Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Annu Rev Med ; 74: 385-400, 2023 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-36706748

RESUMO

In 2020, the nephrology community formally interrogated long-standing race-based clinical algorithms used in the field, including the kidney function estimation equations. A comprehensive understanding of the history of kidney function estimation and racial essentialism is necessary to understand underpinnings of the incorporation of a Black race coefficient into prior equations. We provide a review of this history, as well as the considerations used to develop race-free equations that are a guidepost for a more equity-oriented, scientifically rigorous future for kidney function estimation and other clinical algorithms and processes in which race may be embedded as a variable.


Assuntos
Rim , Grupos Raciais , Humanos , Rim/fisiologia , População Negra
2.
BMC Infect Dis ; 22(1): 591, 2022 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-35787257

RESUMO

BACKGROUND: Microalbuminuria is an independent risk factor for cardiovascular and kidney disease and a predictor of end organ damage, both in the general population and in persons with HIV (PWH). Microalbuminuria is also an important risk factor for mortality in PWH treated with antiretroviral therapy (ART). In the ongoing Renal Risk Reduction (R3) study in Nigeria, we identified a high prevalence of microalbuminuria confirmed by two measurements 4-8 weeks apart in ART-experienced, virologically suppressed PWH. Although Stage 1 or 2 hypertension and exposure to potentially nephrotoxic antiretroviral medications were common in R3 participants, other traditional risk factors for albuminuria and kidney disease, including diabetes, APOL1 high-risk genotype, and smoking were rare. Co-infection with endemic pathogens may also be significant contributors to albuminuria, but co-infections were not evaluated in the R3 study population. METHODS: In Aim 1, we will cross-sectionally compare the prevalence of albuminuria and established kidney disease risk factors in a cohort of PWH to age- and sex-matched HIV-negative adults presenting for routine care at the Aminu Kano Teaching Hospital in Kano, Nigeria. We will leverage stored specimens from 2500 R3 participants and enroll an additional 500 PLWH recently initiated on ART (≤ 24 months) and 750 age- and sex-matched HIV-negative adults to determine the contribution of HIV, hypertension, and other comorbid medical conditions to prevalent albuminuria. In Aim 2, we will follow a cohort of 1000 HIV-positive, ART-treated and 500 HIV-negative normoalbuminuric adults for 30 months to evaluate the incidence and predictors of albuminuria. DISCUSSION: The findings from this study will support the development of interventions to prevent or address microalbuminuria in PWH to reduce kidney and cardiovascular morbidity and mortality. Such interventions might include more intensive monitoring and treatment of traditional risk factors, the provision of renin-angiotensin aldosterone system or sodium-glucose cotransporter-2 inhibitors, consideration of changes in ART regimen, and screening and treatment for relevant co-infections.


Assuntos
Coinfecção , Diabetes Mellitus Tipo 2 , Infecções por HIV , Hipertensão , Nefropatias , Inibidores do Transportador 2 de Sódio-Glicose , Adulto , Albuminúria/epidemiologia , Albuminúria/etiologia , Apolipoproteína L1 , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Humanos , Hipertensão/complicações , Hipertensão/epidemiologia , Nigéria/epidemiologia , Inibidores do Transportador 2 de Sódio-Glicose/uso terapêutico
3.
J Am Soc Nephrol ; 32(7): 1765-1778, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33853887

RESUMO

BACKGROUND: APOL1 variants contribute to the markedly higher incidence of ESKD in Blacks compared with Whites. Genetic testing for these variants in patients with African ancestry who have nephropathy is uncommon, and no specific treatment or management protocol for APOL1-associated nephropathy currently exists. METHODS: A multidisciplinary, racially diverse group of 14 experts and patient advocates participated in a Delphi consensus process to establish practical guidance for clinicians caring for patients who may have APOL1-associated nephropathy. Consensus group members took part in three anonymous voting rounds to develop consensus statements relating to the following: (1) counseling, genotyping, and diagnosis; (2) disease awareness and education; and (3) a vision for management of APOL1-associated nephropathy in a future when treatment is available. A systematic literature search of the MEDLINE and Embase databases was conducted to identify relevant evidence published from January 1, 2009 to July 14, 2020. RESULTS: The consensus group agreed on 55 consensus statements covering such topics as demographic and clinical factors that suggest a patient has APOL1-associated nephropathy, as well as key considerations for counseling, testing, and diagnosis in current clinical practice. They achieved consensus on the need to increase awareness among key stakeholders of racial health disparities in kidney disease and of APOL1-associated nephropathy and on features of a successful education program to raise awareness among the patient community. The group also highlighted the unmet need for a specific treatment and agreed on best practice for management of these patients should a treatment become available. CONCLUSIONS: A multidisciplinary group of experts and patient advocates defined consensus-based guidance on the care of patients who may have APOL1-associated nephropathy.

4.
Kidney Int ; 100(1): 19-21, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34154709

RESUMO

Apolipoprotein L1 (APOL1) high-risk genotypes strongly associate with HIV-associated nephropathy, and antiretroviral therapy reduces the incidence of HIV-associated nephropathy and progression to end-stage kidney disease. Wudil et al. report cross-sectional APOL1 associations with proteinuria and estimated glomerular filtration rate in a northern Nigerian sample with HIV infection on antiretroviral therapy. Multiple ethnic groups with different APOL1 risk variant frequencies were included. Overall, APOL1 was associated with proteinuric chronic kidney disease; however, relationships with underlying causes of nephropathy and progression rates require further study.


Assuntos
Apolipoproteína L1 , Infecções por HIV , Adulto , Apolipoproteína L1/genética , Apolipoproteínas/genética , Estudos Transversais , Genótipo , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Infecções por HIV/epidemiologia , Humanos , Rim , Lipoproteínas HDL/genética , Nigéria , Fenótipo
5.
J Am Soc Nephrol ; 31(9): 2083-2096, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32675303

RESUMO

BACKGROUND: Two coding renal risk variants (RRVs) of the APOL1 gene (G1 and G2) are associated with large increases in CKD rates among populations of recent African descent, but the underlying molecular mechanisms are unknown. Mammalian cell culture models are widely used to study cytotoxicity of RRVs, but results have been contradictory. It remains unclear whether cytotoxicity is RRV-dependent or driven solely by variant-independent overexpression. It is also unknown whether expression of the reference APOL1 allele, the wild-type G0, could prevent cytotoxicity of RRVs. METHODS: We generated tetracycline-inducible APOL1 expression in human embryonic kidney HEK293 cells and examined the effects of increased expression of APOL1 (G0, G1, G2, G0G0, G0G1, or G0G2) on known cytotoxicity phenotypes, including reduced viability, increased swelling, potassium loss, aberrant protein phosphorylation, and dysregulated energy metabolism. Furthermore, whole-genome transcriptome analysis examined deregulated canonical pathways. RESULTS: At moderate expression, RRVs but not G0 caused cytotoxicity in a dose-dependent manner that coexpression of G0 did not reduce. RRVs also have dominant effects on canonical pathways relevant for the cellular stress response. CONCLUSIONS: In HEK293 cells, RRVs exhibit a dominant toxic gain-of-function phenotype that worsens with increasing expression. These observations suggest that high steady-state levels of RRVs may underlie cellular injury in APOL1 nephropathy, and that interventions that reduce RRV expression in kidney compartments may mitigate APOL1 nephropathy.


Assuntos
Apolipoproteína L1/genética , Apolipoproteína L1/fisiologia , Sobrevivência Celular , Metabolismo Energético , Perfilação da Expressão Gênica , Variação Genética , Células HEK293 , Humanos , Potássio/metabolismo , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
6.
Proc Natl Acad Sci U S A ; 113(4): 830-7, 2016 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-26699492

RESUMO

Two specific genetic variants of the apolipoprotein L1 (APOL1) gene are responsible for the high rate of kidney disease in people of recent African ancestry. Expression in cultured cells of these APOL1 risk variants, commonly referred to as G1 and G2, results in significant cytotoxicity. The underlying mechanism of this cytotoxicity is poorly understood. We hypothesized that this cytotoxicity is mediated by APOL1 risk variant-induced dysregulation of intracellular signaling relevant for cell survival. To test this hypothesis, we conditionally expressed WT human APOL1 (G0), the APOL1 G1 variant, or the APOL1 G2 variant in human embryonic kidney cells (T-REx-293) using a tetracycline-mediated (Tet-On) system. We found that expression of either G1 or G2 APOL1 variants increased apparent cell swelling and cell death compared with G0-expressing cells. These manifestations of cytotoxicity were preceded by G1 or G2 APOL1-induced net efflux of intracellular potassium as measured by X-ray fluorescence, resulting in the activation of stress-activated protein kinases (SAPKs), p38 MAPK, and JNK. Prevention of net K(+) efflux inhibited activation of these SAPKs by APOL1 G1 or G2. Furthermore, inhibition of SAPK signaling and inhibition of net K(+) efflux abrogated cytotoxicity associated with expression of APOL1 risk variants. These findings in cell culture raise the possibility that nephrotoxicity of APOL1 risk variants may be mediated by APOL1 risk variant-induced net loss of intracellular K(+) and subsequent induction of stress-activated protein kinase pathways.


Assuntos
Apolipoproteínas/genética , Transporte de Íons/genética , Nefropatias/genética , Lipoproteínas HDL/genética , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Mutação de Sentido Incorreto , Potássio/metabolismo , Substituição de Aminoácidos , Apolipoproteína L1 , Apolipoproteínas/fisiologia , População Negra/genética , Morte Celular , Tamanho Celular , Receptor gp130 de Citocina/biossíntese , Receptor gp130 de Citocina/genética , Progressão da Doença , Ativação Enzimática , Frequência do Gene , Predisposição Genética para Doença , Células HEK293 , Humanos , Nefropatias/etnologia , Lipoproteínas HDL/fisiologia , Sistema de Sinalização das MAP Quinases , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/metabolismo , Risco , Fator de Transcrição STAT3/metabolismo , Transfecção
10.
Clin Nephrol ; 86 (2016)(13): 114-118, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27509583

RESUMO

BACKGROUND: Risk variant Apolipoprotein L1 (G1/G2) are strongly associated with a spectrum of kidney disease in people of recent African descent. The mechanism of ApoL1 nephropathy is unknown. Podocytes and/or endothelial cells are the presumed target kidney cells. Given the close homology in structure and function of zebrafish (ZF) pronephros and human nephron, we studied the effect of podocyte-specific or endothelium-specific expression of ApoL1 (G0, G1, or G2) on the structure and function of ZF pronephros. METHODS: Wild type (G0) or risk variant ApoL1 (G1/G2) were expressed in podocyte-specific or endothelium-specific under podocin/Flk promoters, respectively, using Gal4-UAS system. Structural pronephric changes were studied with light and electron microscopy (EM). Proteinuria was assayed by measuring renal excretion of GFP-vitamin D binding protein. Puromycin aminonucleoside (PAN) was used as inducer of podocyte injury. RESULTS: Endothelial-specific transgenic expression of G1/G2 is associated with endothelial injury indicated by endothelial cell swelling, segmental early double contours, and loss of endothelium fenestrae. Podocyte specific expression of G1 is associated with segmental podocyte foot process effacement and irregularities relative to G0. Despite the histological changes, the expression of G1/G2 alone in podocyte or endothelium compartment is not associated with edema, proteinuria, or gross whole fish phenotype. Moreover, PAN produced equal pericardial edema in all transgenic fish as well as nontransgenic controls. CONCLUSIONS: Transgenic expression human ApoL1 (G1/G2) is associated with histologic abnormalities in ZF glomeruli but is insufficient to cause quantifiable renal dysfunction. This finding supports the necessity of a "second hit" in the pathogenesis/progression of ApoL1-associated nephropathy.


Assuntos
Apolipoproteínas/genética , Nefropatias/genética , Lipoproteínas HDL/genética , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Proteínas de Ligação a DNA/genética , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Variação Genética/genética , Genótipo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Glomérulos Renais/metabolismo , Proteínas de Membrana/genética , Podócitos/metabolismo , Podócitos/patologia , Regiões Promotoras Genéticas/genética , Pronefro/metabolismo , Pronefro/patologia , Proteinúria/urina , Fatores de Transcrição/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Proteína de Ligação a Vitamina D/urina , Peixe-Zebra
11.
J Biol Chem ; 288(5): 3477-88, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23258544

RESUMO

Insulin resistance, hyperlipidemia, and cardiovascular complications are common dysregulations of metabolic syndrome. Transplant patients treated with immunosuppressant drugs such as cyclosporine A (CsA), an inhibitor of calcineurin phosphatase, frequently develop similar metabolic complications. Although calcineurin is known to mediate insulin sensitivity by regulating ß-cell growth and adipokine gene transcription, its role in lipid homeostasis is poorly understood. Here, we examined lipid homeostasis in mice lacking calcineurin Aß (CnAß(-/-)). We show that mice lacking calcineurin Aß are hyperlipidemic and develop age-dependent insulin resistance. Hyperlipidemia found in CnAß(-/-) mice is, in part, due to increased lipolysis in adipose tissues, a process mediated by ß-adrenergic G-protein-coupled receptor signaling pathways. CnAß(-/-) mice also exhibit additional pathophysiological phenotypes caused by the potentiated GPCR signaling pathways. A cell autonomous mechanism with sustained cAMP/PKA activation is found in CnAß(-/-) mice or upon CsA treatment to inhibit calcineurin. Increased PKA activation and cAMP accumulation in CnAß(-/-) mice, however, are sensitive to phosphodiesterase inhibitor. Indeed, we show that calcineurin regulates degradation of phosphodiesterase 3B, in addition to phosphodiesterase 4D. These results establish a role for calcineurin in lipid homeostasis. These data also indicate that potentiated cAMP signaling pathway may provide an alternative molecular pathogenesis for the metabolic complications elicited by CsA in transplant patients.


Assuntos
Calcineurina/deficiência , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Hiperlipidemias/enzimologia , Transdução de Sinais , Envelhecimento/efeitos dos fármacos , Envelhecimento/patologia , Sequência de Aminoácidos , Animais , Células COS , Calcineurina/metabolismo , Chlorocebus aethiops , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/química , Ciclosporina/farmacologia , Embrião de Mamíferos/citologia , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Hiperlipidemias/patologia , Resistência à Insulina , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Inibidores de Fosfodiesterase/farmacologia , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais/efeitos dos fármacos , Triglicerídeos/biossíntese
12.
J Clin Invest ; 134(5)2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38227370

RESUMO

Two coding variants of apolipoprotein L1 (APOL1), called G1 and G2, explain much of the excess risk of kidney disease in African Americans. While various cytotoxic phenotypes have been reported in experimental models, the proximal mechanism by which G1 and G2 cause kidney disease is poorly understood. Here, we leveraged 3 experimental models and a recently reported small molecule blocker of APOL1 protein, VX-147, to identify the upstream mechanism of G1-induced cytotoxicity. In HEK293 cells, we demonstrated that G1-mediated Na+ import/K+ efflux triggered activation of GPCR/IP3-mediated calcium release from the ER, impaired mitochondrial ATP production, and impaired translation, which were all reversed by VX-147. In human urine-derived podocyte-like epithelial cells (HUPECs), we demonstrated that G1 caused cytotoxicity that was again reversible by VX-147. Finally, in podocytes isolated from APOL1 G1 transgenic mice, we showed that IFN-γ-mediated induction of G1 caused K+ efflux, activation of GPCR/IP3 signaling, and inhibition of translation, podocyte injury, and proteinuria, all reversed by VX-147. Together, these results establish APOL1-mediated Na+/K+ transport as the proximal driver of APOL1-mediated kidney disease.


Assuntos
Apolipoproteína L1 , Nefropatias , Compostos Organotiofosforados , Camundongos , Animais , Humanos , Apolipoproteína L1/genética , Células HEK293 , Variação Genética , Nefropatias/genética , Camundongos Transgênicos
13.
JCI Insight ; 7(11)2022 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-35472001

RESUMO

COVID-19 infection causes collapse of glomerular capillaries and loss of podocytes, culminating in a severe kidney disease called COVID-19-associated nephropathy (COVAN). The underlying mechanism of COVAN is unknown. We hypothesized that cytokines induced by COVID-19 trigger expression of pathogenic APOL1 via JAK/STAT signaling, resulting in podocyte loss and COVAN phenotype. Here, based on 9 biopsy-proven COVAN cases, we demonstrated for the first time, to the best of our knowledge, that APOL1 protein was abundantly expressed in podocytes and glomerular endothelial cells (GECs) of COVAN kidneys but not in controls. Moreover, a majority of patients with COVAN carried 2 APOL1 risk alleles. We show that recombinant cytokines induced by SARS-CoV-2 acted synergistically to drive APOL1 expression through the JAK/STAT pathway in primary human podocytes, GECs, and kidney micro-organoids derived from a carrier of 2 APOL1 risk alleles, but expression was blocked by a JAK1/2 inhibitor, baricitinib. We demonstrate that cytokine-induced JAK/STAT/APOL1 signaling reduced the viability of kidney organoid podocytes but was rescued by baricitinib. Together, our results support the conclusion that COVID-19-induced cytokines are sufficient to drive COVAN-associated podocytopathy via JAK/STAT/APOL1 signaling and that JAK inhibitors could block this pathogenic process. These findings suggest JAK inhibitors may have therapeutic benefits for managing cytokine-induced, APOL1-mediated podocytopathy.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19 , Citocinas , Inibidores de Janus Quinases , Nefropatias , Apolipoproteína L1/genética , Azetidinas/farmacologia , COVID-19/metabolismo , Citocinas/metabolismo , Células Endoteliais/metabolismo , Humanos , Inibidores de Janus Quinases/farmacologia , Janus Quinases/metabolismo , Nefropatias/tratamento farmacológico , Nefropatias/metabolismo , Nefropatias/virologia , Organoides/metabolismo , Purinas/farmacologia , Pirazóis/farmacologia , SARS-CoV-2/isolamento & purificação , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
14.
Mol Cell Biol ; 26(20): 7372-87, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16908540

RESUMO

Compromised immunoregulation contributes to obesity and complications in metabolic pathogenesis. Here, we demonstrate that the nuclear factor of activated T cell (NFAT) group of transcription factors contributes to glucose and insulin homeostasis. Expression of two members of the NFAT family (NFATc2 and NFATc4) is induced upon adipogenesis and in obese mice. Mice with the Nfatc2-/- Nfatc4-/- compound disruption exhibit defects in fat accumulation and are lean. Nfatc2-/- Nfatc4-/- mice are also protected from diet-induced obesity. Ablation of NFATc2 and NFATc4 increases insulin sensitivity, in part, by sustained activation of the insulin signaling pathway. Nfatc2-/- Nfatc4-/- mice also exhibit an altered adipokine profile, with reduced resistin and leptin levels. Mechanistically, NFAT is recruited to the transcription loci and regulates resistin gene expression upon insulin stimulation. Together, these results establish a role for NFAT in glucose/insulin homeostasis and expand the repertoire of NFAT function to metabolic pathogenesis and adipokine gene transcription.


Assuntos
Glucose/metabolismo , Homeostase , Insulina/metabolismo , Fatores de Transcrição NFATC/metabolismo , Proteínas Quinases Ativadas por AMP , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipócitos/patologia , Animais , Diferenciação Celular , Linhagem Celular , Chlorocebus aethiops , Gorduras na Dieta/farmacologia , Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Complexos Multienzimáticos/metabolismo , Fatores de Transcrição NFATC/deficiência , Fatores de Transcrição NFATC/genética , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Resistina/metabolismo , Sensibilidade e Especificidade , Transdução de Sinais
16.
Artigo em Inglês | MEDLINE | ID: mdl-30513574

RESUMO

Chronic kidney disease (CKD) is a major non-communicable disease associated with high rates of premature morbidity and mortality. The prevalence of hypovitaminosis D (deficiency of 25(OH)D or 25D) is greater in racial/ethnic minorities and in patients with CKD than the general population. Low 25D is associated with bone and mineral disorders as well as immune, cardiometabolic and cardiovascular (CV) diseases. Thus, it has been suggested that low 25D contributes to the poor outcomes in patients with CKD. The prevalence of hypovitaminosis D rises progressively with advancing severity of kidney disease with over 30% of patients with CKD stage 3 and 70% patients with CKD stage 5 estimated to have low levels of 25D. This report describes several of the abnormal physiologic and counter-regulatory actions related to low 25D in CKD such as those in oxidative stress and inflammatory systems, and some of the preclinical and clinical evidence, or lack thereof, of normalizing serum 25D levels to improve outcomes in patients with CKD, and especially for the high risk subset of racial/ethnic minorities who suffer from higher rates of advanced CKD and hypovitaminosis D.


Assuntos
Estresse Oxidativo/fisiologia , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/fisiopatologia , Deficiência de Vitamina D/complicações , Deficiência de Vitamina D/fisiopatologia , Vitamina D/sangue , Etnicidade , Inquéritos Epidemiológicos , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/epidemiologia , Estados Unidos/epidemiologia , Deficiência de Vitamina D/sangue , Deficiência de Vitamina D/epidemiologia
17.
Semin Nephrol ; 37(6): 546-551, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29110762

RESUMO

Apolipoprotein L1 (APOL1) protein is the human serum factor that protect human beings against Trypanosoma brucei brucei, the cause of trypanosomiasis. Subspecies of T b brucei that cause human sleeping sickness-T b gambiense and T b rhodesiense evolved molecular mechanisms that enabled them to evade killing by APOL1. Sequence changes (termed G1 and G2) in the APOL1 gene that restored its ability to kill T b rhodesiense also increase the risk of developing glomerular diseases and accelerate progression to end-stage kidney disease. To lyse trypanosome parasites, APOL1 forms pores in the trypanosome endolysosomal and mitochondrial membranes, resulting in rapid membrane depolarization. However, the molecular mechanism underlying APOL1 nephropathy is unknown. Recent experimental evidence has shown that aberrant efflux of intracellular potassium is an early event in APOL1-induced death of human embryonic kidney cells. Here, we discuss the possibility that abnormal efflux of cellular potassium or other cations may be relevant to the pathogenesis of APOL1 nephropathy.


Assuntos
Apolipoproteína L1/genética , Morte Celular/genética , Transporte de Íons/genética , Nefropatias/genética , Animais , Células HEK293 , Humanos , Canais Iônicos , Mutação , Podócitos , Potássio/metabolismo
19.
Artigo em Inglês | MEDLINE | ID: mdl-15093963

RESUMO

It is well known that cocaine's psychomotor stimulant properties derive from enhanced monoamines via synaptic transporter/reuptake inhibition and release mechanisms. However, to further understand mechanisms of action for cocaine, which may be receptor-related, ketanserin, a selective 5-HT(2A/2C) antagonist was used to ascertain a possible mediation for 5-HT(2A//2C) receptors in the monoamine and behavioral responses to cocaine. The studies were performed in the freely moving and behaving animal with In Vivo Microvoltammetry. Miniature carbon sensors, BRODERICK PROBE microelectrodes detected dopamine (DA) and serotonin (5-HT) concentrations in Nucleus Accumbens (NAcc) of male, Sprague-Dawley laboratory rats in separate signals and within seconds while at the same time, locomotor behavior was monitored with infrared photobeams. Synaptic release of each monoamine was detected because separate studies showed that the depolarization blocker, gamma-butyrolactone (gamma BL), decreased steady-state values [Pharmacol. Biochem. Behav. 40 (1991) 969]. Acute studies (Day 1) were performed; the animals received single injection of drug(s) in the faradaic behavioral chamber after a stable baseline during habituation behavior was achieved. After completion of the study, the animals were returned to their home cages. Subacute studies (Day 2) were also performed; these took place 24 h later in the faradaic behavioral chamber; same animal control was used and no further drug was administered. Day 2 data were compared to baseline (habituation data) on Day 1. Results showed that (1) Acute administration of Cocaine (10 mg/kg, i.p.) (N=5) increased DA and 5-HT release above baseline (p<0.001) while locomotion was also increased above baseline (p<0.001). (2) In Subacute studies in the cocaine group, when no further drug was administered, DA release decreased (p<0.001) and decreases in 5-HT release also occurred throughout the time course (p<0.05). Locomotor behavior increased above baseline and showed a trend toward statistical significance (p<0.07). (3) Acute administration of Ketanserin/Cocaine (3 mg/kg s.c. and 10 mg/kg i.p., respectively) (N=6) showed that ketanserin antagonized DA and 5-HT release (p<0.001), while locomotion was antagonized as well (p<0.001). (4) In Subacute studies, in the ketanserin/cocaine group, when no further drug was administered, DA decreased (p<0.001), but 5-HT increased (p<0.001), while locomotor activity increased above baseline and a trend toward statistical significance was seen (p<0.07). Additional saline controls were without effect (p>0.05). In summary, Acute studies showed that cocaine produced its psychostimulant responses on monoamines and behavior and ketanserin antagonized these responses, likely via a 5-HT(2A/2C) receptor mediation. Presynaptic and postsynaptic responses were not distinguished, suggesting, in addition, a role for 5-HT-ergic modulation of DA, likely DA(2) postsynaptic modulation Subacute cocaine studies showed that on Day 2, deficiencies in monoamines occurred, reflecting cocaine withdrawal mechanisms neurochemically, while locomotor behavior did not show such dramatic deficiencies. Indeed, behavior increased above baseline. Moreover, ketanserin reversed 5-HT-related and not DA-related cocaine monoamine responses, while locomotion continued to be insignificantly increased above baseline as was seen in the Subacute cocaine group. The data suggest that presynaptic 5-HT(2A/2C) receptor mechanisms may be important during withdrawal from single injection of cocaine.


Assuntos
Monoaminas Biogênicas/metabolismo , Cocaína/farmacologia , Ketanserina/farmacologia , Atividade Motora/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Receptor 5-HT2A de Serotonina/efeitos dos fármacos , Receptor 5-HT2C de Serotonina/efeitos dos fármacos , Antagonistas da Serotonina/farmacologia , Animais , Calibragem , Dopamina/metabolismo , Eletrofisiologia , Masculino , Microeletrodos , Núcleo Accumbens/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serotonina/metabolismo
20.
Methods Mol Biol ; 780: 47-55, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21870253

RESUMO

The enzymatic function of poly(adenosine diphosphate (ADP)-ribose) polymerase (PARP) is central to many of its function as a component of DNA repair machinery, modulator of gene transcription, and cell differentiation. While the auto-modification domain of PARP has been shown to be a primary acceptor site of poly-ADP ribose (pADPr), other DNA binding nuclear proteins are also modified by pADPr. It is -generally accepted that pADPr polymer is built upon the carboxyl side chain of specific Glu, Asp, and/or Lys residues within the target protein. Identification of the unique amino acid acceptors of pADPr in these nuclear proteins is an active area of study. Because of the heterogeneity of pADPr chain on modified -protein targets, the resulting modified proteins have unpredictable final masses, making it difficult to -identify acceptor amino acids. Using recombinant proteins, in vitro pADP ribosylation assay and mass spectrometry, we have been able to identify conserved Glu residue in transcription factor NFAT that is enzymatically modified in vitro with pADPr by PARP-1. We discuss this protocol here as a model approach for identifying pADPr acceptors in other nuclear proteins.


Assuntos
Poli Adenosina Difosfato Ribose/metabolismo , Espectrometria de Massas , Fatores de Transcrição NFATC/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Processamento de Proteína Pós-Traducional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA