Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Am J Physiol Renal Physiol ; 323(3): F272-F287, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35862649

RESUMO

Wilms' tumor interacting protein (Wtip) has been implicated in cell junction assembly and cell differentiation and interacts with proteins in the podocyte slit diaphragm, where it regulates podocyte phenotype. To define Wtip expression and function in the kidney, we created a Wtip-deleted mouse model using ß-galactosidase-neomycin (ß-geo) gene trap technology. Wtip gene trap mice were embryonic lethal, suggesting additional developmental roles outside kidney function. Using ß-geo heterozygous and normal mice, Wtip expression was identified in the developing kidneys, heart, and eyes. In the kidney, expression was restricted to podocytes, which appeared initially at the capillary loop stage coinciding with terminal podocyte differentiation. Heterozygous mice had an expected lifespan and showed no evidence of proteinuria or glomerular pathology. However, heterozygous mice were more susceptible to glomerular injury than wild-type littermates and developed more significant and prolonged proteinuria in response to lipopolysaccharide or adriamycin. In normal human kidneys, WTIP expression patterns were consistent with observations in mice and were lost in glomeruli concurrent with loss of synaptopodin expression in disease. Mechanistically, we identified the Rho guanine nucleotide exchange factor 12 (ARHGEF12) as a binding partner for WTIP. ARHGEF12 was expressed in human podocytes and formed high-affinity interactions through their LIM- and PDZ-binding domains. Our findings suggest that Wtip is essential for early murine embryonic development and maintaining normal glomerular filtration barrier function, potentially regulating slit diaphragm and foot process function through Rho effector proteins.NEW & NOTEWORTHY This study characterized dynamic expression patterns of Wilms' tumor interacting protein (Wtip) and demonstrates the novel role of Wtip in murine development and maintenance of the glomerular filtration barrier.


Assuntos
Nefropatias , Podócitos , Tumor de Wilms , Animais , Proteínas Correpressoras/metabolismo , Proteínas do Citoesqueleto/metabolismo , Feminino , Barreira de Filtração Glomerular , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Nefropatias/metabolismo , Glomérulos Renais/metabolismo , Camundongos , Podócitos/metabolismo , Gravidez , Proteinúria/genética , Proteinúria/metabolismo , Tumor de Wilms/metabolismo
2.
Curr Opin Nephrol Hypertens ; 30(3): 317-323, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33767059

RESUMO

PURPOSE OF REVIEW: Allelic variants in the gene for apolipoprotein L1 (APOL1), found only in individuals of African ancestry, explain a majority of the excess risk of kidney disease in African Americans. However, a clear understanding how the disease-associated APOL1 variants cause kidney injury and the identity of environmental stressors that trigger the injury process have not been determined. RECENT FINDINGS: Basic mechanistic studies of APOL1 biochemistry and cell biology, bolstered by new antibody reagents and inducible pluripotent stem cell-derived cell systems, have focused on the cytotoxic effect of the risk variants when APOL1 gene expression is induced. Since the APOL1 variants evolved to alter a key protein-protein interaction with the trypanosome serum resistance-associated protein, additional studies have begun to address differences in APOL1 interactions with other proteins expressed in podocytes, including new observations that APOL1 variants may alter podocyte cytoskeleton dynamics. SUMMARY: A unified mechanism of pathogenesis for the various APOL1 nephropathies still remains unclear and controversial. As ongoing studies have consistently implicated the pathogenic gain-of-function effects of the variant proteins, novel therapeutic development inhibiting the synthesis or function of APOL1 proteins is moving toward clinical trials.


Assuntos
Apolipoproteína L1 , Nefropatias , Apolipoproteína L1/genética , Suscetibilidade a Doenças , Predisposição Genética para Doença , Humanos , Nefropatias/genética , Nefropatias/terapia , Podócitos
3.
Clin Transplant ; 35(4): e14234, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33511679

RESUMO

Variants in apolipoprotein L1 (APOL1) gene are associated with nondiabetic kidney diseases in black subjects and reduced kidney transplant graft survival. Living and deceased black kidney donors (n = 107) were genotyped for APOL1 variants. To determine whether allografts from high-risk APOL1 donors have reduced podocyte densities contributing to allograft failure, we morphometrically estimated podocyte number, glomerular volume, and podocyte density. We compared allograft loss and eGFR trajectories stratified by APOL1 high-risk and low-risk genotypes. Demographic characteristics were similar in high-risk (n = 16) and low-risk (n = 91) donors. Podocyte density was significantly lower in high-risk than low-risk donors (108 ± 26 vs 127 ± 40 podocytes/106 um3 , P = .03). Kaplan-Meier graft survival (high-risk 61% vs. low-risk 91%, p-value = 0.049) and multivariable Cox models (hazard ratio = 2.6; 95% CI, 0.9-7.8) revealed higher graft loss in recipients of APOL1 high-risk allografts over 48 months. More rapid eGFR decline was seen in recipients of high-risk APOL1 allografts (P < .001). At 60 months, eGFR was 27 vs. 51 mL/min/1.73 min2 in recipients of APOL1 high-risk vs low-risk kidney allografts, respectively. Kidneys from high-risk APOL1 donors had worse outcomes versus low-risk APOL1 genotypes. Lower podocyte density in kidneys from high-risk APOL1 donors may increase susceptibility to CKD from subsequent stresses in both the recipients and donors.


Assuntos
Apolipoproteína L1 , Transplante de Rim , Podócitos , Aloenxertos , Apolipoproteína L1/genética , Genótipo , Sobrevivência de Enxerto , Humanos , Rim
4.
BMC Med Genet ; 21(1): 110, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32434471

RESUMO

BACKGROUND: Black women in the United States and Africa are at an increased risk for preeclampsia. Allelic variants in the gene for apolipoprotein LI, APOL1, are found only in populations of African ancestry, and have been shown to contribute significant risk for kidney disease. Recent studies suggest these APOL1 variants also may contribute risk for preeclampsia. METHODS: The association of preeclampsia with carriage of APOL1 risk alleles was evaluated in a case-control study of deliveries from black women at a single center in Cleveland, Ohio that included gross and histopathologic evaluations of placental tissues (395 cases and 282 controls). Using logistic regression models, associations between fetal APOL1 genotype and preeclampsia were evaluated using several case definitions based on prematurity and severity of preeclampsia, with uncomplicated term pregnancies as controls. Associations between APOL1 genotype and pathological features were also examined. RESULTS: The infant APOL1 genotype was significantly associated with preeclampsia in a dominant inheritance pattern with odds ratio of 1.41 (P=0.029, 95% CI 1.037, 1.926). Stratifying preeclampsia cases by preterm birth, significant associations were detected for both recessive (O.R.=1.70, P=0.038) and additive (O.R.=1.33, P=0.028) inheritance patterns. APOL1 genotype, however, was not significantly associated with pathological changes or other perinatal observations. CONCLUSIONS: Preeclampsia appears to be another disease associated with APOL1 variants, however, further studies are needed to increase confidence in the mode of inheritance. By understanding the association of APOL1 variants with preeclampsia, genetic screening tests for APOL1 may be useful to predict at-risk pregnancies and targeted interventions may be developed to improve pregnancy outcomes.


Assuntos
Apolipoproteína L1/genética , Negro ou Afro-Americano/genética , Predisposição Genética para Doença , Variação Genética , Genótipo , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/genética , Alelos , Estudos de Casos e Controles , Feminino , Frequência do Gene , Estudos de Associação Genética , Testes Genéticos , Humanos , Modelos Logísticos , Masculino , Modelos Genéticos , Razão de Chances , Placenta/metabolismo , Gravidez
5.
Am J Physiol Renal Physiol ; 316(1): F1-F8, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30332315

RESUMO

The mechanism that explains the association of APOL1 variants with nondiabetic kidney diseases in African Americans remains unclear. Kidney disease risk is inherited as a recessive trait, and many studies investigating the intracellular function of APOL1 have indicated the APOL1 variants G1 and G2 are associated with cytotoxicity. Whether cytotoxicity results from the absence of a protective effect conferred by the G0 allele or is induced by a deleterious effect of variant allele expression has not be conclusively established. A central issue hampering basic biology studies is the lack of model systems that authentically replicate APOL1 expression patterns. APOL1 is present in humans and a few other primates and appears to have important functions in the kidney, as the kidney is the primary target for disease associated with the genetic variance. There have been no studies to date assessing the function of untagged APOL1 protein under native expression in human or primate kidney cells, and no studies have examined the heterozygous state, a disease-free condition in humans. A second major issue is the chronic kidney disease (CKD)-associated APOL1 variants are conditional mutations, where the disease-inducing function is only evident under the appropriate environmental stimulus. In addition, it is possible there may be more than one mechanism of pathogenesis that is dependent on the nature of the stressor or other genetic variabilities. Studies addressing the function of APOL1 and how the CKD-associated APOL1 variants cause kidney disease are challenging and remain to be fully investigated under conditions that faithfully model known human genetics and physiology.


Assuntos
Apolipoproteína L1/genética , Mutação com Ganho de Função , Mutação com Perda de Função , Polimorfismo Genético , Insuficiência Renal Crônica/genética , Negro ou Afro-Americano/genética , Animais , Apolipoproteína L1/metabolismo , Interação Gene-Ambiente , Predisposição Genética para Doença , Hereditariedade , Humanos , Fenótipo , Podócitos/metabolismo , Podócitos/patologia , Insuficiência Renal Crônica/etnologia , Insuficiência Renal Crônica/patologia , Fatores de Risco
6.
J Am Soc Nephrol ; 29(3): 869-879, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29180397

RESUMO

Coding variants in the APOL1 gene are associated with kidney diseases in African ancestral populations; yet, the underlying biologic mechanisms remain uncertain. Variant-dependent autophagic and cytotoxic cell death have been proposed as pathogenic pathways mediating kidney injury. To examine this possibility, we conditionally expressed APOL1-G0 (reference), -G1, and -G2 (variants) using a tetracycline-regulated system in HEK293 cells. Autophagy was monitored biochemically and cell death was measured using multiple assays. We measured intracellular Na+ and K+ content with atomic absorption spectroscopy and APOL1-dependent currents with whole-cell patch clamping. Neither reference nor variant APOL1s induced autophagy. At high expression levels, APOL1-G0, -G1, and -G2 inserted into the plasma membrane and formed pH-sensitive cation channels, causing collapse of cellular Na+ and K+ gradients, phosphorylation of p38 mitogen-activated protein kinase, and cell death, without variant-dependent differences. APOL1-G0 and -G2 exhibited similar channel properties in whole-cell patch clamp experiments. At low expression levels, neither reference nor variant APOL1s localized on the plasma membrane, Na+ and K+ gradients were maintained, and cells remained viable. Our results indicate that APOL1-mediated pore formation is critical for the trypanolytic activity of APOL1 and drives APOL1-mediated cytotoxicity in overexpression systems. The absence of cytotoxicity at physiologic expression levels suggests variant-dependent intracellular K+ loss and cytotoxicity does not drive kidney disease progression.


Assuntos
Apolipoproteína L1/genética , Autofagia/genética , Variação Genética , Nefropatias/genética , Potássio/metabolismo , Sódio/metabolismo , Apolipoproteína L1/fisiologia , Cálcio/metabolismo , Membrana Celular/fisiologia , Expressão Gênica/efeitos dos fármacos , Genótipo , Células HEK293 , Humanos , Canais Iônicos , Técnicas de Patch-Clamp , Fosforilação , Tetraciclina/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
7.
J Am Soc Nephrol ; 29(5): 1501-1512, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29572404

RESUMO

Background FSGS is a pattern of podocyte injury that leads to loss of glomerular function. Podocytes support other podocytes and glomerular capillary structure, oppose hemodynamic forces, form the slit diaphragm, and have mechanical properties that permit these functions. However, the biophysical characteristics of glomeruli and podocytes in disease remain unclear.Methods Using microindentation, atomic force microscopy, immunofluorescence microscopy, quantitative RT-PCR, and a three-dimensional collagen gel contraction assay, we studied the biophysical and structural properties of glomeruli and podocytes in chronic (Tg26 mice [HIV protein expression]) and acute (protamine administration [cytoskeletal rearrangement]) models of podocyte injury.Results Compared with wild-type glomeruli, Tg26 glomeruli became progressively more deformable with disease progression, despite increased collagen content. Tg26 podocytes had disordered cytoskeletons, markedly abnormal focal adhesions, and weaker adhesion; they failed to respond to mechanical signals and exerted minimal traction force in three-dimensional collagen gels. Protamine treatment had similar but milder effects on glomeruli and podocytes.Conclusions Reduced structural integrity of Tg26 podocytes causes increased deformability of glomerular capillaries and limits the ability of capillaries to counter hemodynamic force, possibly leading to further podocyte injury. Loss of normal podocyte mechanical integrity could injure neighboring podocytes due to the absence of normal biophysical signals required for podocyte maintenance. The severe defects in podocyte mechanical behavior in the Tg26 model may explain why Tg26 glomeruli soften progressively, despite increased collagen deposition, and may be the basis for the rapid course of glomerular diseases associated with severe podocyte injury. In milder injury (protamine), similar processes occur but over a longer time.


Assuntos
Fenômenos Biofísicos , Citoesqueleto/fisiologia , Glomerulonefrite/fisiopatologia , Nefrose Lipoide/fisiopatologia , Podócitos/fisiologia , Animais , Adesão Celular , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Módulo de Elasticidade , Glomerulonefrite/genética , Glomerulonefrite/patologia , HIV/genética , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Camundongos , Camundongos Transgênicos , Microscopia de Força Atômica , Microscopia de Fluorescência , Nefrose Lipoide/induzido quimicamente , Nefrose Lipoide/patologia , Paxilina/metabolismo , Podócitos/patologia , Protaminas , Reação em Cadeia da Polimerase em Tempo Real
10.
J Am Soc Nephrol ; 27(12): 3600-3610, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27026370

RESUMO

APOL1 risk variants are associated with kidney disease in blacks, but the mechanisms of renal injury associated with APOL1 risk variants are unknown. Because APOL1 is unique to humans and some primates, we created transgenic (Tg) mice using the promoter of nephrin-encoding Nphs1 to express the APOL1 reference sequence (G0) or the G2 risk variant in podocytes, establishing Tg lines with a spectrum of APOL1 expression levels. Podocytes from Tg-G0 and Tg-G2 mice did not undergo necrosis, apoptosis, or autophagic cell death in vivo, even in lines with highly expressed transgenes. Further, Tg-G0 and Tg-G2 mice did not develop kidney pathology, proteinuria, or azotemia as of 300 days of age. However, by 200 days of age, Tg-G2 mice had significantly lower podocyte density than age-matched WT and Tg-G0 mice had, a difference that was not evident at weaning. Notably, a pregnancy-associated phenotype that encompassed eclampsia, preeclampsia, fetal/neonatal deaths, and small litter sizes occurred in some Tg-G0 mice and more severely in Tg-G2 mice. Similar to human placenta, placentas of Tg mice expressed APOL1. Overall, these results suggest podocyte depletion could predispose individuals with APOL1 risk genotypes to kidney disease in response to a second stressor, and add to other published evidence associating APOL1 expression with preeclampsia.


Assuntos
Apolipoproteínas/genética , Nefropatias/genética , Lipoproteínas HDL/genética , Pré-Eclâmpsia/genética , Animais , Apolipoproteína L1 , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Transgênicos , Podócitos/fisiologia , Gravidez
11.
J Biol Chem ; 290(1): 338-49, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25411248

RESUMO

Glomerular podocytes are highly specialized terminally differentiated cells that act as a filtration barrier in the kidney. Mutations in the actin-binding protein, α-actinin 4 (ACTN4), are linked to focal segmental glomerulosclerosis (FSGS), a chronic kidney disease characterized by proteinuria. Aberrant activation of NF-κB pathway in podocytes is implicated in glomerular diseases including proteinuria. We demonstrate here that stable knockdown of ACTN4 in podocytes significantly reduces TNFα-mediated induction of NF-κB target genes, including IL-1ß and NPHS1, and activation of an NF-κB-driven reporter without interfering with p65 nuclear translocation. Overexpression of ACTN4 and an actin binding-defective variant increases the reporter activity. In contrast, an FSGS-linked ACTN4 mutant, K255E, which has increased actin binding activity and is predominantly cytoplasmic, fails to potentiate NF-κB activity. Mechanistically, IκBα blocks the association of ACTN4 and p65 in the cytosol. In response to TNFα, both NF-κB subunits p65 and p50 translocate to the nucleus, where they bind and recruit ACTN4 to their targeted promoters, IL-1ß and IL-8. Taken together, our data identify ACTN4 as a novel coactivator for NF-κB transcription factors in podocytes. Importantly, this nuclear function of ACTN4 is independent of its actin binding activity in the cytoplasm.


Assuntos
Actinina/genética , NF-kappa B/genética , Podócitos/metabolismo , Transcrição Gênica , Actinina/antagonistas & inibidores , Actinina/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Linhagem Celular Transformada , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Podócitos/citologia , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
13.
J Am Soc Nephrol ; 25(3): 634-44, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24231663

RESUMO

Polymorphisms in APOL1 are associated with CKD, including HIV-related CKD, in individuals of African ancestry. The apolipoprotein L1 (APOL1) protein circulates and is localized in kidney cells, but the contribution of APOL1 location to CKD pathogenesis is unclear. We examined associations of plasma APOL1 levels with plasma cytokine levels, dyslipidemia, and APOL1 genotype in a nested case-control study (n=270) of HIV-infected African Americans enrolled in a multicenter prospective observational study. Patients were designated as having CKD when estimated GFR (eGFR) decreased to <60 ml/min per 1.73 m(2) (eGFR<60 cohort) or protein-to-creatinine ratios became >3.5 g/g (nephrotic proteinuria cohort). Circulating APOL1 levels did not associate with APOL1 genotype, CKD status, or levels of proinflammatory cytokines, but did correlate with fasting cholesterol, LDL cholesterol, and triglyceride levels. At ascertainment, CKD-associated polymorphisms (risk variants) in APOL1 associated with the eGFR<60 cohort, but not the nephrotic-range proteinuria cohort. Of note, in both the eGFR<60 and nephrotic proteinuria cohorts, CKD cases with two APOL1 risk variants had significant declines in eGFR over a median of 4 years compared with individuals with one or no risk variants. APOL1 risk genotype was not associated with changes in proteinuria. Higher circulating proinflammatory cytokine levels were independently associated with CKD but not APOL1 genotype. In conclusion, the function of variant APOL1 proteins derived from circulation or synthesized in the kidney, but not the level of circulating APOL1, probably mediates APOL1-associated kidney disease in HIV-infected African Americans.


Assuntos
Nefropatia Associada a AIDS/sangue , Apolipoproteínas/sangue , Lipoproteínas HDL/sangue , Insuficiência Renal Crônica/sangue , Nefropatia Associada a AIDS/genética , Adulto , Negro ou Afro-Americano/genética , Apolipoproteína L1 , Apolipoproteínas/genética , Estudos de Casos e Controles , Citocinas/sangue , Progressão da Doença , Dislipidemias/sangue , Feminino , Genótipo , Taxa de Filtração Glomerular , Humanos , Lipoproteínas HDL/genética , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Fenótipo , Ensaios Clínicos Controlados Aleatórios como Assunto , Insuficiência Renal Crônica/genética
14.
Clin Infect Dis ; 59(9): e96-138, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25234519

RESUMO

It is important to realize that guidelines cannot always account for individual variation among patients. They are not intended to supplant physician judgment with respect to particular patients or special clinical situations. IDSA considers adherence to these guidelines to be voluntary, with the ultimate determination regarding their application to be made by the physician in the light of each patient's individual circumstances.


Assuntos
Infecções por HIV/complicações , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/terapia , Humanos , Transplante de Rim , Estados Unidos
15.
bioRxiv ; 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38948788

RESUMO

RATIONALE: Early steps in glomerular injury are poorly understood in collagen IV nephropathies. OBJECTIVES: We characterized structural, functional, and biophysical properties of glomerular capillaries and podocytes in Col4α3-/- mice and analyzed kidney cortex transcriptional profiles at various disease stages. We investigated the effects of TUDCA (suppresses ER stress) on these parameters and used human FSGS transcriptomic data to identify pathways rescued by TUDCA. FINDINGS: In Col4α3-/- mice, podocyte injury develops by 3 months, with maximum glomerular deformability and 40% podocyte loss at 4 months. This period is followed is followed by glomerular capillary stiffening, proteinuria, reduced renal function, inflammatory infiltrates, and fibrosis. Bulk RNA sequencing at sequential time points revealed progressive increases in inflammatory and injury gene expression, and activation of the TNF pathway. Mapping Podocyte-enriched genes from FSGS patients to mice showed that TUDCA, which mitigated renal injury suppressed molecular pathways associated with podocyte stress, hypertrophy and tubulo-interstitial injury. CONCLUSIONS: Col4α3-/- nephropathy progresses in two phases. The first is characterized by podocytopathy, increased glomerular capillary deformability and accelerated podocyte loss, and the second by increased capillary wall stiffening and renal inflammatory and profibrotic pathway activation. The response of podocytes to TUDCA treatment provides insights into signaling pathways in Alport and related nephropathies.

16.
J Biol Chem ; 287(15): 12027-35, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22351778

RESUMO

Mutations in α-actinin 4 (ACTN4) are linked to familial forms of focal segmental glomerulosclerosis (FSGS), a kidney disease characterized by proteinuria due to podocyte injury. The mechanisms underlying ACTN4 mutant-associated FSGS are not completely understood. Although α-actinins are better known to cross-link actin filaments and modulate cytoskeletal organization, we have previously shown that ACTN4 interacts with transcription factors including estrogen receptor and MEF2s and potentiates their transcriptional activity. Nuclear receptors including retinoic acid receptor (RAR) have been proposed to play a protective role in podocytes. We show here that ACTN4 interacts with and enhances transcriptional activation by RARα. In addition, FSGS-linked ACTN4 mutants not only mislocalized to the cytoplasm, but also lost their ability to associate with nuclear receptors. Consequently, FSGS-linked ACTN4 mutants failed to potentiate transcriptional activation by nuclear hormone receptors in podocytes. In addition, overexpression of these mutants suppressed the transcriptional activity mediated by endogenous wild-type ACTN4 possibly by a cytoplasmic sequestration mechanism. Our data provide the first link between FSGS-linked ACTN4 mutants and transcriptional activation by nuclear receptor such as RARα and peroxisome proliferator-activated receptor γ.


Assuntos
Actinina/genética , Glomerulosclerose Segmentar e Focal/genética , Mutação de Sentido Incorreto , Receptores do Ácido Retinoico/metabolismo , Transcrição Gênica , Actinina/metabolismo , Linhagem Celular , Humanos , Proteínas Mutantes/metabolismo , PPAR gama/metabolismo , Podócitos/metabolismo , Ligação Proteica , Transporte Proteico , Receptores do Ácido Retinoico/agonistas , Receptores do Ácido Retinoico/genética , Proteínas Recombinantes/metabolismo , Receptor alfa de Ácido Retinoico , Ativação Transcricional , Tretinoína/fisiologia
17.
Exp Cell Res ; 318(10): 1075-85, 2012 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-22472346

RESUMO

The glomerular capillary wall, composed of endothelial cells, the glomerular basement membrane and the podocytes, is continually subjected to hemodynamic force arising from tractional stress due to blood pressure and shear stress due to blood flow. Exposure of glomeruli to abnormal hemodynamic force such as hyperfiltration is associated with glomerular injury and progressive renal disease, and the conversion of mechanical stimuli to chemical signals in the regulation of the process is poorly understood in podocytes. By examining DNA fragmentation, apoptotic nuclear changes and cytochrome c release, we found that shear stress induced cell apoptosis in cultured podocytes. Meanwhile, podocytes exposed to shear stress also stimulated c-Src phosphorylation, phospholipase D (PLD) activation and mammalian target of rapamycin (mTOR) signaling. Using the antibodies against c-Src, PLD(1), and PLD(2) to perform reciprocal co-immunoprecipitations and in vitro PLD activity assay, our data indicated that c-Src interacted with and activated PLD(1) but not PLD(2). The inhibition of shear stress-induced c-Src phosphorylation by PP(2) (a specific inhibitor of c-Src kinase) resulted in reduced PLD activity. Phosphatidic acid, produced by shear stress-induced PLD activation, stimulated mTOR signaling, and caused podocyte hypertrophy and apoptosis.


Assuntos
Apoptose , Fosfolipase D/metabolismo , Podócitos/fisiologia , Proteínas Tirosina Quinases/metabolismo , Estresse Fisiológico , Serina-Treonina Quinases TOR/metabolismo , Animais , Proteína Tirosina Quinase CSK , Células Cultivadas , Camundongos , Fosfatidilcolinas/metabolismo , Fosforilação , Podócitos/enzimologia , Podócitos/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Resistência ao Cisalhamento , Transdução de Sinais , Quinases da Família src
20.
Neonatology ; 120(4): 532-536, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37062283

RESUMO

BACKGROUND: Hypertensive disorders of pregnancy cause fetal growth restriction and increased maternal morbidity and mortality, especially in women of African ancestry. Recently, preeclampsia risk was associated with polymorphisms in the apolipoprotein L1 (APOL1) gene in women of African ancestry. OBJECTIVES: We assessed APOL1 genotype effects on pregnancies with and without preeclampsia. METHOD: We conducted an unmatched case-control study of 1,358 mother-infant pairs from two independent cohorts of black women. RESULTS: Term preeclampsia cases with high-risk APOL1 genotypes were more likely to be small for gestational age compared to APOL1 low-risk term cases (odds ratio [OR] 2.8) and APOL1 high-risk controls (OR 5.5). Among preterm pregnancies, fetal APOL1 genotype was associated with preeclampsia. CONCLUSIONS: Fetal APOL1 genotype was associated with preeclampsia in preterm infants and with altered fetal growth in term infants. This may indicate APOL1 genotype impacts a spectrum of pregnancy complications mediated by a common pathophysiological event of placental insufficiency.


Assuntos
Pré-Eclâmpsia , Humanos , Feminino , Lactente , Recém-Nascido , Gravidez , Pré-Eclâmpsia/genética , Apolipoproteína L1/genética , Retardo do Crescimento Fetal/genética , Estudos de Casos e Controles , Idade Gestacional , Placenta , Recém-Nascido Prematuro , Genótipo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa