Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Diabetes ; 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38656940

RESUMEN

Reduced kidney AMPK activity is associated with nutrient stress-induced chronic kidney disease (CKD) in male mice. In contrast, female mice resist nutrient stress-induced CKD. The role of kidney AMPK in sex-related organ protection against nutrient stress and metabolite changes were evaluated in diabetic kidney tubule-specific AMPKγ2KO (KTAMPKγ2KO) male and female mice. In WT males, diabetes increased albuminuria, urinary kidney injury molecule-1, hypertension, kidney p70S6K phosphorylation, and kidney matrix accumulation; these features were not exacerbated with KTAMPKγ2KO. Whereas WT females had protection against diabetes induced kidney injury, KTAMPKγ2KO led to loss of female protection against kidney disease. 17ß-estradiol ameliorated high glucose-induced AMPK inactivation, p70S6K phosphorylation and matrix protein accumulation in kidney tubule cells. The mechanism for female protection against diabetes-induced kidney injury is likely via an estrogen-AMPK pathway, as inhibition of AMPK led to loss of estrogen protection to glucose-induced mTORC1 activation and matrix production. RNA-seq and metabolomic analysis identified a decrease in the degradation pathway of phenylalanine and tyrosine resulting in increased urinary phenylalanine and tyrosine levels in females. The metabolite levels correlated with loss of female protection. The findings provide new insights to explain evolutionary advantages to females during states of nutrient challenges.

3.
FEBS Lett ; 597(9): 1300-1316, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36775967

RESUMEN

The plasticity of proximal tubular epithelial cells in response to TGFß contributes to the expression of TWIST1 to drive renal fibrosis. The mechanism of TWIST1 expression is not known. We show that both PI3 kinase and its target mTORC2 increase TGFß-induced TWIST1 expression. TGFß enhances phosphorylation on Ser-660 in the protein kinase C ßII (PKCßII) hydrophobic motif site. Remarkably, phosphorylation-deficient PKCßIIS660A, kinase-dead PKCßII, and PKCßII knockdown blocked TWIST1 expression by TGFß. Inhibition of TWIST1 arrested TGFß-induced tubular cell hypertrophy and the expression of fibronectin, collagen I (α2), and α-smooth muscle actin. By contrast, TWIST1 overexpression induced these pathologies. Interestingly, the inhibition of PKCßII reduced these phenomena, which were countered by the expression of TWIST1. These results provide the first evidence for the involvement of the mTORC2-PKCßII axis in TWIST1 expression to promote tubular cell pathology.


Asunto(s)
Serina-Treonina Quinasas TOR , Factor de Crecimiento Transformador beta , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal , Proteína Quinasa C beta , Células Epiteliales/metabolismo
4.
J Biol Chem ; 298(9): 102246, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35835217

RESUMEN

Proximal tubular epithelial cells respond to transforming growth factor ß (TGFß) to synthesize collagen I (α2) during renal fibrosis. The oncoprotein DJ-1 has previously been shown to promote tumorigenesis and prevent apoptosis of dopaminergic neurons; however, its role in fibrosis signaling is unclear. Here, we show TGFß-stimulation increased expression of DJ-1, which promoted noncanonical mTORC1 and mTORC2 activities. We show DJ-1 augmented the phosphorylation/activation of PKCßII, a direct substrate of mTORC2. In addition, coimmunoprecipitation experiments revealed association of DJ-1 with Raptor and Rictor, exclusive subunits of mTORC1 and mTORC2, respectively, as well as with mTOR kinase. Interestingly, siRNAs against DJ-1 blocked TGFß-stimulated expression of collagen I (α2), while expression of DJ-1 increased expression of this protein. In addition, expression of dominant negative PKCßII and siRNAs against PKCßII significantly inhibited TGFß-induced collagen I (α2) expression. In fact, constitutively active PKCßII abrogated the effect of siRNAs against DJ-1, suggesting a role of PKCßII downstream of this oncoprotein. Moreover, we demonstrate expression of collagen I (α2) stimulated by DJ-1 and its target PKCßII is dependent on the transcription factor hypoxia-inducible factor 1α (Hif1α). Finally, we show in the renal cortex of diabetic rats that increased TGFß was associated with enhanced expression of DJ-1 and activation of mTOR and PKCßII, concomitant with increased Hif1α and collagen I (α2). Overall, we identified that DJ-1 affects TGFß-induced expression of collagen I (α2) via an mTOR-, PKCßII-, and Hif1α-dependent mechanism to regulate renal fibrosis.


Asunto(s)
Colágeno Tipo I , Diabetes Mellitus Experimental , Nefropatías Diabéticas , Subunidad alfa del Factor 1 Inducible por Hipoxia , Riñón , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Proteínas Oncogénicas , Proteína Desglicasa DJ-1 , Animales , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/genética , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Fibrosis , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Riñón/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/metabolismo , Proteína Quinasa C beta/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
6.
Cell Signal ; 86: 110072, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34224844

RESUMEN

Function of mTORC1 and mTORC2 has emerged as a driver of mesangial cell pathologies in diabetic nephropathy. The mechanism of mTOR activation is poorly understood in this disease. Deptor is a constitutive subunit and a negative regulator of both mTOR complexes. Mechanistic investigation in mesangial cells revealed that high glucose decreased the expression of deptor concomitant with increased mTORC1 and mTORC2 activities, induction of hypertrophy and, expression of fibronectin and PAI-1. shRNAs against deptor mimicked these pathologic outcomes of high glucose. Conversely, overexpression of deptor significantly inhibited all effects of high glucose. To determine the mechanism of deptor suppression, we found that high glucose significantly increased the expression of EZH2, resulting in lysine-27 tri-methylation of histone H3 (H3K27Me3). Employing approaches including pharmacological inhibition, shRNA-mediated downregulation and overexpression of EZH2, we found that EZH2 regulates high glucose-induced deptor suppression along with activation of mTOR, mesangial cell hypertrophy and fibronectin/PAI-1 expression. Moreover, expression of hyperactive mTORC1 reversed shEZH2-mediated inhibition of hypertrophy and expression of fibronectin and PAI-1 by high glucose. Finally, in renal cortex of diabetic mice, we found that enhanced expression of EZH2 is associated with decreased deptor levels and increased mTOR activity and, expression of fibronectin and PAI-1. Together, our findings provide a novel mechanism for mTOR activation via EZH2 to induce mesangial cell hypertrophy and matrix expansion during early progression of diabetic nephropathy. These results suggest a strategy for leveraging the intrinsic effect of deptor to suppress mTOR activity via reducing EZH2 as a novel therapy for diabetic nephropathy.


Asunto(s)
Diabetes Mellitus Experimental , Células Mesangiales , Animales , Diabetes Mellitus Experimental/patología , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Glucosa/metabolismo , Glucosa/farmacología , Péptidos y Proteínas de Señalización Intracelular , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Células Mesangiales/metabolismo , Ratones
7.
Adv Exp Med Biol ; 1315: 17-50, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34302687

RESUMEN

Hydrogen sulfide (H2S) is constitutively synthesized in the kidney. Recent investigations suggest a role for H2S in the regulation of fundamental kidney physiological events including arterial blood flow, glomerular filtration, and electrolyte and water transport. Deficiency of H2S generation has been implicated in acute kidney injury brought on by ischemia, administration of nephrotoxic medications, and obstruction. A role for impaired H2S expression has been shown in chronic kidney injury seen with chronic heart failure, obesity, and diabetes. Deficient H2S generation by the kidney could contribute to blood pressure dysregulation in models of hypertension and preeclampsia. Aging induced chronic kidney impairment is associated with inadequate H2S generation in the kidney. The mechanistic pathways regulated by H2S include but not limited to transcription, mRNA translation, signaling, inflammation, and oxidative stress demonstrating the versatility of the gasotransmitter. In the aforementioned conditions amelioration of kidney injury has been reported by the administration of agents that provide H2S. In renal cancer H2S may participate as an injurious agent. Overall, research on H2S in the kidney is in its early stages, and it is becoming evident that it has a context-dependent nuanced role in various kidney pathologies. There is an urgent need for exploration of H2S in physiology and pathology of the kidney including its role in oxygen sensing and glomerulonephritis. H2S may prove to be a novel therapeutic agent in some kidney disease states.


Asunto(s)
Lesión Renal Aguda , Gasotransmisores , Sulfuro de Hidrógeno , Lesión Renal Aguda/metabolismo , Gasotransmisores/metabolismo , Humanos , Sulfuro de Hidrógeno/metabolismo , Riñón/metabolismo , Estrés Oxidativo
8.
Aging Cell ; 20(7): e13407, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34118180

RESUMEN

The mechanism of kidney injury in aging are not well understood. In order to identify hitherto unknown pathways of aging-related kidney injury, we performed RNA-Seq on kidney extracts of young and aged mice. Expression of chloride (Cl) channel accessory 1 (CLCA1) mRNA and protein was increased in the kidneys of aged mice. Immunostaining showed a marked increase in CLCLA1 expression in the proximal tubules of the kidney from aged mice. Increased kidney CLCA1 gene expression also correlated with aging in marmosets and in a human cohort. In aging mice, increased renal cortical CLCA1 content was associated with hydrogen sulfide (H2 S) deficiency, which was ameliorated by administering sodium hydrosulfide (NaHS), a source of H2 S. In order to study whether increased CLCA1 expression leads to injury phenotype and the mechanisms involved, stable transfection of proximal tubule epithelial cells overexpressing human CLCA1 (hCLCA1) was performed. Overexpression of hCLCA1 augmented Cl- current via the Ca++ -dependent Cl- channel TMEM16A (anoctamin-1) by patch-clamp studies. hCLCA1 overexpression also increased the expression of fibronectin, a matrix protein, and induced the senescence-associated secretory phenotype (SASP). Mechanistic studies underlying these changes showed that hCLCA1 overexpression leads to inhibition of AMPK activity and stimulation of mTORC1 as cellular signaling determinants of injury. Both TMEM16A inhibitor and NaHS reversed these signaling events and prevented changes in fibronectin and SASP. We conclude that CLCA1-TMEM16A-Cl- current pathway is a novel mediator of kidney injury in aging that is regulated by endogenous H2 S.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Canales de Cloruro/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Factores de Edad , Animales , Callithrix , Humanos , Ratones , Ratones Endogámicos C57BL
9.
BMC Nephrol ; 22(1): 112, 2021 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-33773596

RESUMEN

BACKGROUND: Fatigue is prevalent in hemodialysis patients who for survival follow a strict dialysis treatment regimen - dialysis and non-dialysis days. As a result, the daily activities, symptom burden, and clinical outcomes of hemodialysis patients vary significantly between dialysis and non-dialysis days. Fatigue is one of the most reported debilitating symptoms by hemodialysis patients with profound negative impact on their quality of life. Prior studies assessed fatigue during the preceding 7 or 30 days and did not discriminate fatigue characteristics between dialysis and non-dialysis days. We aimed to characterize and compare fatigue severity and fatigue interference with daily activities between dialysis and non-dialysis days. METHODS: Hemodialysis patients self-reported fatigue on consecutive dialysis and non-dialysis days using the 9-item Brief Fatigue Inventory. The differences in fatigue characteristics between dialysis and non-dialysis days were analyzed using one-way ANCOVA. RESULTS: Global fatigue burden was worse on a dialysis day compared to a non-dialysis day (P for all < 0.001). Age and education were associated with fatigue, but hemodialysis-related variables were not. A significant inverse association of physical activity with fatigue severity observed on non-dialysis day; there was also a negative association between the normalized protein catabolic rate and fatigue severity on both dialysis and non-dialysis days. The positive association of depression with fatigue severity and fatigue interference were consistent on both dialysis and non-dialysis days. None of these factors, however, explained differences in fatigue characteristics between dialysis and non-dialysis days. CONCLUSIONS: Fatigue, measured in severity and interference, was more pronounced on a dialysis day relative to a non-dialysis day. These differences were not explained by age, sex, education, hemodialysis-related variables, habitual exercise, nutritional status, and or depression. The quantitative measures of fatigue characteristics may facilitate future interventional trials design and better fatigue management for hemodialysis patients.


Asunto(s)
Fatiga/etiología , Diálisis Renal/efectos adversos , Actividades Cotidianas , Adulto , Análisis de Varianza , Ejercicio Físico , Femenino , Humanos , Fallo Renal Crónico/terapia , Masculino , Persona de Mediana Edad , Calidad de Vida , Autoinforme , Índice de Severidad de la Enfermedad
11.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33400689

RESUMEN

The role of insulin receptor (IR) activated by hyperinsulinemia in obesity-induced kidney injury is not well understood. We hypothesized that activation of kidney proximal tubule epithelial IR contributes to obesity-induced kidney injury. We administered normal-fat diet (NFD) or high-fat diet (HFD) to control and kidney proximal tubule IR-knockout (KPTIRKO) mice for 4 months. Renal cortical IR expression was decreased by 60% in male and female KPTIRKO mice. Baseline serum glucose, serum creatinine, and the ratio of urinary albumin to creatinine (ACR) were similar in KPTIRKO mice compared to those of controls. On HFD, weight gain and increase in serum cholesterol were similar in control and KPTIRKO mice; blood glucose did not change. HFD increased the following parameters in the male control mice: renal cortical contents of phosphorylated IR and Akt, matrix proteins, urinary ACR, urinary kidney injury molecule-1-to-creatinine ratio, and systolic blood pressure. Renal cortical generation of hydrogen sulfide was reduced in HFD-fed male control mice. All of these parameters were ameliorated in male KPTIRKO mice. Interestingly, female mice were resistant to HFD-induced kidney injury in both genotypes. We conclude that HFD-induced kidney injury requires renal proximal tubule IR activation in male mice.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Túbulos Renales Proximales/metabolismo , Receptor de Insulina/metabolismo , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Animales , Epitelio/metabolismo , Femenino , Sulfuro de Hidrógeno/metabolismo , Resistencia a la Insulina , Corteza Renal/metabolismo , Masculino , Ratones , Ratones Noqueados , Obesidad/complicaciones , Obesidad/metabolismo , Receptor de Insulina/deficiencia , Receptor de Insulina/genética , Factores Sexuales , Transducción de Señal
12.
J Biol Chem ; 295(42): 14262-14278, 2020 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-32732288

RESUMEN

Interaction of transforming growth factor-ß (TGFß)-induced canonical signaling with the noncanonical kinase cascades regulates glomerular hypertrophy and matrix protein deposition, which are early features of glomerulosclerosis. However, the specific target downstream of the TGFß receptor involved in the noncanonical signaling is unknown. Here, we show that TGFß increased the catalytic loop phosphorylation of platelet-derived growth factor receptor ß (PDGFRß), a receptor tyrosine kinase expressed abundantly in glomerular mesangial cells. TGFß increased phosphorylation of the PI 3-kinase-interacting Tyr-751 residue of PDGFRß, thus activating Akt. Inhibition of PDGFRß using a pharmacological inhibitor and siRNAs blocked TGFß-stimulated phosphorylation of proline-rich Akt substrate of 40 kDa (PRAS40), an intrinsic inhibitory component of mTORC1, and prevented activation of mTORC1 in the absence of any effect on Smad 2/3 phosphorylation. Expression of constitutively active myristoylated Akt reversed the siPDGFRß-mediated inhibition of mTORC1 activity; however, co-expression of the phospho-deficient mutant of PRAS40 inhibited the effect of myristoylated Akt, suggesting a definitive role of PRAS40 phosphorylation in mTORC1 activation downstream of PDGFRß in mesangial cells. Additionally, we demonstrate that PDGFRß-initiated phosphorylation of PRAS40 is required for TGFß-induced mesangial cell hypertrophy and fibronectin and collagen I (α2) production. Increased activating phosphorylation of PDGFRß is also associated with enhanced TGFß expression and mTORC1 activation in the kidney cortex and glomeruli of diabetic mice and rats, respectively. Thus, pursuing TGFß noncanonical signaling, we identified how TGFß receptor I achieves mTORC1 activation through PDGFRß-mediated Akt/PRAS40 phosphorylation to spur mesangial cell hypertrophy and matrix protein accumulation. These findings provide support for targeting PDGFRß in TGFß-driven renal fibrosis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Fibronectinas/metabolismo , Humanos , Corteza Renal/metabolismo , Células Mesangiales/citología , Células Mesangiales/metabolismo , Ratones , Ratones Transgénicos , Mutagénesis Sitio-Dirigida , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
13.
Am J Nephrol ; 51(7): 565-571, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32575099

RESUMEN

BACKGROUND: Fatigue is one of the most debilitating symptoms reported by maintenance hemodialysis (MHD) patients. Hemodialysis causes marked depletion in plasma essential amino acids. We studied the cross-sectional relationship of pre- and post-hemodialysis branched-chain amino acids (BCAAs) concentrations with fatigue in MHD patients. METHODS: MHD patients self-reported fatigue during a dialysis session using the Brief Fatigue Inventory. Pre- and post-dialysis plasma levels of BCAAs (valine, leucine, and isoleucine) were measured using HPLC-mass spectrometry. RESULTS: The mean age of study participants (n = 114) was 54.8 ± 12.8 years. Plasma levels of BCAAs decreased significantly post-dialysis compared to pre-dialysis (303.8 ± 9.4 vs. 392.1 ± 9.4 µM/L, p < 0.0001). Fatigue score increased as a function of age (p = 0.015). There was no association between pre-dialysis plasma levels of BCAAs and fatigue. A significant negative correlation was found between post-dialysis plasma levels of BCAAs and fatigue (p < 0.05). CONCLUSIONS: These preliminary findings suggest that disruption in BCAAs homeostasis may play a role in precipitating fatigue.


Asunto(s)
Aminoácidos de Cadena Ramificada/sangre , Fatiga/epidemiología , Fallo Renal Crónico/terapia , Diálisis Renal/efectos adversos , Adulto , Anciano , Aminoácidos de Cadena Ramificada/metabolismo , Estudios de Cohortes , Estudios Transversales , Fatiga/sangre , Fatiga/diagnóstico , Fatiga/etiología , Femenino , Humanos , Fallo Renal Crónico/sangre , Masculino , Persona de Mediana Edad , Autoinforme/estadística & datos numéricos
14.
Nat Commun ; 10(1): 4720, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31624249

RESUMEN

Understanding the molecular components of insulin signaling is relevant to effectively manage insulin resistance. We investigated the phenotype of the TMEM127 tumor suppressor gene deficiency in vivo. Whole-body Tmem127 knockout mice have decreased adiposity and maintain insulin sensitivity, low hepatic fat deposition and peripheral glucose clearance after a high-fat diet. Liver-specific and adipose-specific Tmem127 deletion partially overlap global Tmem127 loss: liver Tmem127 promotes hepatic gluconeogenesis and inhibits peripheral glucose uptake, while adipose Tmem127 downregulates adipogenesis and hepatic glucose production. mTORC2 is activated in TMEM127-deficient hepatocytes suggesting that it interacts with TMEM127 to control insulin sensitivity. Murine hepatic Tmem127 expression is increased in insulin-resistant states and is reversed by diet or the insulin sensitizer pioglitazone. Importantly, human liver TMEM127 expression correlates with steatohepatitis and insulin resistance. Our results suggest that besides tumor suppression activities, TMEM127 is a nutrient-sensing component of glucose/lipid homeostasis and may be a target in insulin resistance.


Asunto(s)
Tejido Adiposo/metabolismo , Genes Supresores de Tumor , Resistencia a la Insulina/genética , Hígado/metabolismo , Proteínas de la Membrana/genética , Adipogénesis/genética , Animales , Dieta Alta en Grasa , Perfilación de la Expresión Génica/métodos , Gluconeogénesis/genética , Humanos , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Especificidad de Órganos/genética
15.
FEBS Lett ; 593(16): 2261-2272, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31240704

RESUMEN

The mechanism of PTEN repression by high glucose in diabetic nephropathy is not known. Using proximal tubular cells, we show that inhibition of PI3 kinase/Akt and their inactive enzymes prevents high glucose-induced PTEN downregulation. Similarly, rapamycin (Rapa) and shRaptor block suppression of PTEN by high glucose. In contrast, the constitutive activation of Akt and mechanistic target of rapamycin (mTOR)C1 decrease the expression of PTEN, similarly to high glucose. Remarkably, PI3 kinase/Akt/mTORC1 inhibition significantly attenuates high glucose-stimulated increase in miR-214, which targets PTEN, while constitutively active Akt/mTORC1 increases miR-214. Furthermore, anti-miR-214 and mTORC1 inhibition block high glucose-induced hypertrophy and fibronectin expression. These results reveal the first evidence for the presence of a high glucose-forced positive feedback conduit between the three-layered kinase cascade and miR-214/ PTEN in tubular cell injury.


Asunto(s)
Glucosa/farmacología , Túbulos Renales Proximales/efectos de los fármacos , MicroARNs/genética , Fosfohidrolasa PTEN/genética , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular , Retroalimentación Fisiológica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo
16.
J Biol Chem ; 294(24): 9440-9460, 2019 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-31028173

RESUMEN

S6 kinase acts as a driver for renal hypertrophy and matrix accumulation, two key pathologic signatures of diabetic nephropathy. As a post-translational modification, S6 kinase undergoes acetylation at the C terminus. The role of this acetylation to regulate kidney glomerular cell hypertrophy and matrix expansion is not known. In mesangial cells, high glucose decreased the acetylation and enhanced phosphorylation of S6 kinase and its substrates rps6 and eEF2 kinase that lead to dephosphorylation of eEF2. To determine the mechanism of S6 kinase deacetylation, we found that trichostatin A, a pan-histone deacetylase (HDAC) inhibitor, blocked all high glucose-induced effects. Furthermore, high glucose increased the expression and association of HDAC1 with S6 kinase. HDAC1 decreased the acetylation of S6 kinase and mimicked the effects of high glucose, resulting in mesangial cell hypertrophy and expression of fibronectin and collagen I (α2). In contrast, siRNA against HDAC1 inhibited these effects by high glucose. A C-terminal acetylation-mimetic mutant of S6 kinase suppressed high glucose-stimulated phosphorylation of S6 kinase, rps6 and eEF2 kinase, and inhibited the dephosphorylation of eEF2. Also, the acetylation mimetic attenuated the mesangial cell hypertrophy and fibronectin and collagen I (α2) expression. Conversely, an S6 kinase acetylation-deficient mutant induced all the above effects of high glucose. Finally, in the renal glomeruli of diabetic rats, the acetylation of S6 kinase was significantly reduced concomitant with increased HDAC1 and S6 kinase activity. In aggregate, our data uncovered a previously unrecognized role of S6 kinase deacetylation in high glucose-induced mesangial cell hypertrophy and matrix protein expression.


Asunto(s)
Diabetes Mellitus Experimental/patología , Fibronectinas/metabolismo , Glucosa/farmacología , Hipertrofia/patología , Glomérulos Renales/patología , Células Mesangiales/patología , Proteínas Quinasas S6 Ribosómicas/metabolismo , Acetilación , Animales , Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Humanos , Hipertrofia/etiología , Hipertrofia/metabolismo , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/metabolismo , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas S6 Ribosómicas/genética , Transducción de Señal , Edulcorantes/farmacología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
17.
J Gerontol A Biol Sci Med Sci ; 74(3): 315-324, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30321310

RESUMEN

We evaluated whether the marmoset, a nonhuman primate, can serve as a good model to study aging-related changes in the kidney by employing healthy young and aged marmosets of both sexes. Aging was associated with glomerulosclerosis, interstitial fibrosis, and arteriolosclerosis in both sexes; correspondingly, the content of matrix proteins was increased. Functionally, aging resulted in an increase in urinary albumin and protein excretion. There was a robust correlation between markers of fibrosis and functional changes. We explored signaling pathways as potential mechanistic events. Aging in males, but not in females, was associated with reduced renal cortical activity of AMP-activated protein kinase (AMPK) and a trend toward activation of mechanistic target of rapamycin complex 1 (mTORC1); upstream of AMPK and mTORC1, Akt and IGF-1 receptor were activated. In both sexes, aging promoted kidney activation of transforming growth factor ß-1 signaling pathway. While the expression of cystathionine ß-synthase (CBS), an enzyme involved hydrogen sulfide (H2S) synthesis, was reduced in both aged males and females, decreased H2S generation was seen in only males. Our studies show that the marmoset is a valid model to study kidney aging; some of the signaling pathways involved in renal senescence differ between male and female marmosets.


Asunto(s)
Envejecimiento/fisiología , Callithrix , Modelos Animales de Enfermedad , Riñón/metabolismo , Riñón/patología , Factores de Edad , Animales , Femenino , Riñón/fisiopatología , Masculino , Factores Sexuales , Transducción de Señal
18.
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
19.
Geroscience ; 40(2): 163-176, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29717417

RESUMEN

Aging is associated with replacement of normal kidney parenchyma by fibrosis. Because hydrogen sulfide (H2S) ameliorates kidney fibrosis in disease models, we examined its status in the aging kidney. In the first study, we examined kidney cortical H2S metabolism and signaling pathways related to synthesis of proteins including matrix proteins in young and old male C57BL/6 mice. In old mice, increase in renal cortical content of matrix protein involved in fibrosis was associated with decreased H2S generation and AMPK activity, and activation of insulin receptor (IR)/IRS-2-Akt-mTORC1-mRNA translation signaling axis that can lead to increase in protein synthesis. In the second study, we randomized 18-19 month-old male C57BL/6 mice to receive 30 µmol/L sodium hydrosulfide (NaHS) in drinking water vs. water alone (control) for 5 months. Administration of NaHS increased plasma free sulfide levels. NaHS inhibited the increase in kidney cortical content of matrix proteins involved in fibrosis and ameliorated glomerulosclerosis. NaHS restored AMPK activity and inhibited activation of IR/IRS-2-Akt-mTORC1-mRNA translation axis. NaHS inhibited age-related increase in kidney cortical content of p21, IL-1ß, and IL-6, components of the senescence-associated secretory phenotype. NaHS abolished increase in urinary albumin excretion seen in control mice and reduced serum cystatin C levels suggesting improved glomerular clearance function. We conclude that aging-induced changes in the kidney are associated with H2S deficiency. Administration of H2S ameliorates aging-induced kidney changes probably by inhibiting signaling pathways leading to matrix protein synthesis.


Asunto(s)
Envejecimiento/efectos de los fármacos , Sulfuro de Hidrógeno/metabolismo , Sulfuro de Hidrógeno/farmacología , Riñón/patología , Lesión Renal Aguda/patología , Lesión Renal Aguda/prevención & control , Envejecimiento/metabolismo , Animales , Biomarcadores/metabolismo , Biopsia con Aguja , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Fibrosis/tratamiento farmacológico , Fibrosis/patología , Inmunohistoquímica , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Valores de Referencia , Factores de Riesgo , Transducción de Señal/efectos de los fármacos
20.
Exp Cell Res ; 364(1): 5-15, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29397070

RESUMEN

TGFß contributes to mesangial cell hypertrophy and matrix protein increase in various kidney diseases including diabetic nephropathy. Deptor is an mTOR-interacting protein and suppresses mTORC1 and mTORC2 activities. We have recently shown that TGFß-induced inhibition of deptor increases the mTOR activity. The mechanism by which TGFß regulates deptor expression is not known. Here we identify deptor as a target of the microRNA-181a. We show that in mesangial cells, TGFß increases the expression of miR-181a to downregulate deptor. Decrease in deptor augments mTORC2 activity, resulting in phosphorylation/activation of Akt kinase. Akt promotes inactivating phosphorylation of PRAS40 and tuberin, leading to stimulation of mTORC1. miR-181a-mimic increased mTORC1 and C2 activities, while anti-miR-181a inhibited them. mTORC1 controls protein synthesis via phosphorylation of translation initiation and elongation suppressors 4EBP-1 and eEF2 kinase. TGFß-stimulated miR-181a increased the phosphorylation of 4EBP-1 and eEF2 kinase, resulting in their inactivation. miR-181a-dependent inactivation of eEF2 kinase caused dephosphorylation of eEF2. Consequently, miR-181a-mimic increased protein synthesis and hypertrophy of mesangial cells similar to TGFß. Anti-miR-181a blocked these events in a deptor-dependent manner. Finally, TGFß-miR-181a-driven deptor downregulation increased the expression of fibronectin. Our results identify a novel mechanism involving miR-181a-driven deptor downregulation, which contributes to mesangial cell pathologies in renal complications.


Asunto(s)
Fibronectinas/metabolismo , Regulación de la Expresión Génica , Hipertrofia/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Glomérulos Renales/patología , Células Mesangiales/patología , MicroARNs/genética , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Células Cultivadas , Regulación hacia Abajo , Hipertrofia/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Glomérulos Renales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/genética , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Células Mesangiales/metabolismo , Fosforilación , Ratas , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factor de Crecimiento Transformador beta1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...