Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JBMR Plus ; 4(7): e10080, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32666016

RESUMEN

CSF-1 is a key factor in regulating bone remodeling; osteocytes express CSF-1 and its receptor. Viable osteocytes are essential for bone remodeling through cell-cell contact and secretion of factors that regulate osteoblasts and osteoclasts. Increased oxidative stress contributes to osteocyte death and correlates with bone loss during aging. The NADPH oxidase Nox4 is a major source of ROS in bone. CSF-1 decreases Nox4, suggesting that CSF-1 protects against oxidative stress. Here, we show that osteocyte apoptosis previously reported in our global CSF-1KO mice is associated with increased Nox4, as well as 4-HNE expression in osteocytes. Osteocytes isolated from CSF-1KO mice were less viable and showed increased intracellular ROS, elevated NADPH oxidase activity/Nox4 protein, activation of mTOR/S6K, and downstream apoptosis signals compared with WT osteocytes. Nox4 expression was also increased in CSF-1KO osteocytes and colocalized with MitoTracker Red in mitochondria. Notably, CSF-1 inhibited Nox4 expression and apoptosis cascade signals. In additional studies, shNox4 decreased these signals in CSF-1KO osteocytes, whereas overexpression of Nox4 in WT osteocytes activated the apoptosis pathway. To determine the role of CSF-1 in osteocytes, DMP1Cre-CSF-1cKO (CSF-1cKO) mice that lack CSF-1 in osteocytes/late osteoblasts were developed. Osteocyte defects in CSF-1cKO mice overlapped with those in CSF-1KO mice, including increased apoptosis, Nox4, and 4-HNE-expressing osteocytes. CSF-1cKO mice showed unbalanced cancellous bone remodeling with decreased bone formation and resorption. Continued exposure to high Nox4/ROS levels may further compromise bone formation and predispose to bone loss and skeletal fragility. Taken together, our findings suggest a novel link between CSF-1, Nox4-derived ROS, and osteocyte survival/function that is crucial for osteocyte-mediated bone remodeling. Results reveal new mechanisms by which CSF-1/oxidative stress regulate osteocyte homeostasis, which may lead to therapeutic strategies to improve skeletal health in aging. © 2018 American Society for Bone and Mineral Research.

2.
Mol Metab ; 36: 100968, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32240965

RESUMEN

OBJECTIVE: Glomerular injury is a prominent pathological feature of diabetic kidney disease (DKD). Constitutively active NADPH oxidase 4 (Nox4) is a major source of reactive oxygen species that mediates hyperglycemia-induced mesangial cell (MC) fibrotic injury. However, the mechanism that Nox4 utilizes to achieve its biological outcome remains elusive, and the signaling pathways that regulate this isoform oxidase are not well understood. Here, our goal is to study the detailed mechanism by which NAPDH oxidase 4 (Nox4) is post-transcriptionally regulated in MC during diabetic pathology. METHODS: We studied the protein expression of HuR, Nox4 and matrix proteins by western blotting, while we assessed the mRNA stability of Nox4 by RT-PCR and polysomal assay, examined in vitro cultured glomerular mesangial cells treated by high glucose (HG) and diabetic animal induced by STZ. The binding assay between HuR and the Nox4 promoter was done by immuno-precipiating with HuR antibody and detecting the presence of Nox4 mRNA, or by pull-down by using biotinlyated labeled Nox4 promoter RNA and detecting the presence of the HuR protein. The binding was also confirmed in MCs where Nox4 promoter-containing luciferage constructs were transfected. ROS levels were measured with DHE/DCF dyes in cells, or lucigenin chemiluminescence for Nox enzymatic levels, or HPLC assay for superoxide. HuR protein was inhibited by antisense oligo that utilized osmotic pumps for continuous delivery in animal models. The H1bAc1 ratio was measured by an ELISA kit for mice. RESULTS: We demonstrate that in MCs, high glucose (HG) elicits a rapid upregulation of Nox4 protein via translational mechanisms. Nox4 mRNA 3' untranslated region (3'-UTR) contains numerous AU-rich elements (AREs) that are potential binding sites for the RNA-binding protein human antigen R (HuR). We show that HG promotes HuR activation/expression and that HuR is required for HG-induced Nox4 protein expression/mRNA translation, ROS generation, and subsequent MC fibrotic injury. Through a series of invitro RNA-binding assays, we demonstrate that HuR acts via binding to AREs in Nox4 3'-UTR in response to HG. The invivo relevance of these observations is confirmed by the findings that increased Nox4 is accompanied by the binding of HuR to Nox4 mRNA in kidneys from type 1 diabetic animals, and further suppressing HuR expression showed a reno-protective role in a type 1 diabetic mouse model via reducing MC injury, along with the improvement of hyperglycemia and renal function. CONCLUSIONS: We established for the first time that HuR-mediated translational regulation of Nox4 contributes to the pathogenesis of fibrosis of the glomerular microvascular bed. Thus therapeutic interventions affecting the interplay between Nox4 and HuR could be exploited as valuable tools in designing treatments for DKD.


Asunto(s)
Nefropatías Diabéticas/genética , Proteína 1 Similar a ELAV/metabolismo , NADPH Oxidasa 4/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus/metabolismo , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/terapia , Proteína 1 Similar a ELAV/genética , Glucosa/metabolismo , Humanos , Hiperglucemia/metabolismo , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 4/genética , Proteínas de Unión al ARN/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
3.
PLoS One ; 13(11): e0207285, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30444896

RESUMEN

TGFß promotes podocyte hypertrophy and expression of matrix proteins in fibrotic kidney diseases such as diabetic nephropathy. Both mTORC1 and mTORC2 are hyperactive in response to TGFß in various renal diseases. Deptor is a component of mTOR complexes and a constitutive inhibitor of their activities. We identified that deptor downregulation by TGFß maintains hyperactive mTOR in podocytes. To unravel the mechanism, we found that TGFß -initiated noncanonical signaling controls deptor inhibition. Pharmacological inhibitor of PI 3 kinase, Ly 294002 and pan Akt kinase inhibitor MK 2206 prevented the TGFß induced downregulation of deptor, resulting in suppression of both mTORC1 and mTORC2 activities. However, specific isoform of Akt involved in this process is not known. We identified Akt2 as predominant isoform expressed in kidney cortex, glomeruli and podocytes. TGFß time-dependently increased the activating phosphorylation of Akt2. Expression of dominant negative PI 3 kinase and its signaling inhibitor PTEN blocked Akt2 phosphorylation by TGFß. Inhibition of Akt2 using a phospho-deficient mutant that inactivates its kinase activity, as well as siRNA against the kinase markedly diminished TGFß -mediated deptor suppression, its association with mTOR and activation of mTORC1 and mTORC2. Importantly, inhibition of Akt2 blocked TGFß -induced podocyte hypertrophy and expression of the matrix protein fibronectin. This inhibition was reversed by the downregulation of deptor. Interestingly, we detected increased phosphorylation of Akt2 concomitant with TGFß expression in the kidneys of diabetic rats. Thus, our data identify previously unrecognized Akt2 kinase as a driver of TGFß induced deptor downregulation and sustained mTORC1 and mTORC2 activation. Furthermore, we provide the first evidence that deptor downstream of Akt2 contributes to podocyte hypertrophy and matrix protein expression found in glomerulosclerosis in different renal diseases.


Asunto(s)
Regulación hacia Abajo , Fibronectinas/biosíntesis , Regulación Enzimológica de la Expresión Génica , Podocitos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/biosíntesis , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular , Cromonas/farmacología , Hipertrofia , Diana Mecanicista del Complejo 1 de la Rapamicina/biosíntesis , Diana Mecanicista del Complejo 2 de la Rapamicina/biosíntesis , Morfolinas/farmacología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Podocitos/patología , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta/farmacología
4.
Oxid Med Cell Longev ; 2017: 3296294, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28690762

RESUMEN

Sestrin2 (SESN2), a highly conserved stress-inducible metabolic protein, is known to repress reactive oxygen species (ROS) and provide cytoprotection against various noxious stimuli including genotoxic and oxidative stress, endoplasmic reticulum (ER) stress, and hypoxia. Studies demonstrate that the upregulation of Sestrin2 under conditions of oxidative stress augments autophagy-directed degradation of Kelch-like ECH-associated protein 1 (Keap1), which targets and breaks down nuclear erythroid-related factor 2 (Nrf2), a key regulator of various antioxidant genes. Moreover, ER stress and hypoxia are shown to induce Sestrins, which ultimately reduce cellular ROS levels. Sestrin2 also plays a pivotal role in metabolic regulation through activation of the key energy sensor AMP-dependent protein kinase (AMPK) and inhibition of mammalian target of rapamycin complex 1 (mTORC1). Other downstream effects of Sestrins include autophagy activation, antiapoptotic effects in normal cells, and proapoptotic effects in cancer cells. As perturbations in the aforementioned pathways are well documented in multiple diseases, Sestrin2 might serve as a potential therapeutic target for various diseases. Thus, the aim of this review is to discuss the upstream regulators and the downstream effectors of Sestrins and to highlight the significance of Sestrin2 as a biomarker and a therapeutic target in diseases such as metabolic disorders, cardiovascular and neurodegenerative diseases, and cancer.


Asunto(s)
Biomarcadores/metabolismo , Animales , Antioxidantes/metabolismo , Autofagia/fisiología , Estrés del Retículo Endoplásmico/fisiología , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Nucleares , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo
5.
J Biol Chem ; 292(14): 5665-5675, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28188286

RESUMEN

High-glucose increases NADPH oxidase 4 (NOX4) expression, reactive oxygen species generation, and matrix protein synthesis by inhibiting AMP-activated protein kinase (AMPK) in renal cells. Because hydrogen sulfide (H2S) inhibits high glucose-induced matrix protein increase by activating AMPK in renal cells, we examined whether H2S inhibits high glucose-induced expression of NOX4 and matrix protein and whether H2S and NO pathways are integrated. High glucose increased NOX4 expression and activity at 24 h in renal proximal tubular epithelial cells, which was inhibited by sodium hydrosulfide (NaHS), a source of H2S. High glucose decreased AMPK phosphorylation and activity, which was restored by NaHS. Compound C, an AMPK inhibitor, prevented NaHS inhibition of high glucose-induced NOX4 expression. NaHS inhibition of high glucose-induced NOX4 expression was abrogated by N(ω)-nitro-l-arginine methyl ester, an inhibitor of NOS. NaHS unexpectedly augmented the expression of inducible NOS (iNOS) but not endothelial NOS. iNOS siRNA and 1400W, a selective iNOS inhibitor, abolished the ameliorative effects of NaHS on high glucose-induced NOX4 expression, reactive oxygen species generation, and, matrix laminin expression. Thus, H2S recruits iNOS to generate NO to inhibit high glucose-induced NOX4 expression, oxidative stress, and matrix protein accumulation in renal epithelial cells; the two gasotransmitters H2S and NO and their interaction may serve as therapeutic targets in diabetic kidney disease.


Asunto(s)
Células Epiteliales/enzimología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Sulfuro de Hidrógeno/farmacología , Túbulos Renales Proximales/enzimología , NADPH Oxidasas/biosíntesis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Amidinas/farmacología , Animales , Bencilaminas/farmacología , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/terapia , Células Epiteliales/patología , Proteínas de la Matriz Extracelular/metabolismo , Túbulos Renales Proximales/patología , Ratones , NADPH Oxidasa 4 , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos
6.
Antioxid Redox Signal ; 25(12): 639-641, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27357313

RESUMEN

Both acute kidney injury (AKI) and chronic kidney disease (CKD) are major causes of renal failure in humans and are associated with high incidences of morbidity and mortality rates. AKI and CKD are closely interconnected, and fueled by the obesity and diabetes epidemic, their prevalence is alarmingly increasing to the point that it currently represents a major heath issue worldwide. The kidney is an organ that is particularly sensitive to redox imbalance, resulting in excessive production of reactive oxygen species. Oxidative stress is viewed as a critical pathogenic factor implicated in the initiation, development, and progression of most renal diseases. This Forum discusses the redox-dependent factors and mechanisms accounting for the perturbation of renal function and circulation in the context of the major kidney pathologies linked to hypertension, diabetes, and cancer. Antioxid. Redox Signal. 25, 639-641.


Asunto(s)
Lesión Renal Aguda/metabolismo , Fallo Renal Crónico/metabolismo , Riñón/metabolismo , Insuficiencia Renal Crónica/metabolismo , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Complicaciones de la Diabetes , Progresión de la Enfermedad , Humanos , Hipertensión/metabolismo , Hipertensión/patología , Riñón/patología , Fallo Renal Crónico/patología , Obesidad/complicaciones , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/patología
7.
Antioxid Redox Signal ; 25(13): 703-719, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27393154

RESUMEN

AIM: Podocyte apoptosis is a critical mechanism for excessive loss of urinary albumin that eventuates in kidney fibrosis. Oxidative stress plays a critical role in hyperglycemia-induced glomerular injury. We explored the hypothesis that mammalian target of rapamycin complex 2 (mTORC2) mediates podocyte injury in diabetes. RESULTS: High glucose (HG)-induced podocyte injury reflected by alterations in the slit diaphragm protein podocin and podocyte depletion/apoptosis. This was paralleled by activation of the Rictor/mTORC2/Akt pathway. HG also increased the levels of Nox4 and NADPH oxidase activity. Inhibition of mTORC2 using small interfering RNA (siRNA)-targeting Rictor in vitro decreased HG-induced Nox1 and Nox4, NADPH oxidase activity, restored podocin levels, and reduced podocyte depletion/apoptosis. Inhibition of mTORC2 had no effect on mammalian target of rapamycin complex 1 (mTORC1) activation, described by our group to be increased in diabetes, suggesting that the mTORC2 activation by HG could mediate podocyte injury independently of mTORC1. In isolated glomeruli of OVE26 mice, there was a similar activation of the Rictor/mTORC2/Akt signaling pathway with increase in Nox4 and NADPH oxidase activity. Inhibition of mTORC2 using antisense oligonucleotides targeting Rictor restored podocin levels, reduced podocyte depletion/apoptosis, and attenuated glomerular injury and albuminuria. INNOVATION: Our data provide evidence for a novel function of mTORC2 in NADPH oxidase-derived reactive oxygen species generation and podocyte apoptosis that contributes to urinary albumin excretion in type 1 diabetes. CONCLUSION: mTORC2 and/or NADPH oxidase inhibition may represent a therapeutic modality for diabetic kidney disease. Antioxid. Redox Signal. 25, 703-719.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Tipo 1/complicaciones , Glucosa/farmacología , Complejos Multiproteicos/metabolismo , NADPH Oxidasas/genética , Podocitos/citología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis , Proteínas Portadoras/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina , Proteínas de la Membrana/metabolismo , Ratones , NADPH Oxidasa 4 , NADPH Oxidasas/metabolismo , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Am J Physiol Renal Physiol ; 311(1): F1-F11, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27147669

RESUMEN

Systemic fibrosis from gadolinium-based magnetic resonance imaging contrast is a scourge for the afflicted. Although gadolinium-associated systemic fibrosis is a rare condition, the threat of litigation has vastly altered clinical practice. Most theories concerning the etiology of the fibrosis are grounded in case reports rather than experiment. This has led to the widely accepted conjecture that the relative affinity of certain contrast agents for the gadolinium ion inversely correlates with the risk of succumbing to the disease. How gadolinium-containing contrast agents trigger widespread and site-specific systemic fibrosis and how chronicity is maintained are largely unknown. This review highlights experimentally-derived information from our laboratory and others that pertain to our understanding of the pathophysiology of gadolinium-associated systemic fibrosis.


Asunto(s)
Medios de Contraste/efectos adversos , Fibrosis/inducido químicamente , Gadolinio/efectos adversos , Imagen por Resonancia Magnética/efectos adversos , Fibrosis/epidemiología , Fibrosis/patología , Humanos
9.
FASEB J ; 30(9): 3026-38, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27221979

RESUMEN

Systemic fibrosis can be induced in humans with gadolinium-based contrast, and cumulative doses correlate with severity. Bone marrow-derived fibrocytes accumulate in the dermis. Whether target organs liberate chemokines to recruit these fibrocytes or whether fibrocytes are stimulated to home to the affected tissue is unknown. Transgenic (tagged) donor rats were treated with gadolinium-based contrast. Bone marrow was obtained from diseased animals and age-matched controls. Rats with subtotal nephrectomies were lethally irradiated and underwent salvage transplantation with either the contrast-naïve or contrast-exposed bone marrow. Groups were randomly assigned to control or contrast treatment. Contrast treatment led to dermal fibrosis, and this was exacerbated in recipients of contrast-exposed marrow. Fibronectin, C-C chemokine receptors (CCRs)2 and 7, and oxidative stress were all increased in skin from contrast-treated animals-all parameters more severe in recipients of contrast-treated animals. The respective ligands, monocyte chemoattractant protein and C-C motif ligand 19, were both elevated in skin from contrast-treated animals. Coadministration of gadolinium-based contrast and a CCR2 inhibitor reduced the severity of skin disease as well as dermal cellularity. The functional role of chemokines in the effects of gadolinium-based contrast was further confirmed in in situ coculture studies using neutralizing CCR2 antibodies. These data implicate dermal liberation of specific chemokines in the recruitment of circulating bone marrow-derived cells. The disease is augmented by bone marrow exposure to contrast, which explains why multiple exposures correlate with severity.-Drel, V. R., Tan, C., Barnes, J. L., Gorin, Y., Lee, D.-Y., Wagner, B. Centrality of bone marrow in the severity of gadolinium-based contrast-induced systemic fibrosis.


Asunto(s)
Médula Ósea/efectos de los fármacos , Medios de Contraste/efectos adversos , Gadolinio DTPA/efectos adversos , Dermopatía Fibrosante Nefrogénica/inducido químicamente , Animales , Animales Modificados Genéticamente , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Trasplante de Médula Ósea , Medios de Contraste/administración & dosificación , Medios de Contraste/farmacología , Femenino , Gadolinio DTPA/metabolismo , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Masculino , NADPH Oxidasa 4 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Dermopatía Fibrosante Nefrogénica/patología , Distribución Aleatoria , Ratas , Especies Reactivas de Oxígeno , Receptores CCR2/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Piel/efectos de los fármacos , Piel/metabolismo
10.
Am J Pathol ; 185(8): 2168-80, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26071397

RESUMEN

Transforming growth factor (TGF)-ß contributes to tubulointerstitial fibrosis. We investigated the mechanism by which TGF-ß exerts its profibrotic effects and specifically the role of AMP-activated protein kinase (AMPK) in kidney tubular epithelial cells and interstitial fibroblasts. In proximal tubular epithelial cells, TGF-ß1 treatment causes a decrease in AMPK phosphorylation and activation together with increased fibronectin and α-smooth muscle actin expression and decreased in E-cadherin. TGF-ß1 causes similar changes in interstitial fibroblasts. Activation of AMPK with 5-aminoimidazole-4-carboxamide 1-ß-d-ribofuranoside, metformin, or overexpression of constitutively active AMPK markedly attenuated TGF-ß1 functions. Conversely, inhibition of AMPK with adenine 9-ß-d-arabinofuranoside or siRNA-mediated knockdown of AMPK (official name PRKAA1) mimicked the effect of TGF-ß1 and enhanced basal and TGF-ß1-induced phenotypic changes. Importantly, we found that tuberin contributed to the protective effects of AMPK and that TGF-ß1 promoted cell injury by blocking AMPK-mediated tuberin phosphorylation and activation. In the kidney cortex of TGF-ß transgenic mice, the significant decrease in AMPK phosphorylation and tuberin phosphorylation on its AMPK-dependent activating site was associated with an increase in mesenchymal markers and a decrease in E-cadherin. Collectively, the data indicate that TGF-ß exerts its profibrotic action in vitro and in vivo via inactivation of AMPK. AMPK and tuberin activation prevent tubulointerstitial injury induced by TGF-ß. Activators of AMPK provide potential therapeutic strategy to prevent kidney fibrosis and progressive kidney disease.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Miofibroblastos/metabolismo , Factor de Crecimiento Transformador beta1/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Cadherinas/metabolismo , Fibronectinas/metabolismo , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Ratones , Ratones Noqueados , Miofibroblastos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ribonucleósidos/farmacología , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
11.
Mol Cells ; 38(4): 285-96, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25824546

RESUMEN

Oxidative stress has been linked to the pathogenesis of diabetic nephropathy, the complication of diabetes in the kidney. NADPH oxidases of the Nox family, and in particular the homologue Nox4, are a major source of reactive oxygen species in the diabetic kidney and are critical mediators of redox signaling in glomerular and tubulointerstitial cells exposed to the diabetic milieu. Here, we present an overview of the current knowledge related to the understanding of the role of Nox enzymes in the processes that control mesangial cell, podocyte and tubulointerstitial cell injury induced by hyperglycemia and other predominant factors enhanced in the diabetic milieu, including the renin-angiotensin system and transforming growth factor-ß. The nature of the upstream modulators of Nox enzymes as well as the downstream targets of the Nox NADPH oxidases implicated in the propagation of the redox processes that alter renal biology in diabetes will be highlighted.


Asunto(s)
Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , NADPH Oxidasas/metabolismo , Animales , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/patología , Humanos , Terapia Molecular Dirigida , Estrés Oxidativo , Transducción de Señal
12.
J Biol Chem ; 290(19): 12014-26, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25752605

RESUMEN

Diabetes-induced kidney cell injury involves an increase in matrix protein expression that is only partly alleviated by current treatment, prompting a search for new modalities. We have previously shown that hydrogen sulfide (H2S) inhibits high glucose-induced protein synthesis in kidney podocytes. We tested whether tadalafil, a phosphodiesterase 5 inhibitor used to treat erectile dysfunction, ameliorates high glucose stimulation of matrix proteins by generating H2S in podocytes. Tadalafil abrogated high glucose stimulation of global protein synthesis and matrix protein laminin γ1. Tadalafil inhibited high glucose-induced activation of mechanistic target of rapamycin complex 1 and laminin γ1 accumulation in an AMP-activated protein kinase (AMPK)-dependent manner. Tadalafil increased AMPK phosphorylation by stimulating calcium-calmodulin kinase kinase ß. Tadalafil rapidly increased the expression and activity of the H2S-generating enzyme cystathionine γ-lyase (CSE) by promoting its translation. dl-Propargylglycine, a CSE inhibitor, and siRNA against CSE inhibited tadalafil-induced AMPK phosphorylation and abrogated the tadalafil effect on high glucose stimulation of laminin γ1. In tadalafil-treated podocytes, we examined the interaction between H2S and nitric oxide (NO). N(ω)-Nitro-L-arginine methyl ester and 1H-[1,2,4]-oxadiazolo-[4,3-a]-quinoxalin-1-one, inhibitors of NO synthase (NOS) and soluble guanylyl cyclase, respectively, abolished tadalafil induction of H2S and AMPK phosphorylation. Tadalafil rapidly augmented inducible NOS (iNOS) expression by increasing its mRNA, and siRNA for iNOS and 1400W, an iNOS blocker, inhibited tadalafil stimulation of CSE expression and AMPK phosphorylation. We conclude that tadalafil amelioration of high glucose stimulation of synthesis of proteins including matrix proteins in podocytes requires integration of the NO-H2S-AMPK axis leading to the inhibition of high glucose-induced mechanistic target of rapamycin complex 1 activity and mRNA translation.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Carbolinas/química , Glucosa/química , Sulfuro de Hidrógeno/química , Óxido Nítrico/química , Podocitos/metabolismo , Transducción de Señal , Animales , Calcio/química , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Riñón/citología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Inhibidores de Fosfodiesterasa 5/química , Fosforilación , Podocitos/citología , Polirribosomas/metabolismo , Ratas , Serina-Treonina Quinasas TOR/metabolismo , Tadalafilo
13.
Am J Physiol Renal Physiol ; 308(11): F1276-87, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25656366

RESUMEN

Reactive oxygen species (ROS) generated by Nox NADPH oxidases may play a critical role in the pathogenesis of diabetic nephropathy (DN). The efficacy of the Nox1/Nox4 inhibitor GKT137831 on the manifestations of DN was studied in OVE26 mice, a model of type 1 diabetes. Starting at 4-5 mo of age, OVE26 mice were treated with GKT137831 at 10 or 40 mg/kg, once-a-day for 4 wk. At both doses, GKT137831 inhibited NADPH oxidase activity, superoxide generation, and hydrogen peroxide production in the renal cortex from diabetic mice without affecting Nox1 or Nox4 protein expression. The increased expression of fibronectin and type IV collagen was reduced in the renal cortex, including glomeruli, of diabetic mice treated with GKT137831. GKT137831 significantly reduced glomerular hypertrophy, mesangial matrix expansion, urinary albumin excretion, and podocyte loss in OVE26 mice. GKT137831 also attenuated macrophage infiltration in glomeruli and tubulointerstitium. Collectively, our data indicate that pharmacological inhibition of Nox1/4 affords broad renoprotection in mice with preexisting diabetes and established kidney disease. This study validates the relevance of targeting Nox4 and identifies GKT137831 as a promising compound for the treatment of DN in type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Inhibidores Enzimáticos/farmacología , NADH NADPH Oxidorreductasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Animales , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , Riñón/metabolismo , Riñón/patología , Ratones , NADPH Oxidasa 1 , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Pirazolonas , Piridonas , Especies Reactivas de Oxígeno/metabolismo
14.
Cell Signal ; 27(1): 1-5, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25283600

RESUMEN

Circulating symmetric dimethylarginine (SDMA) is increased in patients with chronic kidney disease. SDMA is considered an inert metabolite, but because it can transported into cells, we studied the effect of SDMA on glomerular endothelial cells. SDMA suppressed VEGF-induced endothelial nitric oxide synthase (eNOS) phosphorylation and nitric oxide production, but not VEGFR2 activation and signaling leading to eNOS activation. SDMA caused eNOS uncoupling and increased superoxide anion production in response to VEGF. All these effects were blocked by preventing cellular uptake of SDMA with a molar excess of arginine. These data show that SDMA interferes with nitric oxide production by uncoupling eNOS and leads to oxidative stress in glomerular endothelial cells. In conclusion, our data show that SDMA is not an inert metabolite and that it could contribute to oxidative stress in the renal endothelium.


Asunto(s)
Arginina/análogos & derivados , Células Endoteliales/metabolismo , Glomérulos Renales/citología , Óxido Nítrico/metabolismo , Animales , Arginina/farmacología , Canales de Calcio/metabolismo , Línea Celular , Células Endoteliales/efectos de los fármacos , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo , Multimerización de Proteína/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Superóxidos/metabolismo , Canales Catiónicos TRPV/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
15.
Am J Physiol Renal Physiol ; 307(2): F159-71, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24872317

RESUMEN

The small G proteins Rac1 and RhoA regulate actin cytoskeleton, cell shape, adhesion, migration, and proliferation. Recent studies in our laboratory have shown that NADPH oxidase Nox4-derived ROS are involved in transforming growth factor (TGF)-ß1-induced rat kidney myofibroblast differentiation assessed by the acquisition of an α-smooth muscle actin (α-SMA) phenotype and expression of an alternatively spliced fibronectin variant (Fn-EIIIA). Rac1 and RhoA are essential in signaling by some Nox homologs, but their role as effectors of Nox4 in kidney myofibroblast differentiation is not known. In the present study, we explored a link among Rac1 and RhoA and Nox4-dependent ROS generation in TGF-ß1-induced kidney myofibroblast activation. TGF-ß1 stimulated an increase in Nox4 protein expression, NADPH oxidase activity, and abundant α-SMA and Fn-EIIIA expression. RhoA but not Rac1 was involved in TGF-ß1 induction of Nox4 signaling of kidney myofibroblast activation. TGF-ß1 stimulated active RhoA-GTP and increased Rho kinase (ROCK). Inhibition of RhoA with small interfering RNA and ROCK using Y-27632 significantly reduced TGF-ß1-induced stimulation of Nox4 protein, NADPH oxidase activity, and α-SMA and Fn-EIIIA expression. Treatment with diphenyleneiodonium, an inhibitor of NADPH oxidase, did not decrease RhoA activation but inhibited TGF-ß1-induced α-SMA and Fn-EIIIA expression, indicating that RhoA is upstream of ROS generation. RhoA/ROCK also regulated polymerase (DNA-directed) δ-interacting protein 2 (Poldip2), a newly discovered Nox4 enhancer protein. Collectively, these data indicate that RhoA/ROCK is upstream of Poldip2-dependent Nox4 regulation and ROS production and induces redox signaling of kidney myofibroblast activation and may broader implications in the pathophysiology of renal fibrosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Riñón/enzimología , Miofibroblastos/enzimología , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Diferenciación Celular , Línea Celular , Movimiento Celular , Activación Enzimática , Fibronectinas/metabolismo , Fibrosis , Riñón/patología , Miofibroblastos/patología , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Oxidación-Reducción , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Ratas , Transducción de Señal , Factores de Tiempo , Transfección , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética
16.
Am J Physiol Cell Physiol ; 306(11): C1089-100, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24740537

RESUMEN

Platelet-derived growth factor BB and its receptor (PDGFRß) play a pivotal role in the development of renal glomerular mesangial cells. Their roles in increased mesangial cell proliferation during mesangioproliferative glomerulonephritis have long been noted, but the operating logic of signaling mechanisms regulating these changes remains poorly understood. We examined the role of a recently identified MAPK, Erk5, in this process. PDGF increased the activating phosphorylation of Erk5 and tyrosine phosphorylation of proteins in a time-dependent manner. A pharmacologic inhibitor of Erk5, XMD8-92, abrogated PDGF-induced DNA synthesis and mesangial cell proliferation. Similarly, expression of dominant negative Erk5 or siRNAs against Erk5 blocked PDGF-stimulated DNA synthesis and proliferation. Inhibition of Erk5 attenuated expression of cyclin D1 mRNA and protein, resulting in suppression of CDK4-mediated phosphorylation of the tumor suppressor protein pRb. Expression of cyclin D1 or CDK4 prevented the dominant negative Erk5- or siErk5-mediated inhibition of DNA synthesis and mesangial cell proliferation induced by PDGF. We have previously shown that phosphatidylinositol 3-kinase (PI3-kinase) contributes to PDGF-induced proliferation of mesangial cells. Inhibition of PI3-kinase blocked PDGF-induced phosphorylation of Erk5. Since PI3-kinase acts through Akt, we determined the role of Erk5 on Akt phosphorylation. XMD8-92, dominant negative Erk5, and siErk5 inhibited phosphorylation of Akt by PDGF. Interestingly, we found inhibition of PDGF-induced Erk5 phosphorylation by a pharmacological inhibitor of Akt kinase and kinase dead Akt in mesangial cells. Thus our data unfold the presence of a positive feedback microcircuit between Erk5 and Akt downstream of PI3-kinase nodal point for PDGF-induced mesangial cell proliferation.


Asunto(s)
Proliferación Celular , Retroalimentación Fisiológica/fisiología , Células Mesangiales/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/biosíntesis , Proteína Oncogénica v-akt/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Retroalimentación Fisiológica/efectos de los fármacos , Células Mesangiales/efectos de los fármacos , Ratas
17.
Am J Physiol Renal Physiol ; 306(1): F85-97, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24197068

RESUMEN

The adult kidney is derived from the interaction between the metanephric blastema and the ureteric bud. Platelet-derived growth factor (PDGF) receptor ß is essential for the development of the mature glomerular tuft, as mice deficient for this receptor lack mesangial cells. This study investigated the role of Src tyrosine kinase in PDGF-mediated reactive oxygen species (ROS) generation and migration of metanephric mesenchymal cells (MMCs). Cultured embryonic MMCs from wild-type and PDGF receptor-deficient embryos were established. Migration was determined via wound-healing assay. Unlike PDGF AA, PDGF BB-induced greater migration in MMCs with respect to control. This was abrogated by neutralizing an antibody to PDGF BB. Phosphatidylinositol 3-kinase (PI3K) inhibitors suppressed PDGF BB-induced migration. Conversely, mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) inhibitors had no effect. Src inhibitors inhibited PDGF-induced cell migration, PI3K activity, and Akt phosphorylation. Adenoviral dominant negative Src (AD DN Src) abrogated PDGF BB-induced Akt phosphorylation. Hydrogen peroxide stimulated cell migration. PDGF BB-induced wound closure was inhibited by the antioxidants N-acetyl-l-cysteine, tiron, and the flavoprotein inhibitor diphenyleneiodonium. These cells express the NADPH oxidase homolog Nox4. Inhibiting Nox4 with antisense oligonucleotides or small interfering RNA (siRNA) suppressed PDGF-induced wound closure. Inhibition of Src with siRNA reduced PDGF BB-induced ROS generation as assessed by 2',7'-dichlorodihydrofluorescein diacetate fluorescence. Furthermore, PDGF BB-stimulated ROS generation and migration were similarly suppressed by Ad DN Src. In MMCs, PDGF BB-induced migration is mediated by PI3K and Src in a redox-dependent manner involving Nox4. Src may be upstream to PI3K and Nox4.


Asunto(s)
Células Madre Mesenquimatosas/metabolismo , Proteínas Proto-Oncogénicas c-sis/farmacología , Familia-src Quinasas/metabolismo , Animales , Becaplermina , Movimiento Celular , Células Cultivadas , Cromonas/farmacología , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Morfolinas/farmacología , NADPH Oxidasa 4 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Oxidación-Reducción , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Familia-src Quinasas/genética
18.
J Biol Chem ; 288(40): 28668-86, 2013 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-23940049

RESUMEN

Activation of glomerular mesangial cells (MCs) by angiotensin II (Ang II) leads to extracellular matrix accumulation. Here, we demonstrate that, in MCs, Ang II induces endothelial nitric-oxide synthase (eNOS) uncoupling with enhanced generation of reactive oxygen species (ROS) and decreased production of NO. Ang II promotes a rapid increase in 3-nitrotyrosine formation, and uric acid attenuates Ang II-induced decrease in NO bioavailability, demonstrating that peroxynitrite mediates the effects of Ang II on eNOS dysfunction. Ang II rapidly up-regulates Nox4 protein. Inhibition of Nox4 abolishes the increase in ROS and peroxynitrite generation as well as eNOS uncoupling triggered by Ang II, indicating that Nox4 is upstream of eNOS. This pathway contributes to Ang II-mediated fibronectin accumulation in MCs. Ang II also elicits an increase in mitochondrial abundance of Nox4 protein, and the oxidase contributes to ROS production in mitochondria. Overexpression of mitochondrial manganese superoxide dismutase prevents the stimulatory effects of Ang II on mitochondrial ROS production, loss of NO availability, and MC fibronectin accumulation, whereas manganese superoxide dismutase depletion increases mitochondrial ROS, NO deficiency, and fibronectin synthesis basally and in cells exposed to Ang II. This work provides the first evidence that uncoupled eNOS is responsible for Ang II-induced MC fibronectin accumulation and identifies Nox4 and mitochondrial ROS as mediators of eNOS dysfunction. These data shed light on molecular processes underlying the oxidative signaling cascade engaged by Ang II and identify potential targets for intervention to prevent renal fibrosis.


Asunto(s)
Angiotensina II/farmacología , Fibronectinas/metabolismo , Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ácido Peroxinitroso/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Disponibilidad Biológica , Fibrosis , Silenciador del Gen/efectos de los fármacos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Células Mesangiales/efectos de los fármacos , Células Mesangiales/enzimología , Células Mesangiales/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Modelos Biológicos , NADPH Oxidasa 4 , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Ratas , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Regulación hacia Arriba/efectos de los fármacos
19.
Mol Cell Biol ; 33(17): 3439-60, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23816887

RESUMEN

Mesangial matrix accumulation is an early feature of glomerular pathology in diabetes. Oxidative stress plays a critical role in hyperglycemia-induced glomerular injury. Here, we demonstrate that, in glomerular mesangial cells (MCs), endothelial nitric oxide synthase (eNOS) is uncoupled upon exposure to high glucose (HG), with enhanced generation of reactive oxygen species (ROS) and decreased production of nitric oxide. Peroxynitrite mediates the effects of HG on eNOS dysfunction. HG upregulates Nox4 protein, and inhibition of Nox4 abrogates the increase in ROS and peroxynitrite generation, as well as the eNOS uncoupling triggered by HG, demonstrating that Nox4 functions upstream from eNOS. Importantly, this pathway contributes to HG-induced MC fibronectin accumulation. Nox4-mediated eNOS dysfunction was confirmed in glomeruli of a rat model of type 1 diabetes. Sestrin 2-dependent AMP-activated protein kinase (AMPK) activation attenuates HG-induced MC fibronectin synthesis through blockade of Nox4-dependent ROS and peroxynitrite generation, with subsequent eNOS uncoupling. We also find that HG negatively regulates sestrin 2 and AMPK, thereby promoting Nox4-mediated eNOS dysfunction and increased fibronectin. These data identify a protective function for sestrin 2/AMPK and potential targets for intervention to prevent fibrotic injury in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Hiperglucemia/metabolismo , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas Nucleares/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus Experimental/genética , Fibronectinas/genética , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Mesangio Glomerular/citología , Mesangio Glomerular/metabolismo , Glucosa/metabolismo , Hiperglucemia/genética , Masculino , NADPH Oxidasa 4 , NADPH Oxidasas/genética , Óxido Nítrico/metabolismo , Ácido Peroxinitroso/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Superóxidos/metabolismo
20.
Am J Physiol Renal Physiol ; 305(5): F691-700, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23804455

RESUMEN

Podocyte injury, a major contributor to the pathogenesis of diabetic nephropathy, is caused at least in part by the excessive generation of reactive oxygen species (ROS). Overproduction of superoxide by the NADPH oxidase isoform Nox4 plays an important role in podocyte injury. The plant extract silymarin is attributed antioxidant and antiproteinuric effects in humans and in animal models of diabetic nephropathy. We investigated the effect of silybin, the active constituent of silymarin, in cultures of mouse podocytes and in the OVE26 mouse, a model of type 1 diabetes mellitus and diabetic nephropathy. Exposure of podocytes to high glucose (HG) increased 60% the intracellular superoxide production, 90% the NADPH oxidase activity, 100% the Nox4 expression, and 150% the number of apoptotic cells, effects that were completely blocked by 10 µM silybin. These in vitro observations were confirmed by similar in vivo findings. The kidney cortex of vehicle-treated control OVE26 mice displayed greater Nox4 expression and twice as much superoxide production than cortex of silybin-treated mice. The glomeruli of control OVE26 mice displayed 35% podocyte drop out that was not present in the silybin-treated mice. Finally, the OVE26 mice experienced 54% more pronounced albuminuria than the silybin-treated animals. In conclusion, this study demonstrates a protective effect of silybin against HG-induced podocyte injury and extends this finding to an animal model of diabetic nephropathy.


Asunto(s)
Antioxidantes/uso terapéutico , Nefropatías Diabéticas/prevención & control , Glucosa/farmacología , Estrés Oxidativo/efectos de los fármacos , Podocitos/efectos de los fármacos , Silimarina/uso terapéutico , Animales , Diabetes Mellitus Tipo 1/fisiopatología , Nefropatías Diabéticas/metabolismo , Modelos Animales de Enfermedad , Ratones , NADPH Oxidasa 4 , NADPH Oxidasas/biosíntesis , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Silibina , Superóxidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA