Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Am J Physiol Renal Physiol ; 292(2): F701-10, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17047167

RESUMO

In addition to forming the selective filtration barrier for the renal glomerulus, podocytes maintain glomerular capillary architecture by opposing distending hemodynamic forces. To understand the relationship of cytoskeletal properties and the mechanical characteristics of podocytes, we studied filamin expression and distribution and measured cell membrane deformability in conditionally immortalized wild-type (WT) mouse podocytes, and in podocytes derived from a mouse model of HIV-associated nephropathy (HIVAN). In the WT cells, filamin and F-actin were localized at the periphery and in prominent stress fibers. In the HIVAN cells, filamin expression was reduced, and stress fibers were sparse. In a microaspiration assay, HIVAN cells ruptured under minimal negative pressure. Atomic force microscopy demonstrated that the WT cells had a stiffness of 17 kPa, whereas the value for the HIVAN cells was 4 kPa. These results demonstrate that the mechanical properties of WT and HIVAN podocytes are markedly different in a manner that is consistent with differences in the composition and arrangement of their cytoskeletons. The mechanical properties of the WT podocytes suggest that these cells can better maintain capillary integrity than the HIVAN podocytes and implicate pathological assembly of the cytoskeleton as a mechanism of HIVAN.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Infecções por HIV/fisiopatologia , Podócitos/metabolismo , Actinina/biossíntese , Actinas/biossíntese , Animais , Fenômenos Biomecânicos , Células Cultivadas , Proteínas Contráteis/biossíntese , Filaminas , Quinase 1 de Adesão Focal/biossíntese , Camundongos , Proteínas dos Microfilamentos/biossíntese , Microscopia de Força Atômica , Microscopia de Fluorescência , Microscopia de Contraste de Fase , Podócitos/fisiologia
2.
Cell Mol Biol (Noisy-le-grand) ; 52(8): 115-21, 2006 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-17535745

RESUMO

NHE1 is a ubiquitously expressed Na+/H+ exchanger, which is important for vital cell functions. Using in vivo models of kidney podocyte injury and renal tubular epithelial cell (RTC) culture systems, we previously demonstrated that NHE1 defends against apoptosis by a mechanism involving ezrin binding to the NHE1 cytoplasmic domain. We now extend the NHE1 role to diabetic mouse models and refine the mechanism of NHE1-dependent ezrin activation. Streptozotocin induced diabetes resulted in greater azotemia, albuminuria and tubulointerstitial pathology in NHE1-deficient swe/swe compared to wild-type control mice. Increased RTC apoptosis was noted in swe/swe mice, suggesting that loss of NHE1 function leads to tubular atrophy, which predicts kidney disease progression. In vitro, proximal RTC derived from swe/swe mice also underwent increased apoptosis in response to staurosporine or a hypertonic environment. Activated ezrin normally resides in the apical domain of the proximal RTC, while NHE1 is a basolateral protein. After NHE1 activation by intracellular acidification or extracellular hypertonicity, confocal immunofluorescence microscopy in polarized LLC-PK1 cells demonstrated transient ezrin localization to lateral membrane domains, where it is positioned to interact with NHE1. We conclude that cell stresses promote NHE1-ezrin interaction, which activate cell survival pathways to prevent apoptosis in diabetic and non-diabetic kidney diseases.


Assuntos
Proteínas de Transporte de Cátions/fisiologia , Proteínas do Citoesqueleto/metabolismo , Diabetes Mellitus Experimental/metabolismo , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia , Animais , Apoptose , Linhagem Celular , Membrana Celular/metabolismo , Polaridade Celular/fisiologia , Sobrevivência Celular/fisiologia , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Células Epiteliais/fisiologia , Camundongos , Camundongos Mutantes , Fosforilação , Transporte Proteico , Trocador 1 de Sódio-Hidrogênio , Estreptozocina
3.
Kidney Int ; 60(1): 65-76, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11422737

RESUMO

BACKGROUND: In chronic renal disease, renal tubular epithelial cell (RTC) Fas expression is up-regulated, leading to apoptotic RTC deletion and tubular atrophy. In vitro, cytokine- or hypoxia-induced up-regulation of Fas expression is associated with RTC apoptosis. In contrast, constitutively expressed, low level RTC Fas does not mediate apoptosis, suggesting that Fas may be coupled to expression level-dependent RTC signaling pathways. Fas is known to signal through JNK in many systems, but the requirement of JNK activation for apoptosis remains controversial. METHODS: To determine if RTC Fas regulates JNK activity and apoptosis, human RTC were transfected with graded concentrations of a eukaryotic expression vector for murine Fas. Apoptosis was measured by annexin V, TUNEL and PARP cleavage assays. JNK activity was determined by immune complex kinase assay and/or immunoblots with phospho-specific JNK antibodies, in the presence or absence of co-expressed dominant negative JNK constructs. RESULTS: Fas antibody stimulation of RTC with high Fas expression levels (to model RTC phenotype in renal disease) caused a tenfold increase in apoptosis, while RTC with low level Fas expression (to model normal RTC phenotype) were apoptosis-resistant. Fas ligation activated JNK in RTC expressing low levels of Fas, but not in apoptosis-sensitive RTC with increased Fas expression. Dominant negative JNK co-expression failed to inhibit apoptosis in RTC expressing high levels of Fas, suggesting that JNK is neither necessary, nor sufficient, for Fas-dependent apoptosis. CONCLUSIONS: At high levels of expression, RTC Fas promotes apoptosis in a JNK-independent manner. At low basal expression, Fas induces JNK activation, but not apoptosis, consistent with novel roles for RTC Fas as a mediator of cell stress or chronic inflammation.


Assuntos
Apoptose/fisiologia , Túbulos Renais Proximais/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptor fas/metabolismo , Animais , Membrana Celular/metabolismo , Células Cultivadas , DNA Complementar/fisiologia , Ativação Enzimática/fisiologia , Células Epiteliais/fisiologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno , Camundongos , Proteína Quinase 8 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Plasmídeos/fisiologia , Transfecção , Receptor fas/genética
4.
Nat Cell Biol ; 3(5): 527-30, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11331884

RESUMO

Interactions between Eph receptor tyrosine kinases (RTKs) and membrane-anchored ephrin ligands critically regulate axon pathfinding and development of the cardiovascular system, as well as migration of neural cells. Similar to other RTKs, ligand-activated Eph kinases recruit multiple signalling and adaptor proteins, several of which are involved in growth regulation. However, in contrast to other RTKs, activation of Eph receptors fails to promote cell proliferation or to transform rodent fibroblasts, indicating that Eph kinases may initiate signalling pathways that are distinct from those transmitted by other RTKs. Here we show that stimulation of endogenous EphA kinases with ephrin-A1 potently inhibits the Ras/MAPK cascade in a range of cell types, and attenuates activation of mitogen-activated protein kinase (MAPK) by receptors for platelet-derived growth factor (PDGF), epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF). In prostatic epithelial cells and endothelial cells, but not fibroblasts, treatment with ephrin-A1 inhibits cell proliferation. Our results identify EphA kinases as negative regulators of the Ras/MAPK pathway that exert anti-mitogenic functions in a cell-type-specific manner.


Assuntos
Sistema de Sinalização das MAP Quinases , Proteínas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Proteínas ras/antagonistas & inibidores , Animais , Divisão Celular , Linhagem Celular , Fatores de Crescimento Endotelial/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Efrina-A1 , Fator de Crescimento Epidérmico/metabolismo , Fibroblastos/metabolismo , Humanos , Immunoblotting , Queratinócitos/metabolismo , Linfocinas/metabolismo , Masculino , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Fator de Crescimento Derivado de Plaquetas/metabolismo , Testes de Precipitina , Neoplasias da Próstata/metabolismo , Ratos , Receptor EphA1 , Receptor EphA2 , Transdução de Sinais , Fatores de Tempo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Proteínas ras/metabolismo
5.
Am J Kidney Dis ; 37(3): 638-47, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11228193

RESUMO

Diabetic nephropathy (DN) clusters in families and specific ethnic groups, suggesting a genetic basis of disease transmission. Identification of DN susceptibility loci should reveal new therapeutic targets but requires accurate phenotyping. A powerful family-based strategy, which is novel to the pursuit of nephropathy genes in type 2 diabetes, is being used to collect a sample for candidate gene and genome scan analyses. Sib pairs that include DN index cases plus (1) sibs concordant for type 2 diabetes and DN (affected sib pairs [ASPs]) and (2) sibs concordant for type 2 diabetes but discordant for DN (discordant sib pairs [DSPs]) are targeted specifically for recruitment. Type 2 diabetes and DN phenotype criteria for index cases include diabetes onset after 38 years of age, duration 10 years or longer, no initial insulin treatment, diabetic retinopathy, end-stage renal disease (ESRD), and history of nephrotic proteinuria. ESRD patients were screened by questionnaire and medical record review (n = 2114). Of 666 patients with ESRD secondary to DN, 227 had a family history of ESRD, 150 had a living diabetic sib, and 124 families were enrolled. Sixty-five families, with 86 diabetic relative pairs (69 sibs, 17 children), have been completely phenotyped. If nephropathy in diabetic sibs is defined as albuminuria greater than 0.3 g/24 h, 31 ASPs and 26 DSPs (diabetic sib with albuminuria <0.3 g/24 h) were identified. Applying more stringent criteria, only 12 ASPs (sib with diabetes >10 years, diabetic retinopathy, and nephrotic proteinuria) and 9 DSPs (sib with diabetes >10 years and normal urine albumin excretion) were identified. Extrapolating from the number of subjects recruited using stringent phenotyping criteria, nearly 10,000 ESRD patients are required for screening to achieve adequate statistical power for linkage analysis (80% power to detect locus-specific relative risk of 2.2 at a lod score of 3.0). Careful phenotyping requires a large recruitment effort but is necessary to reduce population heterogeneity, a strategy that increases the likelihood of identifying DN loci.


Assuntos
Diabetes Mellitus Tipo 2/genética , Nefropatias Diabéticas/genética , Marcadores Genéticos , Predisposição Genética para Doença , Idade de Início , Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/complicações , Progressão da Doença , Família , Feminino , Genes , Humanos , Falência Renal Crônica/etnologia , Falência Renal Crônica/etiologia , Falência Renal Crônica/genética , Masculino , Inquéritos e Questionários
6.
Nephrol Dial Transplant ; 15(11): 1801-7, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11071968

RESUMO

BACKGROUND: Multiple studies suggest that hypertension-induced end-stage renal disease (ESRD) is heritable. Identification of nephropathy susceptibility genes absolutely requires accurate phenotyping, but the clinical hypertensive nephrosclerosis (HN) phenotype is poorly characterized. We hypothesized that many patients with HN as the indicated cause of ESRD on the Health Care Financing Administration (HCFA) 2728 form, fail to satisfy stringent HN phenotyping criteria. METHODS: Since renal biopsy documentation of HN is uncommon, clinical parameters for HN phenotype were applied: family history of hypertension, left ventricular hypertrophy, proteinuria <0.5 g/day, and hypertension preceding renal dysfunction (Schlessinger et al., 1994) or urine protein:creatinine (prot:creat) ratio <2.0 and no evidence of other renal diseases (AASK Trial Group, 1997). RESULTS: ESRD patients (n=607, 73% African American, 25% Caucasian) were enrolled in a study to identify HN susceptibility genes. HN was the most common cause of ESRD according to HCFA 2728 forms (37% prevalence). Phenotyping of randomly selected patients with HN from the total cohort revealed that 4/100 subjects satisfied the Schlessinger criteria, and 28/91 African Americans met AASK criteria for HN. From these figures, the adjusted prevalence of HN was only 1.5-13.5%. Of patients that could not be phenotyped for HN, 14 were misdiagnosed, 14 had urine prot:creat >2.0, and insufficient data were available in the remainder. Four patients underwent renal biopsy, but histology from only one was consistent with HN. If the HN phenotype definitions are revised to exclude 'hypertension preceding renal dysfunction', or proteinuria limits, then 44/100 and 39/91 patients respectively satisfy clinical phenotyping parameters for HN. CONCLUSIONS: (i) We provide the strongest evidence to date that HN is less frequent in an ESRD population than commonly assumed if strict clinical criteria are used; many patients clinically diagnosed with HN may have undetected, treatable renal disease from other causes; (ii) relaxing HN phenotype criteria may erroneously include patients with glomerular diseases and secondary hypertension; (iii) reliance on HCFA 2728 diagnoses will confound identification of HN susceptibility genes; (iv) to attain adequate statistical power for genotype analysis, rigorous HN phenotyping will require screening an extremely large number of patients, which can be reasonably accomplished only in a multi-centre trial design.


Assuntos
Predisposição Genética para Doença , Falência Renal Crônica/genética , Nefroesclerose/genética , População Negra/genética , Diabetes Mellitus Tipo 1/fisiopatologia , Diabetes Mellitus Tipo 2/fisiopatologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/fisiopatologia , Humanos , Nefroesclerose/epidemiologia , Nefroesclerose/fisiopatologia , Fenótipo , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/fisiopatologia , Prevalência , Estados Unidos/epidemiologia , População Branca/genética
7.
Kidney Int ; 58(4): 1742-50, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11012908

RESUMO

BACKGROUND: A homogeneous patient population is necessary to identify genetic factors that regulate complex disease pathogenesis. In this study, we evaluated clinical and biochemical phenotyping criteria for type 2 diabetes in end-stage renal disease (ESRD) probands of families in which nephropathy is clustered. C-peptide concentrations accurately discriminate type 1 from type 2 diabetic patients with normal renal function, but have not been extensively evaluated in ESRD patients. We hypothesized that C-peptide concentrations may not accurately reflect insulin synthesis in ESRD subjects, since the kidney is the major site of C-peptide catabolism and would poorly correlate with accepted clinical criteria used to classify diabetics as types 1 and 2. METHODS: Consenting diabetic ESRD patients (N = 341) from northeastern Ohio were enrolled. Clinical history was obtained by questionnaire, and predialysis blood samples were collected for C-peptide levels from subjects with at least one living diabetic sibling (N = 127, 48% males, 59% African Americans). RESULTS: Using clinical criteria, 79% of the study population were categorized as type 1 (10%) or type 2 diabetics (69%), while 21% of diabetic ESRD patients could not be classified. In contrast, 98% of the patients were classified as type 2 diabetics when stratified by C-peptide concentrations using criteria derived from the Diabetes Control and Complications Trial Research Group (DCCT) and UREMIDIAB studies. Categorization was concordant in only 70% of ESRD probands when C-peptide concentration and clinical classification algorithms were compared. Using clinical phenotyping criteria as the standard for comparison, C-peptide concentrations classified diabetic ESRD patients with 100% sensitivity, but only 5% specificity. The mean C-peptide concentrations were similar in diabetic ESRD patients (3.2 +/- 1.9 nmol/L) and nondiabetic ESRD subjects (3.5 +/- 1.7 nmol/L, N = 30, P = NS), but were 2.5-fold higher compared with diabetic siblings (1.3 +/- 0.7 nmol/L, N = 30, P < 0.05) with normal renal function and were indistinguishable between type 1 and type 2 diabetics. Although 10% of the diabetic ESRD study population was classified as type 1 diabetics using clinical criteria, only 1.5% of these patients had C-peptide levels less than 0.20 nmol/L, the standard cut-off used to discriminate type 1 from type 2 diabetes in patients with normal renal function. However, the criteria of C-peptide concentrations> 0.50 nmol/L and diabetes onset in patients who are more than 38 years old identify type 2 diabetes with a 97% positive predictive value in our ESRD population. CONCLUSIONS: Accepted clinical criteria, used to discriminate type 1 and type 2 diabetes, failed to classify a significant proportion of diabetic ESRD patients. In contrast to previous reports, C-peptide levels were elevated in the majority of type 1 ESRD diabetic patients and did not improve the power of clinical parameters to separate them from type 2 diabetic or nondiabetic ESRD subjects. Accurate classification of diabetic ESRD patients for genetic epidemiological studies requires both clinical and biochemical criteria, which may differ from norms used in diabetic populations with normal renal function.


Assuntos
Peptídeo C/sangue , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/genética , Falência Renal Crônica/sangue , Falência Renal Crônica/genética , Adulto , Idade de Início , Algoritmos , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/genética , Nefropatias Diabéticas/sangue , Nefropatias Diabéticas/classificação , Nefropatias Diabéticas/genética , Feminino , Predisposição Genética para Doença , Humanos , Insulina/metabolismo , Secreção de Insulina , Falência Renal Crônica/classificação , Masculino , Pessoa de Meia-Idade , Fenótipo , Valor Preditivo dos Testes , Sensibilidade e Especificidade
8.
Am J Physiol Renal Physiol ; 279(2): F383-92, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10919859

RESUMO

Chronic renal disease initiation and progression remain incompletely understood. Genome-wide expression monitoring should clarify mechanisms that cause progressive renal disease by determining how clusters of genes coordinately change their activity. Serial analysis of gene expression (SAGE) is a technique of expression profiling, which permits simultaneous, comparative, and quantitative analysis of gene-specific, 9- to 13-bp sequence tags. Using SAGE, we have constructed a tag expression library from ROP-+/+ mouse kidney. Tag sequences were sorted by abundance, and identity was determined by sequence homology searching. Analyses of 3,868 tags yielded 1,453 unique kidney transcripts. Forty-two percent of these transcripts matched mRNA sequence entries with known function, 35% of the transcripts corresponded to expressed sequence tag (EST) entries or cloned genes, whose function has not been established, and 23% represented unidentified genes. Previously characterized transcripts were clustered into functional groups, and those encoding metabolic enzymes, plasma membrane proteins (transporters/receptors), and ribosomal proteins were most abundant (39, 14, and 12% of known transcripts, respectively). The most common, kidney-specific transcripts were kidney androgen-regulated protein (4% of all transcripts), sodium-phosphate cotransporter (0.3%), renal cytochrome P-450 (0.3%), parathyroid hormone receptor (0.1%), and kidney-specific cadherin (0.1%). Comprehensively characterizing and contrasting gene expression patterns in normal and diseased kidneys will provide an alternative strategy to identify candidate pathways, which regulate nephropathy susceptibility and progression, and novel targets for therapeutic intervention.


Assuntos
Expressão Gênica , Biblioteca Gênica , Técnicas Genéticas , Rim/fisiologia , Animais , Sequência de Bases/genética , Nefropatias/genética , Masculino , Camundongos , Valores de Referência , Transcrição Gênica
9.
Clin Nephrol ; 53(1): 61-5, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10661484

RESUMO

Severe symptomatic hypermagnesemia is a rare clinical problem that predominantly results from excess exogenous magnesium intake in patients with renal failure. This report describes an elderly woman who was given a magnesium-containing cathartic for pre-operative bowel preparation in the context of unrecognized acute renal failure. She subsequently developed one of the highest serum magnesium concentrations ever reported. The hypermagnesemia was successfully treated with continuous arteriovenous hemodialysis, but she ultimately died from complications of hypermagnesemia, that included junctional bradycardia, myocardial infarction and respiratory failure. This case illustrates the importance of ensuring intact renal function prior to administering large quantities of oral magnesium. More specifically, large doses of magnesium salts should be avoided in patients with acute renal failure.


Assuntos
Injúria Renal Aguda/sangue , Magnésio/sangue , Doenças Metabólicas/sangue , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/complicações , Idoso , Idoso de 80 Anos ou mais , Catárticos/efeitos adversos , Catárticos/uso terapêutico , Ácido Cítrico/efeitos adversos , Ácido Cítrico/uso terapêutico , Evolução Fatal , Feminino , Humanos , Magnésio/efeitos adversos , Doenças Metabólicas/induzido quimicamente , Compostos Organometálicos/efeitos adversos , Compostos Organometálicos/uso terapêutico
10.
Kidney Int ; 56(4): 1313-6, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10504483

RESUMO

Renal tubular atrophy predicts a poor prognosis in chronic renal failure, but the molecular mechanisms that regulate tubular atrophy are unknown. Because the Fas apoptosis pathway has been implicated in disease pathogenesis and Fas is expressed in kidney, we hypothesized that Fas-mediated renal tubule epithelial cell (RTC) apoptosis contributes to tubular atrophy in chronic renal failure. Immunohistochemical analyses of renal sections from two murine models of progressive renal disease revealed increases in RTC Fas expression and apoptosis compared with tissue sections from age-matched control kidneys. Increased RTC apoptosis was not accompanied by compensatory hyperplasia, suggesting that RTCs targeted for Fas-dependent apoptotic deletion contribute to tubular atrophy. These data are supported by in vitro studies that showed that interleukin-1alpha or tumor necrosis factor-alpha, cytokines that are secreted in chronic renal failure, stimulated increases in Fas expression in cultured RTCs. Both murine kidney cortex and RTCs in culture demonstrated constitutive expression of Fas ligand, a feature that is characteristically restricted to lymphocytes and immune-privileged tissues and previously unrecognized in RTCs. Functional studies revealed that interleukin-1alpha-stimulated RTC Fas expression was accompanied by increased apoptosis, which was inhibited by blocking anti-Fas ligand antibodies. The data suggest that up-regulated RTC Fas binds to Fas ligand on adjacent RTCs, which then leads to RTC death by fratricide. We propose this pathway as an initiating mechanism of tubular atrophy.


Assuntos
Apoptose/fisiologia , Células Epiteliais/citologia , Falência Renal Crônica/patologia , Túbulos Renais Proximais/patologia , Receptor fas/metabolismo , Animais , Atrofia , Células Epiteliais/química , Células Epiteliais/metabolismo , Proteína Ligante Fas , Humanos , Imunoglobulina G/farmacologia , Marcação In Situ das Extremidades Cortadas , Interleucina-1/farmacologia , Falência Renal Crônica/metabolismo , Túbulos Renais Proximais/metabolismo , Glicoproteínas de Membrana/análise , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Mutantes , Receptor fas/análise , Receptor fas/imunologia
11.
Curr Opin Nephrol Hypertens ; 8(4): 465-72, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10491742

RESUMO

New methods have been developed to uncover the genotypes that result in complex diseases. End-stage renal disease is a complex disease, without a simple correspondence between genotype and phenotype. Both population-based and family-based epidemiological studies and analysis of model organisms suggest that the pathogenesis of end-stage renal disease may have a genetic component. A number of studies have analyzed candidate nephropathy genes with little success, but recently several well-designed studies of multiplex families with diabetic nephropathy have identified candidate nephropathy susceptibility loci. To date, kidney disease-oriented research has focused on effector mechanisms responsible for the initiation and progression of chronic renal disease. However, because end-stage renal disease is a complex disease, interruption of a single effector pathway is unlikely to result in significant therapeutic benefit. Further understanding of the pathogenesis of kidney disease and the development of new kidney disease therapies will require continued application of genetic and genomic tools to kidney disease research.


Assuntos
Predisposição Genética para Doença , Falência Renal Crônica/genética , Animais , Família , Feminino , Humanos , Falência Renal Crônica/fisiopatologia , Masculino , Linhagem
12.
Lab Invest ; 79(9): 1089-99, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10496527

RESUMO

Renal tubular atrophy characterizes chronic progressive renal disease, but the molecular mechanisms of renal tubular cell (RTC) deletion are unclear. Because glomerular sclerosis leads to impaired peritubular blood flow, we tested the hypothesis that chronic hypoxia contributes to RTC apoptosis. Tubule hypoxia in mice with progressive renal disease (Os/+) was assessed by injecting EF5, a nitroimidazole compound that preferentially binds to cells undergoing anaerobic metabolism. Hypoxic tubules, as determined by direct immunofluorescence with anti-EF5 antibodies, were identified in kidneys from Os/+ mice, but not in age-matched controls (+/+) at 12 weeks, coincident with the onset of glomerular pathology. Hypoxia can cause apoptosis, but apoptotic RTCs were rare and equivalent in number in 12 week Os/+ and +/+ kidneys. However, by 16 weeks apoptotic RTCs were significantly more frequent in Os/+ versus +/+ mice, demonstrating that tubule hypoxia preceded RTC apoptosis. Importantly, apoptotic RTCs co-localized to hypoxic, but not normoxic tubules, indicating that tubular atrophy may result from hypoxic stimulation of RTC apoptosis. We have previously demonstrated enhanced, diffuse expression of the Fas apoptosis receptor in Os/+ tubules, providing a potential intermediary between hypoxia and apoptosis. To determine whether hypoxia stimulates Fas-dependent apoptosis, RTCs were cultured within a hypoxia chamber or in the presence of the cyanide analog, sodium azide. Both in vitro hypoxic conditions stimulated RTC plasma membrane Fas expression, and caused RTC apoptosis upon ligation with agonistic Fas antibodies. The data suggest that in the context of progressive renal disease, chronic hypoxia stimulates Fas-dependent RTC apoptosis, which represents the first definitive link between hypoxia and tubular atrophy. We believe that hypoxic induction of RTC apoptosis provides a unifying mechanism for the pathogenesis of tubular atrophy, and this paradigm identifies novel targets for chronic renal failure therapy.


Assuntos
Apoptose/fisiologia , Células Epiteliais/patologia , Hipóxia/patologia , Falência Renal Crônica/patologia , Túbulos Renais/patologia , Animais , Camundongos , Receptor fas/análise
13.
J Clin Invest ; 103(11): 1561-70, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10359565

RESUMO

IL-1-stimulated mesenchymal cells model molecular mechanisms of inflammation. Binding of IL-1 to the type I IL-1 receptor (IL-1R) clusters a multi-subunit signaling complex at focal adhesion complexes. Since Rho family GTPases coordinately organize actin cytoskeleton and signaling to regulate cell phenotype, we hypothesized that the IL-1R signaling complex contained these G proteins. IL-1 stimulated actin stress fiber formation in serum-starved HeLa cells in a Rho-dependent manner and rapidly activated nucleotide exchange on RhoA. Glutathione S-transferase (GST) fusion proteins, containing either the full-length IL-1R cytosolic domain (GST-IL-1Rcd) or the terminal 68 amino acids of IL-1R required for IL-1-dependent signal transduction, specifically coprecipitated both RhoA and Rac-1, but not p21(ras), from Triton-soluble HeLa cell extracts. In whole cells, a small-molecular-weight G protein coimmunoprecipitated by anti-IL-1R antibody was a substrate for C3 transferase, which specifically ADP-ribosylates Rho GTPases. Constitutively activated RhoA, loaded with [gamma-32P]GTP, directly interacted with GST-IL-1Rcd in a filter-binding assay. The IL-1Rcd-RhoA interaction was functionally important, since a dominant inhibitory mutant of RhoA prevented IL-1Rcd-directed transcriptional activation of the IL-6 gene. Consistent with our previous data demonstrating that IL-1R-associated myelin basic protein (MBP) kinases are necessary for IL-1-directed gene expression, cellular incorporation of C3 transferase inhibited IL-1R-associated MBP kinase activity both in solution and in gel kinase assays. In summary, IL-1 activated RhoA, which was physically associated with IL-1Rcd and necessary for activation of cytosolic nuclear signaling pathways. These findings suggest that IL-1-stimulated, Rho-dependent cytoskeletal reorganization may cluster signaling molecules in specific architectures that are necessary for persistent cell activation in chronic inflammatory disease.


Assuntos
Toxinas Botulínicas , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Inflamação/metabolismo , Receptores de Interleucina-1/metabolismo , ADP Ribose Transferases/metabolismo , Actinas/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Extratos Celulares , Linhagem Celular , Precipitação Química , Citoesqueleto/fisiologia , Ativação Enzimática , Glutationa Transferase , Quinase 3 da Glicogênio Sintase , Células HeLa , Humanos , Interleucina-1/metabolismo , Interleucina-1/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Especificidade por Substrato , Proteínas rac de Ligação ao GTP , Proteína rhoA de Ligação ao GTP
14.
Kidney Int ; 55(5): 1740-9, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10231436

RESUMO

BACKGROUND: We have studied interleukin-1 (IL-1)-stimulated signals and gene expression in mesangial cells (MCs) to identify molecular mechanisms of MC activation, a process characteristic of glomerular inflammation. The JNK1 pathway has been implicated in cell fate decisions, and IL-1 stimulates the Jun N-terminal/stress-activated protein kinases (JNK1/SAPK). However, early postreceptor mechanisms by which IL-1 activates these enzymes remain unclear. Free arachidonic acid (AA) activates several protein kinases, and because IL-1 rapidly stimulates phospholipase A2 (PLA2) activity release AA, IL-1-induced activation of JNK1/SAPK may be mediated by AA release. METHODS: MCs were grown from collagenase-treated glomeruli, and JNK/SAPK activity in MC lysates was determined using an immunocomplex kinase assay. RESULT: Treatment of MCs with IL-1 alpha induced a time-dependent increase in JNK1/SAPK kinase activity, assessed by phosphorylation of the activating transcription factor-2 (ATF-2). Using similar incubation conditions, IL-1 also increased [3H]AA release from MCs. Pretreatment of MCs with aristolochic acid, a PLA2 inhibitor, concordantly reduced IL-1-regulated [3H]AA release and JNK1/SAPK activity, suggesting that cytosolic AA in part mediates IL-1-induced JNK1/SAPK activation. Addition of AA stimulated JNK1/SAPK activity in a time- and concentration-dependent manner. This effect was AA specific, as only AA and its precursor linoleic acid stimulated JNK1/SAPK activity. Other fatty acids failed to activate JNK1/SAPK. Pretreatment of MCs with specific inhibitors of AA oxidation by cyclooxygenase, lipoxygenase, and cytochrome P-450 epoxygenase had no effect on either IL-1- or AA-induced JNK1/SAPK activation. Furthermore, stimulation of MCs with the exogenous cyclooxygenase-, lipoxygenase-, phosphodiesterase-, and epoxygenase-derived arachidonate metabolites, in contrast to AA itself, did not activate JNK1/SAPK. CONCLUSION: We conclude that IL-1-stimulated AA release, in part, mediates stimulation of JNK1/SAPK activity and that AA activates JNK1/SAPK by a mechanism that does not require enzymatic oxygenation. JNK1 signaling pathway components may provide molecular switches that mediate structural rearrangements and biochemical processes characteristic of MC activation and could provide a novel target(s) for therapeutic intervention.


Assuntos
Ácido Araquidônico/metabolismo , Ácidos Aristolóquicos , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Mesângio Glomerular/enzimologia , Interleucina-1/farmacologia , Proteínas Quinases Ativadas por Mitógeno , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacologia , Animais , Ácido Araquidônico/farmacologia , Células Cultivadas , Dinoprostona/farmacologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ácidos Graxos Insaturados/farmacologia , Mesângio Glomerular/citologia , Mesângio Glomerular/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno , Leucotrienos/farmacologia , Peróxidos Lipídicos/farmacologia , Nefrite/enzimologia , Fenantrenos/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Fosfolipases A/metabolismo , Fosfolipases A2 , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Ácidos Esteáricos/farmacologia , Trítio , Vasoconstritores/farmacologia
15.
Lab Invest ; 78(7): 813-24, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9690559

RESUMO

Renal tubular atrophy predicts a poor prognosis in chronic renal failure, but the molecular mechanisms regulating this process remain unknown. Because the Fas apoptosis pathway has recently been implicated in disease pathogenesis and Fas is expressed in the kidney, we hypothesized that Fas-mediated apoptosis of renal tubule epithelial cells (RTC) contributes to tubular atrophy in chronic renal failure. In vivo, immunohistochemical analyses of renal sections from two murine models of progressive renal disease revealed coordinate increases in RTC Fas expression and apoptosis compared with tissue sections from age-matched control kidneys. Increased RTC apoptosis was not accompanied by compensatory hyperplasia, suggesting that RTC targeted for Fas-dependent apoptotic deletion contribute to tubular atrophy. These data are supported by in vitro studies showing that interleukin-1alpha and tumor necrosis factor-alpha, cytokines secreted in chronic renal failure, stimulated increases in Fas expression in cultured RTC. Both murine kidney cortex and RTC in culture demonstrated constitutive expression of transmembrane and soluble forms of RTC Fas ligand, features that are primarily restricted to lymphocytes and immune-privileged tissues and that have been previously unrecognized in RTC. Functional studies revealed that interleukin-1alpha-stimulated RTC Fas expression was accompanied by increased apoptosis, which was inhibited by blocking anti-Fas ligand antibodies. In contrast to the conventional paradigm, which holds that Fas-dependent apoptosis is initiated by the binding of lymphocyte Fas ligand to target cell Fas, our data suggest that up-regulated RTC Fas binds to Fas ligand on adjacent RTC, which then leads to RTC death by fratricide. We propose this pathway as an initiating mechanism of tubular atrophy.


Assuntos
Apoptose , Células Epiteliais/patologia , Genes p53 , Falência Renal Crônica/patologia , Falência Renal Crônica/fisiopatologia , Túbulos Renais/patologia , Receptor fas/biossíntese , Animais , Divisão Celular , Linhagem Celular Transformada , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Citometria de Fluxo , Humanos , Túbulos Renais/citologia , Túbulos Renais/fisiopatologia , Camundongos , Camundongos Transgênicos , Reação em Cadeia da Polimerase , Antígeno Nuclear de Célula em Proliferação/análise , Valores de Referência , Proteína Supressora de Tumor p53/biossíntese , Receptor fas/análise
16.
Ren Fail ; 20(4): 635-44, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9713883

RESUMO

Tumor lysis syndrome (TLS) is a critical illness with few treatment options. This report describes the clinical course of a patient with non-Hodgkin's lymphoma, who developed TLS and required renal replacement therapy. Institution of the standard therapeutic approach, intermittent hemodialysis, was not possible because of persistent hypotension. Instead, the patient was treated with a short course of continuous arteriovenous hemofiltration (CAVH) and conventional continuous arteriovenous hemodialysis (CAVHD) with dialysate flow rate of 1 L/h), which resulted in effective control of serum uric acid, potassium, urea nitrogen, phosphorus, and extracellular fluid volume. This case is in distinction to a previous report of TLS treatment with CAVHD using 4 L/h dialysate flow rate. We conclude that continuous renal replacement therapies with standard dialysate flow rates and replacement volumes should be considered for the treatment of TLS, particulary if the syndrome is accompanied by hypotension.


Assuntos
Injúria Renal Aguda/terapia , Hemofiltração , Diálise Renal/métodos , Síndrome de Lise Tumoral/terapia , Injúria Renal Aguda/etiologia , Feminino , Humanos , Linfoma não Hodgkin/tratamento farmacológico , Pessoa de Meia-Idade , Síndrome de Lise Tumoral/etiologia
17.
Am J Physiol ; 272(5 Pt 1): C1558-66, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9176147

RESUMO

Vascular smooth muscle cells (VSMC) contribute to the pathophysiology of hypertension through cell growth and contraction, and phospholipase C (PLC) is a critical effector enzyme in growth factor and vasoconstrictor signaling. There is indirect evidence that angiotensin II (ANG II) receptors are linked to the PLC-beta isoform signaling pathways. However, recent studies suggest that PLC-beta isoforms may not be expressed in VSMC. Our data demonstrate that in human aortic VSMC, PLC-beta 1 and PLC-gamma 1 proteins were detected by immunoblot analysis, and PLC-beta 1 mRNA was identified by reverse transcriptase-polymerase chain reaction in rat aortic VSMC. Incubation of permeabilized VSMC with anti-PLC-beta 1 or anti-Gq alpha antibodies inhibited ANG II-dependent inositol polyphosphate (IP) formation, while anti-PLC-gamma 1 antibodies did not inhibit ANG II-regulated IP formation. Conversely, anti-PLC-gamma 1 antibodies completely abolished platelet-derived growth factor (PDGF)-dependent IP generation, whereas anti-PLC-beta 1 antibodies had no effect on PDGF-induced PLC activation. Inhibition of tyrosine phosphorylation with genistein or herbimycin A did not diminish ANG II-stimulated IP formation or cytosolic free Ca2+ concentration transients, thereby confirming that ANG II signals via a PLC-gamma 1-independent mechanism. In summary, PLC-beta 1 and PLC-gamma 1 are expressed in human aortic VSMC, and PLC-beta 1 is the isoform that is critical for ANG II-regulated PLC signaling in these cells.


Assuntos
Angiotensina II/farmacologia , Isoenzimas/metabolismo , Músculo Liso Vascular/enzimologia , Fosfolipases Tipo C/metabolismo , Animais , Células Cultivadas , Ativação Enzimática , Proteínas de Ligação ao GTP/fisiologia , Humanos , Immunoblotting , Fosfatos de Inositol/biossíntese , Isoenzimas/genética , Músculo Liso Vascular/citologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Ratos , Transcrição Gênica , Fosfolipases Tipo C/genética
18.
Am J Physiol ; 270(4 Pt 2): F682-90, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8967347

RESUMO

Vasoconstrictor hormones contribute to the pathogenesis of hypertension through intracellular signals that stimulate vascular smooth muscle (VSMC) contraction and/or growth. We previously showed that the glucocorticoid dexamethasone (DEX) inhibited angiotensin II-stimulated inositol trisphosphate (IP3) formation in VSMC, but the mechanism of inhibition is not known. Because glucocorticoids stimulate the expression of annexins and annexin II potently binds phosphoinositides, the role of DEX and annexin II in VSMC G protein-coupled phosphoinositide hydrolysis was investigated. DEX incubation blunted increases in guanosine 5'-O-(3-thiotriphosphate) (GTP gamma S)-stimulated IP3 generation and angiotensin II-induced intracellular Ca2+ mobilization but stimulated elevations in VSMC annexin II content. VSMC incubation with exogenous purified annexin II resulted in concentration-dependent decreases in GTP gamma S-stimulated IP3 formation. In DEX-treated cells, exogenous annexin II did not further diminish GTP gamma S-stimulated IP3 formation, suggesting that endogenous annexin II may be a mediator of DEX-induced inhibition of G protein-coupled IP3 generation. These data represent the first direct evidence of G protein-dependent phosphoinositide hydrolysis regulation by glucocorticoids or annexins. We speculate that annexin II may play a role in the pathogenesis of hypertension through stimulation of VSMC growth.


Assuntos
Anexina A2/farmacologia , Aorta/metabolismo , Proteínas de Ligação ao GTP/fisiologia , Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Músculo Liso Vascular/metabolismo , Angiotensina II/farmacologia , Animais , Anexina A2/fisiologia , Aorta/citologia , Transporte Biológico/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Dexametasona/farmacologia , Ativação Enzimática/efeitos dos fármacos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Inositol 1,4,5-Trifosfato/biossíntese , Músculo Liso Vascular/citologia , Ratos , Ratos Sprague-Dawley , Fosfolipases Tipo C/metabolismo
19.
Am J Physiol ; 267(5 Pt 1): C1239-45, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7977687

RESUMO

Angiotensin II (ANG II) stimulates proximal tubule sodium transport by decreasing adenylyl cyclase activity. The role of ANG II-dependent phospholipase C is less certain. To determine the contribution of phospholipase C and adenylyl cyclase to apical (AP) ANG II-dependent sodium transport, unidirectional (AP to basolateral) 22Na flux was measured in rat proximal tubule cells cultured on permeable supports. AP ANG II (100 nM)-dependent sodium flux was prevented by preincubation with concentrations of the phospholipase C inhibitor U-73122 (1 microM) that blocked ANG II-dependent inositol phosphate formation. AP ANG II-dependent sodium flux was also abolished by preincubation with the intracellular calcium mobilization inhibitor 3,4,5-trimethoxybenzoic acid 8-(diethylamino)octyl ester (TMB-8), further suggesting that ANG II-dependent sodium transport was mediated by inositol phosphates. Neither U-73122 nor TMB-8 prevented ANG II-dependent adenosine 3',5'-cyclic monophosphate (cAMP) decreases. Incubation with dibutyryl cAMP (10 microM) or forskolin (10 microM) prevented ANG II-dependent sodium flux as well as ANG II-dependent inositol phosphate formation. In conclusion, ANG II-dependent proximal tubule sodium transport in cultured cells was transduced by phospholipase C and adenylyl cyclase. The adenylyl cyclase effect on ANG II-dependent sodium transport was mediated by phospholipase C.


Assuntos
Angiotensina II/fisiologia , AMP Cíclico/fisiologia , Túbulos Renais Proximais/metabolismo , Sódio/metabolismo , Fosfolipases Tipo C/metabolismo , Adenilil Ciclases/metabolismo , Animais , Transporte Biológico/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Membrana Celular/metabolismo , Células Cultivadas , Estrenos/farmacologia , Ácido Gálico/análogos & derivados , Ácido Gálico/farmacologia , Túbulos Renais Proximais/citologia , Masculino , Pirrolidinonas/farmacologia , Ratos , Ratos Sprague-Dawley , Fosfolipases Tipo C/antagonistas & inibidores
20.
Kidney Int ; 46(3): 675-82, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7996788

RESUMO

Vascular tone is maintained by both angiotensin II (Ang II) and glucocorticoids, but the effect of glucocorticoids on Ang II function in vascular smooth muscle cells (VSMC) is unclear. To determine the direct influence of glucocorticoids on VSMC Ang II receptor function, the effects of dexamethasone on Ang II receptor binding, Ang II-induced phospholipase C (PLC) activation, and Ang II-dependent cell growth were studied in cultured rat VSMC. Dexamethasone caused concentration- and time-dependent increases in Ang II binding which were prevented by glucocorticoid receptor inhibition with RU 38486. Dexamethasone-induced enhancement of Ang II binding resulted from increased AT1 receptors, as indicated by Northern blot analysis and competitive binding assays. Despite causing increased Ang II receptor number, dexamethasone preincubation prevented Ang II-induced PLC activation, as indicated by phosphatidylinositol 4,5-bisphosphate degradation and inositol trisphosphate formation. When PLC activity was directly measured in VSMC soluble and membrane fractions, Ang II receptor activation caused decreased soluble and increased membrane PLC activity, consistent with the interpretation that Ang II caused cytosol-to-membrane PLC translocation. The effect of Ang II on PLC translocation was prevented by dexamethasone preincubation. Finally, prolonged incubation with dexamethasone and Ang II had additive effects on VSMC hypertrophy. In conclusion, glucocorticoids directly altered Ang II function in VSMC by causing increased Ang II receptor number, Ang II receptor/PLC uncoupling, and enhanced Ang II-dependent hypertrophy.


Assuntos
Angiotensina II/metabolismo , Dexametasona/farmacologia , Músculo Liso Vascular/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Divisão Celular , Células Cultivadas , Relação Dose-Resposta a Droga , Ativação Enzimática , Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Mifepristona/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Fosfatidilinositol 4,5-Difosfato , Fosfatos de Fosfatidilinositol/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Angiotensina/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA