Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Glycoconj J ; 37(6): 729-744, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32915357

RESUMEN

Apolipoprotein L1 (APOL1) wild type (G0) plays a role in the metabolism of sphingolipids, glycosphingolipids, sphingomyelin and ceramide, which constitute bioactive components of the lipid rafts (DRM). We asked whether APOL1 variants (APOL1-Vs) G1 and G2 carry the potential to alter the metabolism of sphingolipids in human podocytes. The sphingolipid pattern in HPs overexpressing either APOL1G0 or APOL1-Vs was analysed by using a thin mono- and bi-dimensional layer chromatography, mass-spectrometry and metabolic labelling with [1-3H]sphingosine. HP G0 and G1/G2-Vs exhibit a comparable decrease in lactosylceramide and an increase in the globotriaosylceramide content. An analysis of the main glycohydrolases activity involved in glycosphingolipid catabolism showed an overall decrease in the activeness of the tested enzymes, irrespective of the type of APOL1-Vs expression. Similarly, the high throughput cell live-based assay showed a comparable increased action of the plasma membrane glycosphingolipid-glycohydrolases in living cells independent of the genetic APOL1 expression profile. Importantly, the most significative modification of the sphingolipid pattern induced by APOL1-Vs occurred in DRM resulted with a drastic reduction of radioactivity associated with sphingolipids. G1/G2-Vs present a decrease amount of globotriaosylceramide and globopentaosylceramide compared to G0. Additionally, ceramide at the DRM site and lactosylceramide in general, showed a greatest fall in G1/G2 in comparison with G0. Additionally, the levels of glucosylceramide decreased only in the DRM of human podocytes overexpressing G1/G2-Vs. These findings suggest that altered sphingolipidsprofiles may contribute to the deranged functionality of the plasma membrane in APOL1 risk milieu.


Asunto(s)
Apolipoproteína L1/genética , Glicósido Hidrolasas/genética , Podocitos/metabolismo , Esfingolípidos/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Metabolismo , Polimorfismo Genético/genética , Esfingolípidos/metabolismo
2.
Am J Physiol Cell Physiol ; 317(2): C209-C225, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31116585

RESUMEN

We hypothesized that a functional apolipoprotein LI (APOL1)-miR193a axis (inverse relationship) preserves, but disruption alters, the podocyte molecular phenotype through the modulation of autophagy flux. Podocyte-expressing APOL1G0 (G0-podocytes) showed downregulation but podocyte-expressing APOL1G1 (G1-podocytes) and APOL1G2 (G2-podocytes) displayed enhanced miR193a expression. G0-, G1-, and G2-podocytes showed enhanced expression of light chain (LC) 3-II and beclin-1, but a disparate expression of p62 (low in wild-type but high in risk alleles). G0-podocytes showed enhanced, whereas G1- and G2-podocytes displayed decreased, phosphorylation of Unc-51-like autophagy-activating kinase (ULK)1 and class III phosphatidylinositol 3-kinase (PI3KC3). Podocytes overexpressing miR193a (miR193a-podocytes), G1, and G2 showed decreased transcription of PIK3R3 (PI3KC3's regulatory unit). Since 3-methyladenine (3-MA) enhanced miR193a expression but inhibited PIK3R3 transcription, it appears that 3-MA inhibits autophagy and induces podocyte dedifferentiation via miR193a generation. miR193a-, G1-, and G2-podocytes also showed decreased phosphorylation of mammalian target of rapamycin (mTOR) that could repress lysosome reformation. G1- and G2-podocytes showed enhanced expression of run domain beclin-1-interacting and cysteine-rich domain-containing protein (Rubicon); however, its silencing prevented their dedifferentiation. Docking, protein-protein interaction, and immunoprecipitation studies with antiautophagy-related gene (ATG)14L, anti-UV radiation resistance-associated gene (UVRAG), or Rubicon antibodies suggested the formation of ATG14L complex I and UVRAG complex II in G0-podocytes and the formation of Rubicon complex III in G1- and G2-podocytes. These findings suggest that the APOL1 risk alleles favor podocyte dedifferentiation through blockade of multiple autophagy pathways.


Asunto(s)
Apolipoproteína L1/metabolismo , Autofagia , Desdiferenciación Celular , MicroARNs/metabolismo , Podocitos/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Apolipoproteína L1/genética , Autofagosomas/metabolismo , Autofagosomas/patología , Proteínas Relacionadas con la Autofagia/metabolismo , Línea Celular Transformada , Regulación de la Expresión Génica , Humanos , MicroARNs/genética , Simulación de Dinámica Molecular , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Podocitos/patología , Mapas de Interacción de Proteínas , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo
3.
Am J Physiol Renal Physiol ; 317(2): F463-F477, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31241995

RESUMEN

The apolipoprotein L1 (APOL1) gene is unique to humans and gorillas and appeared ~33 million years ago. Since the majority of the mammals do not carry APOL1, it seems to be dispensable for kidney function. APOL1 renal risk variants (RRVs; G1 and G2) are associated with the development as well as progression of chronic kidney diseases (CKDs) at higher rates in populations with African ancestry. Cellular expression of two APOL1 RRVs has been demonstrated to induce cytotoxicity, including necrosis, apoptosis, and pyroptosis, in several cell types including podocytes; mechanistically, these toxicities were attributed to lysosomal swelling, K+ depletion, mitochondrial dysfunction, autophagy blockade, protein kinase receptor activation, ubiquitin D degradation, and endoplasmic reticulum stress; notably, these effects were found to be dose dependent and occurred only in overtly APOL1 RRV-expressing cells. However, cellular protein expressions as well as circulating blood levels of APOL1 RRVs were not elevated in patients suffering from APOL1 RRV-associated CKDs. Therefore, the question arises as to whether it is gain or loss of function on the part of APOL1 RRVs contributing to kidney cell injury. The question seems to be more pertinent after the recognition of the role of APOL1 nonrisk (G0) in the transition of parietal epithelial cells and preservation of the podocyte molecular phenotype through modulation of the APOL1-miR-193a axis. With this background, the present review analyzed the available literature in terms of the known function of APOL1 nonrisk and how the loss of these functions could have contributed to two APOL1 RRV-associated CKDs.


Asunto(s)
Apolipoproteína L1/genética , Apolipoproteína L1/fisiología , Riñón/fisiología , Animales , Humanos , Riñón/citología , Riñón/metabolismo , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/fisiopatología
4.
Am J Pathol ; 188(11): 2508-2528, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30201495

RESUMEN

Human parietal epithelial cells (PECs) are progenitor cells that sustain podocyte homeostasis. We hypothesized that the lack of apolipoprotein (APO) L1 ensures the PEC phenotype, but its induction initiates PEC transition (expression of podocyte markers). APOL1 expression and down-regulation of miR193a coincided with the expression of podocyte markers during the transition. The induction of APOL1 also stimulated transition markers in human embryonic kidney cells (cells with undetectable APOL1 protein expression). APOL1 silencing in PECs up-regulated miR193a expression, suggesting the possibility of a reciprocal feedback relationship between APOL1 and miR193a. HIV, interferon-γ, and vitamin D receptor agonist down-regulated miR193a expression and induced APOL1 expression along with transition markers in PECs. Luciferase assay suggested a putative interaction between miR193a and APOL1. Since silencing of APOL1 attenuated HIV-, vitamin D receptor agonist-, miR193a inhibitor-, and interferon-γ-induced expression of transition markers, APOL1 appears to be a critical functional constituent of the miR193a- APOL1 axis in PECs. This notion was confirmed by further enhanced expression of PEC markers in APOL1 mRNA-silenced PECs. In vivo studies, glomeruli in patients with HIV, and HIV/APOL1 transgenic mice had foci of PECs expressing synaptopodin, a transition marker. APOL1 likely regulates PEC molecular phenotype through modulation of miR193a expression, and APOL1 and miR193a share a reciprocal feedback relationship.


Asunto(s)
Nefropatía Asociada a SIDA/patología , Apolipoproteína L1/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica , Glomérulos Renales/patología , MicroARNs/genética , Nefropatía Asociada a SIDA/metabolismo , Nefropatía Asociada a SIDA/virología , Animales , Apolipoproteína L1/genética , Estudios de Casos y Controles , Células Epiteliales/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Glomérulos Renales/metabolismo , Ratones , Ratones Transgénicos
5.
Am J Physiol Renal Physiol ; 314(5): F832-F843, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29357419

RESUMEN

The loss of podocyte (PD) molecular phenotype is an important feature of diabetic podocytopathy. We hypothesized that high glucose (HG) induces dedifferentiation in differentiated podocytes (DPDs) through alterations in the apolipoprotein (APO) L1-microRNA (miR) 193a axis. HG-induced DPD dedifferentiation manifested in the form of downregulation of Wilms' tumor 1 (WT1) and upregulation of paired box 2 (PAX2) expression. WT1-silenced DPDs displayed enhanced expression of PAX2. Immunoprecipitation of DPD cellular lysates with anti-WT1 antibody revealed formation of WT1 repressor complexes containing Polycomb group proteins, enhancer of zeste homolog 2, menin, and DNA methyltransferase (DNMT1), whereas silencing of either WT1 or DNMT1 disrupted this complex with enhanced expression of PAX2. HG-induced DPD dedifferentiation was associated with a higher expression of miR193a, whereas inhibition of miR193a prevented DPD dedifferentiation in HG milieu. HG downregulated DPD expression of APOL1. miR193a-overexpressing DPDs displayed downregulation of APOL1 and enhanced expression of dedifferentiating markers; conversely, silencing of miR193a enhanced the expression of APOL1 and preserved DPD phenotype. Moreover, stably APOL1G0-overexpressing DPDs displayed the enhanced expression of WT1 but attenuated expression of miR193a; nonetheless, silencing of APOL1 reversed these effects. Since silencing of APOL1 enhanced miR193a expression as well as dedifferentiation in DPDs, it appears that downregulation of APOL1 contributed to dedifferentiation of DPDs through enhanced miR193a expression in HG milieu. Vitamin D receptor agonist downregulated miR193a, upregulated APOL1 expression, and prevented dedifferentiation of DPDs in HG milieu. These findings suggest that modulation of the APOL1-miR193a axis carries a potential to preserve DPD molecular phenotype in HG milieu.


Asunto(s)
Apolipoproteína L1/metabolismo , Desdiferenciación Celular/efectos de los fármacos , Glucosa/toxicidad , MicroARNs/metabolismo , Podocitos/efectos de los fármacos , Apolipoproteína L1/genética , Calcitriol/análogos & derivados , Calcitriol/farmacología , Línea Celular Transformada , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , MicroARNs/genética , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX2/metabolismo , Fenotipo , Podocitos/metabolismo , Podocitos/patología , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas WT1/genética , Proteínas WT1/metabolismo
6.
Exp Cell Res ; 352(2): 193-201, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28159470

RESUMEN

HIV-associated nephropathy (HIVAN) is characterized by heavy proteinuria, rapidly progressive renal failure, and distinct morphological features in the kidney. HIV-induced epithelial-mesenchymal transition (EMT) is critically important for the progression of kidney injury. In this study, we tested the role of hedgehog pathway in the HIV-induced EMT and fibrosis of kidney. We used the Tg26 mice, the abundantly used HIVAN mouse model, to investigate the activation of hedgehog pathway by HIV. Western blotting and real time PCR results showed that renal tissue expression of hedgehog pathway related molecules, including hedgehog homologous (Shh, Ihh, Dhh), PTCH, and Gli1, were increased in HIVAN (Tg26) mice; while immunofluorescent staining displayed localization PTCH expression in podocytes. For in vitro studies, we used recombinant sonic hedgehog (Shh) and HIV for their expression by podocytes. Both the methods activated the hedgehog pathway, enhanced the expression of EMT markers, and decreased impermeability. Overexpression of Gli1 by human podocytes also augmented their expression of EMT markers. On the other hand, the blockade of hedgehog pathway with Gant 58, a specific blocker for Gli1-induced transcription, dramatically decreased HIV-induced podocyte EMT and permeability. These results indicate that hedgehog pathway plays an important role in HIV-induced podocyte injury. The present study provides mechanistical insight into a new target for therapeutic strategy.


Asunto(s)
Transición Epitelial-Mesenquimal , Proteínas Hedgehog/genética , Podocitos/metabolismo , Animales , Línea Celular , Femenino , VIH , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Ratones , Podocitos/citología , Podocitos/virología , Piridinas/farmacología , Tiofenos/farmacología
7.
Am J Pathol ; 186(2): 347-58, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26683666

RESUMEN

Dysregulated growth and loss of podocytes are important features of HIV-associated nephropathy. Recently, HIV was reported to induce a new type of programed cell death, pyroptosis, in T lymphocytes through induction of Nod-like receptor protein 3 (NLRP3) inflammasome complexes. We evaluated the role of HIV in podocyte NLRP3 inflammasome formation both in vivo and in vitro. Renal cortical sections of HIV-transgenic mice (Tg26) displayed increased expression of NLRP3, ASC (a CARD protein), caspase-1, and IL-1ß proteins, confirming NLRP3 inflammasome complex formation in podocytes of Tg26 mice. Renal tissues of Tg26 mice also displayed enhanced mRNA levels and protein expressions of inflammasome markers (NLRP3, ASC, and caspase-1, and IL-1ß). Serum of Tg26 mice also showed elevated concentrations of IL-1ß cytokine compared with FVBN mice. HIV induced pyroptosis in a dose- and time-dependent manner within podocytes, a phenotype of inflammasome activation. Caspase-1 inhibitor not only attenuated podocyte expression of caspase-1 and IL-1ß but also provided protection against pyroptosis, suggesting that HIV-induced podocyte injury was mediated by caspase-1 activation. Interestingly, HIV-induced podocyte pyroptosis could be partially inhibited by Tempol (a superoxide dismutase-mimetic agent) and by glyburide (an inhibitor of potassium efflux). These findings suggest that generation of reactive oxygen species and potassium efflux contribute to HIV-induced pyroptosis and NLRP3 inflammasome activation in podocytes.


Asunto(s)
Nefropatía Asociada a SIDA/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Portadoras/metabolismo , Inflamasomas/metabolismo , Podocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/fisiología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Podocitos/virología
8.
Exp Mol Pathol ; 102(1): 97-105, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28069388

RESUMEN

Vitamin D receptor (VDR) deficient status has been shown to be associated with the activation of renin angiotensin system (RAS). We hypothesized that lack of VDR would enhance p53 expression in podocytes through down regulation of SIRT1; the former would enhance the transcription of angiotensinogen (Agt) and angiotensinogen II type 1 receptor (AT1R) leading to the activation of RAS. Renal tissues of VDR mutant (M) mice displayed increased expression of p53, Agt, renin, and AT1R. In vitro studies, VDR knockout podocytes not only displayed up regulation p53 but also displayed enhanced expression of Agt, renin and AT1R. VDR deficient podocytes also displayed an increase in mRNA expression for p53, Agt, renin, and AT1R. Interestingly, renal tissues of VDR-M as well as VDR heterozygous (h) mice displayed attenuated expression of deacetylase SIRT1. Renal tissues of VDR-M mice showed acetylation of p53 at lysine (K) 382 residues inferring that enhanced p53 expression in renal tissues could be the result of ongoing acetylation, a consequence of SIRT1 deficient state. Notably, podocytes lacking SIRT1 not only showed acetylation of p53 at lysine (K) 382 residues but also displayed enhanced p53 expression. Either silencing of SIRT1/VDR or treatment with high glucose enhanced podocyte PPAR-y expression, whereas, immunoprecipitation (IP) of their lysates with anti-retinoid X receptor (RXR) antibody revealed presence of PPAR-y. It appears that either the deficit of SIRT1 has de-repressed expression of PPAR-y or enhanced podocyte expression of PPAR-y (in the absence of VDR) has contributed to the down regulation of SIRT1.


Asunto(s)
Podocitos/metabolismo , Receptores de Calcitriol/genética , Sistema Renina-Angiotensina/genética , Sirtuina 1/genética , Acetilación , Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Animales , Western Blotting , Células Cultivadas , Humanos , Riñón/citología , Riñón/metabolismo , Lisina/genética , Lisina/metabolismo , Ratones Noqueados , Modelos Genéticos , Podocitos/citología , Interferencia de ARN , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Calcitriol/deficiencia , Renina/genética , Renina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
9.
Retrovirology ; 13(1): 63, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27599995

RESUMEN

BACKGROUND: Patients of African ancestry with untreated HIV-1 infection and carrying the G1 or G2 kidney disease risk variants (Vs) at the APOL1 gene have a tenfold higher risk of developing HIV-associated nephropathy (HIVAN) compared to those with the non-risk wild type (WT) G0 variant. However, the mechanistic contribution of the APOL1 allelic state to kidney injury in HIV-1 infection remains to be elucidated. RESULTS: Non-risk WT APOL1 is associated with lower intracellular levels of HIV-1 in conditionally immortalized human podocytes, while the over expression of G1 or G2 risk Vs significantly increases viral accumulation. The priming of podocytes with exogenous IL-1ß facilitates HIV-1 entry, via the up-regulation of DC-SIGN. The over expression of APOL1 G1 and G2 risk Vs in combination with an increase in IL-1ß levels causes a greater increase in viral concentration than either condition alone. In turn, HIV-1 and exogenous IL-1ß together induce a de novo secretion of endogenous IL-1ß and an increase of APOL1 gene expression. CONCLUSIONS: Our findings indicate that the presence of risk Vs of APOL1 is permissive of HIV-1 persistence in human podocytes in synergy with IL-1ß, a cytokine that characterizes the inflammatory milieu of acute and chronic phases of HIV-1 infection. The elucidation of these molecular mechanisms explains, at least in part, the higher frequency of HIVAN in populations carrying the risk polymorphic genetic variant of APOL1 gene.


Asunto(s)
Nefropatía Asociada a SIDA/genética , Apolipoproteínas/genética , Infecciones por VIH/genética , VIH-1/fisiología , Interleucina-1beta/inmunología , Lipoproteínas HDL/genética , Podocitos/virología , Polimorfismo Genético , Nefropatía Asociada a SIDA/inmunología , Nefropatía Asociada a SIDA/virología , África , Alelos , Apolipoproteína L1 , Moléculas de Adhesión Celular/genética , Femenino , Predisposición Genética a la Enfermedad/etnología , Infecciones por VIH/etnología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Interleucina-1beta/biosíntesis , Lectinas Tipo C/genética , Masculino , Persona de Mediana Edad , Receptores de Superficie Celular/genética , Internalización del Virus
10.
Am J Physiol Cell Physiol ; 309(1): C22-37, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25924622

RESUMEN

The apolipoprotein L1 (APOL1) gene (APOL1) product is toxic to kidney cells, and its G1 and G2 alleles are strongly associated with increased risk for kidney disease progression in African Americans. Variable penetrance of the G1 and G2 risk alleles highlights the significance of additional factors that trigger or modify the progression of disease. In this regard, the effect of alternative splicing in the absence or presence of G1 or G2 alleles is unknown. In this study we investigated whether alternative splicing of non-G1, non-G2 APOL1 (APOL1 G0) affects its biological activity. Among seven APOL1 exons, exons 2 and 4 are differentially expressed in major transcripts. We found that, in contrast to APOL1 splice variants B3 or C, variants A and B1 demonstrate strong toxicity in human embryonic kidney (HEK293T) cells. Subsequently, we established that exon 4 is a major determinant of toxicity of variants A and B1 and that extracellular release of these variants is dispensable for their cytotoxicity. Although only variants A and B1 induced nuclear translocation of transcription factor EB (TFEB), a master regulator of lysosomal biogenesis and autophagy, exon 4-positive and -negative APOL1 variants stimulated perinuclear accumulation of unprocessed autophagosomes. Knockdown of endogenous TFEB did not attenuate APOL1 cytotoxicity, indicating that nuclear translocation of TFEB is dispensable for APOL1 toxicity. Our findings that a human podocyte cell line expresses exon 4-positive and -negative APOL1 transcripts suggest that these variants may play a differential role in podocyte pathology. In summary, we have identified exon 4 as a major determinant of APOL1 G0 cytotoxicity.


Asunto(s)
Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Autofagia , Exones , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Podocitos/metabolismo , Transporte Activo de Núcleo Celular , Empalme Alternativo , Secuencia de Aminoácidos , Apolipoproteína L1 , Apolipoproteínas/química , Secuencia de Bases , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Lipoproteínas HDL/química , Datos de Secuencia Molecular , Podocitos/patología , Interferencia de ARN , ARN Mensajero/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Transcripción Genética , Transfección
11.
Am J Physiol Renal Physiol ; 309(3): F189-203, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26084932

RESUMEN

ANG II type 1 receptor blockade (AT1R-BLK) is used extensively to slow down the progression of proteinuric kidney diseases. We hypothesized that AT1R-BLK provides podocyte protection through regulation of silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) and vitamin D receptor (VDR) expression under adverse milieus such as high glucose and human immunodeficiency virus infection. Both AT1R-BLK and VDR agonists (VDAs) stimulated VDR complex formation that differed not only in their composition but also in their functionality. AT1R-BLK-induced VDR complexes contained predominantly unliganded VDR, SMRT, and phosphorylated histone deacetylase 3, whereas VDA-VDR complexes were constituted by liganded VDR and CREB-binding protein/p300. AT1R-BLK-induced complexes attenuated podocyte acetyl-histone 3 levels as well as cytochrome P-450 family 24A1 expression, thus indicating their deacetylating and repressive properties. On the other hand, VDA-VDR complexes not only increased podocyte acetyl-histone 3 levels but also enhanced cytochrome P-450 family 24A1 expression, thus suggesting their acetylating and gene activation properties. AT1R-BLK- induced podocyte SMRT inhibited expression of the proapoptotic gene BAX through downregulation of Wip1 and phosphorylation of checkpoint kinase 2 in high-glucose milieu. Since SMRT-depleted podocytes lacked AT1R-BLK-mediated protection against DNA damage, it appears that SMRT is necessary for DNA repairs during AT1R-BLK. We conclude that AT1R-BLK provides podocyte protection in adverse milieus predominantly through SMRT expression and partly through unliganded VDR expression in 1,25(OH)2D-deficient states; on the other hand, AT1R-BLK contributes to liganded VDR expression in 1,25(OH)2D-sufficient states.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Co-Represor 2 de Receptor Nuclear/fisiología , Acetilación , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Co-Represoras/efectos de los fármacos , Daño del ADN , Relación Dosis-Respuesta a Droga , Histonas/metabolismo , Humanos , Losartán/farmacología , Podocitos/efectos de los fármacos , Podocitos/enzimología , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Sustancias Protectoras/farmacología , Receptores de Calcitriol/efectos de los fármacos , Vitamina D3 24-Hidroxilasa/biosíntesis , Vitamina D3 24-Hidroxilasa/metabolismo
12.
Exp Mol Pathol ; 99(1): 109-15, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26079546

RESUMEN

Collapsing glomerulopathy and microcysts are characteristic histological features of HIV-associated nephropathy (HIVAN). We have previously reported the role of epithelial mesenchymal transition (EMT) in the development of glomerular and tubular cell phenotypes in HIVAN. Since persistent tubular cell activation of NFκB has been reported in HIVAN, we now hypothesize that HIV may be contributing to tubular cell phenotype via lysophosphatidic acid (LPA) mediated downstream signaling. Interestingly, LPA and its receptors have also been implicated in the tubular interstitial cell fibrosis (TIF) and cyst formation in autosomal dominant polycystic kidney disease (PKD). Primary human proximal tubular cells (HRPTCs) were transduced with either empty vector (EV/HRPTCs), HIV (HIV/HRPTCs) or treated with LPA (LPA/HRPTC). Immunoelectrophoresis of HIV/HRPTCs and LPA/HRPTCs displayed enhanced expression of pro-fibrotic markers: a) fibronectin (2.25 fold), b) connective tissue growth factor (CTGF; 4.8 fold), c) α-smooth muscle actin (α-SMA; 12 fold), and d) collagen I (5.7 fold). HIV enhanced tubular cell phosphorylation of ILK-1, FAK, PI3K, Akt, ERKs and P38 MAPK. HIV increased tubular cell transcriptional binding activity of NF-κB; whereas, a LPA biosynthesis inhibitor (AACOCF3), a DAG kinase inhibitor, a LPA receptor blocker (Ki16425), a NF-κB inhibitor (PDTC) and NFκB-siRNA not only displayed downregulation of a NFκB activity but also showed attenuated expression of profibrotic/EMT genes in HIV milieu. These findings suggest that LPA could be contributing to HIV-induced tubular cell phenotype via NFκB activation in HIVAN.


Asunto(s)
Nefropatía Asociada a SIDA/patología , Glomérulos Renales/citología , Túbulos Renales/citología , Lisofosfolípidos/metabolismo , Nefropatía Asociada a SIDA/genética , Actinas/genética , Actinas/metabolismo , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Fibronectinas/genética , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Silenciador del Gen , Vectores Genéticos , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Regulación hacia Arriba , Vimentina/genética , Vimentina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Exp Mol Pathol ; 99(1): 139-44, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26091559

RESUMEN

Increasing lines of evidence have demonstrated that the development of higher rates of non-diabetic glomerulosclerosis (GS) in African Americans can be attributed to two coding sequence variants (G1 and G2) in the Apolipoprotein L1 (APOL) gene. Recent studies indicate that the gene products of these APOL1 risk variants have augmented toxicity to kidney cells. However, the biological characteristics of APOL1 and its risk variants are not well elucidated. The APOL1 protein can be divided into several functional domains, including signal peptide (SP), pore forming domain (PFD), membrane address domain (MAD), and SRA-interacting domain. To investigate the relative contribution of each domain to cell injury, we constructed a serial expression vectors to delete or express each domain. These vectors were transfected into the human embryonic kidney cell line 293T, and then compared the cytotoxicity. In addition, we conducted studies in which APOL1 wild type (G0) was co-transfected in combination with G1 or G2 to see whether G0 could counteract the toxicity of the risk variants. The results showed that deleting the SP did not abolish the toxicity of APOL1, though deletion of 26 amino acid residues of the mature peptide at the N-terminal partially decreased the toxicity. Deleting PFD or MAD or SRA-interacting domain abolished toxicity, while, overexpressing each domain alone could not cause toxicity to the host cells. Deletion of the G2 sites while retaining G1 sites in the risk state resulted in persistent toxicity. Either deletion or exchanging the BH3 domain in the PFD led to complete loss of the toxicity in this experimental platform. Adding G0 to either G1 or G2 did not attenuate the toxicity of the either moiety. These results indicate that the integrity of the mature APOL1 protein is indispensable for its toxicity. Our study not only reveals the contribution of each domain of the APOL1 protein to cell injury, but also highlights some potential suggested targets for drug design to prevent or treat APOL1-associated nephropathy.


Asunto(s)
Apolipoproteínas/genética , Variación Genética , Enfermedades Renales/genética , Lipoproteínas HDL/genética , Negro o Afroamericano/genética , Apolipoproteína L1 , Apolipoproteínas/metabolismo , Genotipo , Células HEK293 , Humanos , Lipoproteínas HDL/metabolismo , Estructura Terciaria de Proteína , Factores de Riesgo
14.
Exp Mol Pathol ; 98(3): 491-501, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25796344

RESUMEN

Clinical reports have demonstrated that higher rates of non-diabetic glomerulosclerosis in African Americans can be attributed to two coding sequence variants (G1 and G2) in the APOL1 gene; however, the underlying mechanism is still unknown. Kidney biopsy data suggest enhanced expression of APOL1/APOL1 variants (Vs) in smooth muscle cells (SMCs) of renal vasculature. Since APOL1 is a secretory protein of relatively low molecular weight (41kDa), SMCs may be a contributory endocrine/paracrine source of APOL1 wild type (WT)/APOL1Vs in the glomerular capillary perfusate percolating podocytes. In the present study, we tested the hypothesis that an HIV milieu stimulated secretion of APOL1 and its risk variants by arterial SMCs contributes to podocyte injury. Human umbilical artery smooth muscle cells (HSMCs)-treated with conditioned media (CM) of HIV-infected peripheral mononuclear cells (PBMC/HIV-CM), CM of HIV-infected U939 cells, or recombinant IFN-γ displayed enhanced expression of APOL1. Podocytes co-cultured in trans-wells with HSMCs-over expressing APOL1WT showed induction of injury; however, podocytes co-cultured with HSMC-over expressing either APOL1G1 or APOL1G2 showed several folds greater injury when compared to HSMC-over expressing APOL1WT. Conditioned media collected from HSMC-over-expressing APOL1G1/APOL1G2 (HSMC/APOL1G1-CM or HSMC/APOL1G2-CM) also displayed higher percentages of injured podocytes in the form of swollen cells, leaky lysosomes, loss of viability, and enhanced sensitivity to adverse host factors when compared to HSMC/APOL1WT-CM. Notably, HSMC/APOL1WT-CM promoted podocyte injury only at a significantly higher concentrations compared to HSMC/APOL1G1/G2-CM. We conclude that HSMCs could serve as an endocrine/paracrine source of APOL1Vs, which mediate accelerated podocyte injury in HIV milieu.


Asunto(s)
Apolipoproteínas/metabolismo , Lipoproteínas HDL/metabolismo , Músculo Liso Vascular/metabolismo , Podocitos/metabolismo , Apolipoproteína L1 , Apolipoproteínas/genética , Línea Celular , Medios de Cultivo Condicionados/farmacología , VIH/patogenicidad , Humanos , Lipoproteínas HDL/genética , Monocitos/metabolismo , Monocitos/virología , Músculo Liso Vascular/efectos de los fármacos , Podocitos/efectos de los fármacos
15.
J Am Soc Nephrol ; 25(9): 1991-2002, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24676636

RESUMEN

FSGS is characterized by segmental scarring of the glomerulus and is a leading cause of kidney failure. Identification of genes causing FSGS has improved our understanding of disease mechanisms and points to defects in the glomerular epithelial cell, the podocyte, as a major factor in disease pathogenesis. Using a combination of genome-wide linkage studies and whole-exome sequencing in a kindred with familial FSGS, we identified a missense mutation R431C in anillin (ANLN), an F-actin binding cell cycle gene, as a cause of FSGS. We screened 250 additional families with FSGS and found another variant, G618C, that segregates with disease in a second family with FSGS. We demonstrate upregulation of anillin in podocytes in kidney biopsy specimens from individuals with FSGS and kidney samples from a murine model of HIV-1-associated nephropathy. Overexpression of R431C mutant ANLN in immortalized human podocytes results in enhanced podocyte motility. The mutant anillin displays reduced binding to the slit diaphragm-associated scaffold protein CD2AP. Knockdown of the ANLN gene in zebrafish morphants caused a loss of glomerular filtration barrier integrity, podocyte foot process effacement, and an edematous phenotype. Collectively, these findings suggest that anillin is important in maintaining the integrity of the podocyte actin cytoskeleton.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Proteínas de Microfilamentos/genética , Mutación , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Movimiento Celular/genética , Secuencia Conservada , Proteínas Contráctiles/genética , Proteínas del Citoesqueleto/metabolismo , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Exoma , Femenino , Técnicas de Silenciamiento del Gen , Barrera de Filtración Glomerular/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Mutantes/genética , Linaje , Podocitos/metabolismo , Homología de Secuencia de Aminoácido , Regulación hacia Arriba , Pez Cebra , Proteínas de Pez Cebra/genética
16.
Am J Physiol Renal Physiol ; 307(3): F326-36, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899058

RESUMEN

Development of higher rates of nondiabetic glomerulosclerosis (GS) in African Americans has been attributed to two coding sequence variants (G1 and G2) in the APOL1 gene. To date, the cellular function and the role of APOL1 variants (Vs) in GS are still unknown. In this study, we examined the effects of overexpressing wild-type (G0) and kidney disease risk variants (G1 and G2) of APOL1 in human podocytes using a lentivirus expression system. Interestingly, G0 inflicted podocyte injury only at a higher concentration; however, G1 and G2 promoted moderate podocyte injury at lower and higher concentrations. APOL1Vs expressing podocytes displayed diffuse distribution of both Lucifer yellow dye and cathepsin L as manifestations of enhanced lysosomal membrane permeability (LMP). Chloroquine attenuated the APOL1Vs-induced increase in podocyte injury, consistent with targeting lysosomes. The chloride channel blocker DIDS prevented APOL1Vs- induced injury, indicating a role for chloride influx in osmotic swelling of lysosomes. Direct exposure of noninfected podocytes with conditioned media from G1- and G2-expressing podocytes also induced injury, suggesting a contributory role of the secreted component of G1 and G2 as well. Adverse host factors (AHFs) such as hydrogen peroxide, hypoxia, TNF-α, and puromycin aminonucleoside augmented APOL1- and APOL1Vs-induced podocyte injury, while the effect of human immunodeficiency virus (HIV) on podocyte injury was overwhelming under conditions of APOLVs expression. We conclude that G0 and G1 and G2 APOL1 variants have the potential to induce podocyte injury in a manner which is further augmented by AHFs, with HIV infection being especially prominent.


Asunto(s)
Apolipoproteínas/genética , Apolipoproteínas/metabolismo , Variación Genética/genética , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Lisosomas/fisiología , Podocitos/metabolismo , Podocitos/patología , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Actinas/metabolismo , Negro o Afroamericano/etnología , Negro o Afroamericano/genética , Apolipoproteína L1 , Células Cultivadas , Canales de Cloruro/antagonistas & inhibidores , Canales de Cloruro/efectos de los fármacos , Cloroquina/farmacología , Predisposición Genética a la Enfermedad/etnología , Predisposición Genética a la Enfermedad/genética , Glomeruloesclerosis Focal y Segmentaria/etnología , Glomeruloesclerosis Focal y Segmentaria/genética , Humanos , Necrosis/fisiopatología , Permeabilidad , Podocitos/efectos de los fármacos
17.
Am J Physiol Renal Physiol ; 307(4): F369-84, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24944268

RESUMEN

Despite its success as a potent antineoplastic agent, ∼25% of patients receiving cisplatin experience acute kidney injury (AKI) and must discontinue therapy. Impaired magnesium homeostasis has been linked to cisplatin-mediated AKI, and because magnesium deficiency is widespread, we examined the effect of magnesium deficiency and replacement on cisplatin-induced AKI in physiologically relevant older female mice. Magnesium deficiency significantly increased cisplatin-associated weight loss and markers of renal damage (plasma blood urea nitrogen and creatinine), histological changes, inflammation, and renal cell apoptosis and modulated signaling pathways (e.g., ERK1/2, p53, and STAT3). Conversely, these damaging effects were reversed by magnesium. Magnesium deficiency alone significantly induced basal and cisplatin-mediated oxidative stress, whereas magnesium replacement attenuated these effects. Similar results were observed using cisplatin-treated LLC-PK1 renal epithelial cells exposed to various magnesium concentrations. Magnesium deficiency significantly amplified renal platinum accumulation, whereas magnesium replacement blocked the augmented platinum accumulation after magnesium deficiency. Increased renal platinum accumulation during magnesium deficiency was accompanied by reduced renal efflux transporter expression, which was reversed by magnesium replacement. These findings demonstrate the role of magnesium in regulating cisplatin-induced AKI by enhancing oxidative stress and thus promoting cisplatin-mediated damage. Additional in vitro experiments using ovarian, breast, and lung cancer cell lines showed that magnesium supplementation did not compromise cisplatin's chemotherapeutic efficacy. Finally, because no consistently successful therapy to prevent or treat cisplatin-mediated AKI is available for humans, these results support developing more conservative magnesium replacement guidelines for reducing cisplatin-induced AKI in cancer patients at risk for magnesium deficiency.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Magnesio/uso terapéutico , Infiltración Neutrófila/efectos de los fármacos , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Animales , Apoptosis/efectos de los fármacos , Nitrógeno de la Urea Sanguínea , Línea Celular Tumoral , Creatinina/sangre , Citocinas/biosíntesis , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Riñón/metabolismo , Células LLC-PK1 , Magnesio/metabolismo , Deficiencia de Magnesio/fisiopatología , Ratones , Estrés Oxidativo/efectos de los fármacos , Platino (Metal)/metabolismo , Factor de Transcripción STAT3/metabolismo , Porcinos
18.
Exp Mol Pathol ; 96(3): 431-7, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24768585

RESUMEN

Mammalian target of rapamycin (mTOR) has been reported to contribute to the development of HIV-associated nephropathy (HIVAN). We hypothesized that HIV may be activating renal tissue mTOR pathway through renin angiotensin system (RAS) via Angiotensin Receptor Type II receptor (AT2R). Renal tissues of Vpr transgenic and Tg26 (HIVAN) mice displayed enhanced phosphorylation of mTOR and p70S6K. Aliskiren, a renin inhibitor attenuated phosphorylation of both mTOR and p70S6K in renal tissues of HIVAN mice. Interestingly, Angiotensin Receptor Type I (AT1R) blockade did not modulate renal tissue phosphorylation of mTOR in HIVAN mice; on the other hand, AT2R blockade attenuated renal tissue phosphorylation of mTOR in HIVAN mice. In vitro studies, both renin and Ang II displayed enhanced mouse tubular cell (MTC) phosphorylation of p70S6K in a dose dependent manner. HIV/MTC also displayed enhanced phosphorylation of both mTOR and p70S6K; interestingly this effect of HIV was further enhanced by losartan (an AT1R blocker). On the other hand, AT2R blockade attenuated HIV-induced tubular cell phosphorylation of mTOR and p70S6K, whereas, AT2R agonist enhanced phosphorylation of mTOR and p70S6K. These findings indicate that HIV stimulates mTOR pathway in HIVAN through the activation of renin angiotensin system via AT2R.


Asunto(s)
Nefropatía Asociada a SIDA/genética , Enfermedades Renales/virología , Receptor de Angiotensina Tipo 2/metabolismo , Sistema Renina-Angiotensina/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Amidas/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/metabolismo , Bloqueadores del Receptor Tipo 2 de Angiotensina II/metabolismo , Animales , Fumaratos/farmacología , VIH , Enfermedades Renales/veterinaria , Losartán/farmacología , Ratones , Ratones Transgénicos , Fosforilación , Receptor de Angiotensina Tipo 2/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética
19.
Exp Cell Res ; 319(6): 779-89, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23384600

RESUMEN

Autophagy is a cellular pathway involved in protein and organelle degradation. It is relevant to many types of cellular homeostasis and human diseases. High level of glucose is known to inflict podocyte injury, but little is reported about the relationship between high concentrations of glucose and autophagy in these cells. The present study demonstrates that high glucose promotes autophagy in podocytes. Rapamycin further enhances this effect, but 3-methyadenine inhibits it. The proautophagic effect of high glucose manifested in the form of enhanced podocyte expression of LC3-2 and beclin-1; interestingly, antioxidants such as NAC were found to inhibit high glucose-induced autophagy. High glucose induced the generation of ROS by podocytes in a time-dependent manner. High glucose also enhanced podocyte expression of MnSOD and catalase. These findings indicate that high glucose-induced autophagy is mediated through podocyte ROS generation.


Asunto(s)
Autofagia , Glucosa/metabolismo , Podocitos/efectos de los fármacos , Acetilcisteína/farmacología , Adenina/análogos & derivados , Adenina/farmacología , Animales , Antioxidantes/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Catalasa/metabolismo , Medios de Cultivo/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Glucosa/farmacología , Masculino , Microscopía Electrónica , Proteínas Asociadas a Microtúbulos/metabolismo , Podocitos/metabolismo , Podocitos/ultraestructura , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Sirolimus/farmacología , Estreptozocina/efectos adversos , Superóxido Dismutasa/metabolismo , Células Tumorales Cultivadas
20.
Exp Cell Res ; 319(14): 2266-74, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23806280

RESUMEN

Mesenchymal stem cells (MSCs) secrete paracrine factors that could be cytoprotective and serve roles in immunoregulation during tissue injury. Although MSCs express HIV receptors, and co-receptors, and are susceptible to HIV infection, whether HIV-1 may affect biological properties of MSCs needs more study. We evaluated cellular proliferation, differentiation and paracrine functions of MSCs isolated from compact bones of healthy control mice and Tg26 HIV-1 transgenic mice. The ability of MSCs to protect against cisplatin toxicity was studied in cultured renal tubular cells as well as in intact mice. We successfully isolated MSCs from healthy mice and Tg26 HIV-1 transgenic mice and found the latter expressed viral Nef, Vpu, NL4-3 and Vif genes. The proliferation and differentiation of Tg26 HIV-1 MSCs was inferior to MSCs from healthy mice. Moreover, transplantation of Tg26 HIV-1 MSCs less effectively improved outcomes compared with healthy MSCs in mice with acute kidney injury. Also, Tg26 HIV-1 MSCs secreted multiple cytokines, but at significantly lower levels than healthy MSCs, which resulted in failure of conditioned medium from these MSCs to protect cultured renal tubular cells from cisplatin toxicity. Therefore, HIV-1 had adverse biological effects on MSCs extending to their proliferation, differentiation, function, and therapeutic potential. These findings will help in advancing mechanistical insight in renal injury and repair in the setting of HIV-1 infection.


Asunto(s)
Lesión Renal Aguda/terapia , Diferenciación Celular/genética , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Células Madre Mesenquimatosas/metabolismo , Comunicación Paracrina/genética , Lesión Renal Aguda/inducido químicamente , Animales , Huesos/citología , Proliferación Celular , Cisplatino , Citocinas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Terapia Genética , VIH-1 , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Péptidos y Proteínas de Señalización Intercelular , Túbulos Renales/patología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/patología , Ratones , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA