescimento & desenvolvimento , Rim/metabolismo , Rim/patologia , Camundongos , Camundongos Transgênicos , Nefrose Lipoide/genética , Nefrose Lipoide/metabolismo , Nefrose Lipoide/patologia , Síndrome Nefrótica/genética , Síndrome Nefrótica/patologia , Podócitos/patologia , Podócitos/ultraestrutura , Proteinúria/patologia , Fator A de Crescimento do Endotélio Vascular/genética
14.
Pediatr Nephrol ; 26(9): 1407-12, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21336944

RESUMO

Semaphorins are guidance proteins that play important roles in organogenesis and disease. Expression of class 3 semaphorins and their receptors is regulated during kidney development. Gain- and loss-of-function experiments demonstrated that tight semaphorin3a gene dosage is required for podocyte differentiation, and for the establishment of a normal glomerular filtration barrier. Sema3a modulates kidney vascular patterning acting as a negative regulator of endothelial cell migration and survival. Excess podocyte semaphorin3a expression causes glomerular disease in mice. In addition, Sema3a is a negative regulator of ureteric bud branching, whereas Sema3c is a positive regulator of ureteric bud and endothelial cell branching morphogenesis. In summary, secreted semaphorins modulate ureteric bud branching, vascular patterning, and podocyte-endothelial crosstalk, suggesting that they play a role in renal disease. Understanding the signaling pathways downstream from semaphorin receptors will provide insight into the mechanism of action of semaphorins in renal pathology.


Assuntos
Comunicação Celular , Células Epiteliais/metabolismo , Nefropatias/metabolismo , Rim/metabolismo , Neovascularização Fisiológica , Podócitos/metabolismo , Semaforina-3A/metabolismo , Ureter/metabolismo , Animais , Humanos , Rim/irrigação sanguínea , Rim/embriologia , Morfogênese , Transdução de Sinais , Ureter/embriologia
16.
Front Med (Lausanne) ; 8: 679518, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34336885

RESUMO

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.

17.
Front Pharmacol ; 12: 788886, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35280251

RESUMO

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.

18.
Kidney Int ; 77(11): 989-99, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20375978

RESUMO

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.


Assuntos
Nefropatias Diabéticas/metabolismo , Nefropatias/metabolismo , Podócitos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores Etários , Animais , Comunicação Autócrina , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Genótipo , Membrana Basal Glomerular/patologia , Nefropatias/genética , Nefropatias/patologia , Metaloproteinase 9 da Matriz/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Comunicação Parácrina , Fenótipo , Fosforilação , Podócitos/patologia , Ligação Proteica , Proteinúria/genética , Proteinúria/metabolismo , Transdução de Sinais , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
FASEB J ; 20(12): 2150-2, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16940438

RESUMO

Class 3 semaphorins (sema 3) are secreted guidance proteins. Sema 3A expressed by endothelial cells controls vascular morphogenesis through integrin inhibition. Sema 3C is required for normal cardiovascular patterning. Here we examined the potential role of sema 3C as regulator of endothelial cell function in vitro using mouse glomerular endothelial cells (MGEC). We determined that MGEC express sema 3C mRNA and protein and its receptors mRNA. Recombinant sema 3C induced MGEC proliferation 18 +/- 2% above control, as assessed by bromodeoxyuridine (BrdU) incorporation, and reduced starvation-induced apoptosis by 46 +/- 3%, as indicated by an in situ marker of activated caspase 3. Sema 3C increased MGEC adhesion to fibronectin 79 +/- 13% and to collagen 55 +/- 12% as compared with control. Sema 3C-induced MGEC adhesion was prevented by integrin blocking antibodies and involved beta1 integrin serine phosphorylation. Sema 3C-induced MGEC adhesion and proliferation were similar to those induced by vascular endothelial growth factor (VEGF)-A. Sema 3C induced a 44 +/- 11% increase in MGEC directional migration and stimulated MGEC capillary-like network formation on collagen I gels. Collectively, our data indicate that sema 3C promotes glomerular endothelial cell proliferation, adhesion, directional migration, and tube formation in vitro by stimulating integrin phosphorylation and VEGF120 secretion, functions that are similar to VEGF-A and opposite to sema 3A.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Integrina beta1/efeitos dos fármacos , Semaforinas/fisiologia , Animais , Adesão Celular , Movimento Celular , Proliferação de Células , Células Endoteliais/citologia , Endotélio Vascular/citologia , Integrina beta1/metabolismo , Integrinas/efeitos dos fármacos , Integrinas/metabolismo , Rim/citologia , Camundongos , Neovascularização Fisiológica , Fosforilação , Proteínas Recombinantes/farmacologia , Semaforinas/farmacologia , Fator A de Crescimento do Endotélio Vascular/farmacologia
20.
Methods Mol Biol ; 1493: 393-399, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27787866

RESUMO

Podocytes are complex epithelial cells with foot processes that are essential for the integrity and function of the kidney glomerular filters. Podocyte foot processes linked by slit diaphragms constitute signaling platforms that tightly regulate the cell shape and the function of the filtration barrier. Semaphorin (Sema) 3A is a class 3 semaphorin secreted by podocytes that has autocrine and paracrine functions in the kidney. We have shown that Sema3A regulates podocyte shape and that excess Sema3A signaling induces glomerular disease and aggravates diabetic nephropathy. MICAL-1 is an actin-binding protein that mediates Sema3A signals in podocytes. This chapter describes the methods used to examine how Sema3A signaling regulates podocyte shape.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Forma Celular , Proteínas do Citoesqueleto/fisiologia , Proteínas com Domínio LIM/fisiologia , Podócitos/citologia , Semaforina-3A/fisiologia , Animais , Linhagem Celular , Linhagem Celular Transformada , Humanos , Camundongos , Proteínas dos Microfilamentos , Oxigenases de Função Mista
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA