Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
FEBS Lett ; 597(9): 1300-1316, 2023 05.
Article in English | MEDLINE | ID: mdl-36775967

ABSTRACT

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.


Subject(s)
TOR Serine-Threonine Kinases , Transforming Growth Factor beta , Mechanistic Target of Rapamycin Complex 2/metabolism , Transforming Growth Factor beta/metabolism , TOR Serine-Threonine Kinases/metabolism , Signal Transduction , Protein Kinase C beta , Epithelial Cells/metabolism
2.
Am J Physiol Endocrinol Metab ; 321(1): E90-E104, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34029162

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a spectrum of disorders ranging from hepatic steatosis [excessive accumulation of triglycerides (TG)] to nonalcoholic steatohepatitis, which can progress to cirrhosis and hepatocellular carcinoma. The molecular pathogenesis of steatosis and progression to more severe NAFLD remains unclear. Obesity and aging, two principal risk factors for NAFLD, are associated with a hyperadrenergic state. ß-Adrenergic responsiveness in liver increases in animal models of obesity and aging, and in both is linked to increased hepatic expression of ß2-adrenergic receptors (ß2-ARs). We previously showed that in aging rodents intracellular signaling from elevated hepatic levels of ß2-ARs may contribute to liver steatosis. In this study we demonstrate that injection of formoterol, a highly selective ß2-AR agonist, to mice acutely results in hepatic TG accumulation. Further, we have sought to define the intrahepatic mechanisms underlying ß2-AR mediated steatosis by investigating changes in hepatic expression and cellular localization of enzymes, transcription factors, and coactivators involved in processes of lipid accrual and disposition-and also functional aspects thereof-in livers of formoterol-treated animals. Our results suggest that ß2-AR activation by formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, increased but incomplete ß-oxidation of fatty acids with accumulation of potentially toxic long-chain acylcarnitine intermediates, and reduced TG secretion-all previously invoked as contributors to fatty liver disease. Experiments are ongoing to determine whether sustained activation of hepatic ß2-AR signaling by formoterol might be utilized to model fatty liver changes occurring in hyperadrenergic states of obesity and aging, and thereby identify novel molecular targets for the prevention or treatment of NAFLD.NEW & NOTEWORTHY Results of our study suggest that ß2-adrenergic receptor (ß2-AR) activation by agonist formoterol leads to increased hepatic TG synthesis and de novo lipogenesis, incomplete ß-oxidation of fatty acids with accumulation of long-chain acylcarnitine intermediates, and reduced TG secretion. These findings may, for the first time, implicate a role for ß2-AR responsive dysregulation of hepatic lipid metabolism in the pathogenetic processes underlying NAFLD in hyperadrenergic states such as obesity and aging.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Fatty Liver/chemically induced , Non-alcoholic Fatty Liver Disease/physiopathology , Receptors, Adrenergic, beta-2/physiology , Animals , Carnitine/analogs & derivatives , Carnitine/analysis , Formoterol Fumarate/pharmacology , Gene Expression/drug effects , Hepatic Stellate Cells , Lipid Metabolism/drug effects , Lipid Metabolism/physiology , Lipogenesis/genetics , Liver/chemistry , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/chemically induced , Phosphatidate Phosphatase/analysis , Triglycerides/biosynthesis
3.
JCI Insight ; 6(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33400689

ABSTRACT

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.


Subject(s)
Diet, High-Fat/adverse effects , Kidney Tubules, Proximal/metabolism , Receptor, Insulin/metabolism , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Animals , Epithelium/metabolism , Female , Hydrogen Sulfide/metabolism , Insulin Resistance , Kidney Cortex/metabolism , Male , Mice , Mice, Knockout , Obesity/complications , Obesity/metabolism , Receptor, Insulin/deficiency , Receptor, Insulin/genetics , Sex Factors , Signal Transduction
4.
J Biol Chem ; 295(42): 14262-14278, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32732288

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Fibronectins/metabolism , Humans , Kidney Cortex/metabolism , Mesangial Cells/cytology , Mesangial Cells/metabolism , Mice , Mice, Transgenic , Mutagenesis, Site-Directed , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
5.
FEBS Lett ; 593(16): 2261-2272, 2019 08.
Article in English | MEDLINE | ID: mdl-31240704

ABSTRACT

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.


Subject(s)
Glucose/pharmacology , Kidney Tubules, Proximal/drug effects , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Signal Transduction/drug effects , Animals , Cell Line , Feedback, Physiological/drug effects , Gene Expression Regulation/drug effects , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Proto-Oncogene Proteins c-akt/metabolism
6.
J Biol Chem ; 294(24): 9440-9460, 2019 06 14.
Article in English | MEDLINE | ID: mdl-31028173

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Fibronectins/metabolism , Glucose/pharmacology , Hypertrophy/pathology , Kidney Glomerulus/pathology , Mesangial Cells/pathology , Ribosomal Protein S6 Kinases/metabolism , Acetylation , Animals , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Hypertrophy/etiology , Hypertrophy/metabolism , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases/genetics , Signal Transduction , Sweetening Agents/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
7.
Am J Physiol Cell Physiol ; 313(4): C430-C447, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28701356

ABSTRACT

Aberrant expression of microRNAs (miRs) contributes to diabetic renal complications, including renal hypertrophy and matrix protein accumulation. Reduced expression of phosphatase and tensin homolog (PTEN) by hyperglycemia contributes to these processes. We considered involvement of miR in the downregulation of PTEN. In the renal cortex of type 1 diabetic mice, we detected increased expression of miR-214 in association with decreased levels of PTEN and enhanced Akt phosphorylation and fibronectin expression. Mesangial and proximal tubular epithelial cells exposed to high glucose showed augmented expression of miR-214. Mutagenesis studies using 3'-UTR of PTEN in a reporter construct revealed PTEN as a direct target of miR-214, which controls its expression in both of these cells. Overexpression of miR-214 decreased the levels of PTEN and increased Akt activity similar to high glucose and lead to phosphorylation of its substrates glycogen synthase kinase-3ß, PRAS40, and tuberin. In contrast, quenching of miR-214 inhibited high-glucose-induced Akt activation and its substrate phosphorylation; these changes were reversed by small interfering RNAs against PTEN. Importantly, respective expression of miR-214 or anti-miR-214 increased or decreased the mammalian target of rapamycin complex 1 (mTORC1) activity induced by high glucose. Furthermore, mTORC1 activity was controlled by miR-214-targeted PTEN via Akt activation. In addition, neutralization of high-glucose-stimulated miR-214 expression significantly inhibited cell hypertrophy and expression of the matrix protein fibronectin. Finally, the anti-miR-214-induced inhibition of these processes was reversed by the expression of constitutively active Akt kinase and hyperactive mTORC1. These results uncover a significant role of miR-214 in the activation of mTORC1 that contributes to high-glucose-induced mesangial and proximal tubular cell hypertrophy and fibronectin expression.


Subject(s)
Blood Glucose/metabolism , Cell Proliferation , Diabetes Mellitus, Type 1/enzymology , Diabetic Nephropathies/enzymology , Epithelial Cells/enzymology , Kidney Glomerulus/enzymology , Kidney Tubules, Proximal/enzymology , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , 3' Untranslated Regions , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetic Nephropathies/blood , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Models, Animal , Epithelial Cells/pathology , Fibronectins/metabolism , Gene Expression Regulation, Enzymologic , Glomerular Mesangium/metabolism , Glomerular Mesangium/pathology , Hypertrophy , Kidney Glomerulus/pathology , Kidney Tubules, Proximal/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , MicroRNAs/genetics , Multiprotein Complexes/metabolism , PTEN Phosphohydrolase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Rats , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transfection , Transforming Growth Factor beta/metabolism
8.
Am J Physiol Renal Physiol ; 313(2): F291-F307, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28424212

ABSTRACT

Increased expression of PDGF receptor-ß (PDGFRß) has been shown in renal proximal tubules in mice with diabetes. The core molecular network used by high glucose to induce proximal tubular epithelial cell collagen I (α2) expression is poorly understood. We hypothesized that activation of PDGFRß by high glucose increases collagen I (α2) production via the Akt/mTORC1 signaling pathway in proximal tubular epithelial cells. Using biochemical and molecular biological techniques, we investigated this hypothesis. We show that high glucose increases activating phosphorylation of the PDGFRß, resulting in phosphorylation of phosphatidylinositol 3-kinase. A specific inhibitor, JNJ-10198409, and small interfering RNAs targeting PDGFRß blocked this phosphorylation without having any effect on MEK/Erk1/2 activation. We also found that PDGFRß regulates high glucose-induced Akt activation, its targets tuberin and PRAS40 phosphorylation, and finally, mTORC1 activation. Furthermore, inhibition of PDGFRß suppressed high glucose-induced expression of collagen I (α2) in proximal tubular cells. Importantly, expression of constitutively active Akt or mTORC1 reversed these processes. As a mechanism, we found that JNJ and PDGFRß knockdown inhibited high glucose-stimulated Hif1α expression. Furthermore, overexpression of Hif1α restored expression of collagen I (α2) that was inhibited by PDGFRß knockdown in high glucose-stimulated cells. Finally, we show increased phosphorylation of PDGFRß and its association with Akt/mTORC1 activation, Hif1α expression, and elevated collagen I (α2) levels in the renal cortex of mice with diabetes. Our results identify PDGFRß as a driver in activating Akt/mTORC1 nexus for high glucose-mediated expression of collagen I (α2) in proximal tubular epithelial cells, which contributes to tubulointerstitial fibrosis in diabetic nephropathy.


Subject(s)
Blood Glucose/metabolism , Collagen Type I/metabolism , Diabetic Nephropathies/enzymology , Kidney Tubules, Proximal/enzymology , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/blood , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Disease Models, Animal , Fibrosis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Tubules, Proximal/pathology , Male , Mechanistic Target of Rapamycin Complex 1 , Mice, Transgenic , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation , RNA Interference , Receptor, Platelet-Derived Growth Factor beta/genetics , Signal Transduction , Time Factors , Transfection , Up-Regulation
9.
J Biol Chem ; 292(14): 5665-5675, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28188286

ABSTRACT

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.


Subject(s)
Epithelial Cells/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Glucose/pharmacology , Hydrogen Sulfide/pharmacology , Kidney Tubules, Proximal/enzymology , NADPH Oxidases/biosynthesis , Nitric Oxide Synthase Type II/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Amidines/pharmacology , Animals , Benzylamines/pharmacology , Diabetic Nephropathies/enzymology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/therapy , Epithelial Cells/pathology , Extracellular Matrix Proteins/metabolism , Kidney Tubules, Proximal/pathology , Mice , NADPH Oxidase 4 , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/antagonists & inhibitors , Oxidative Stress/drug effects
10.
J Gerontol A Biol Sci Med Sci ; 71(7): 850-7, 2016 07.
Article in English | MEDLINE | ID: mdl-26442901

ABSTRACT

We examined the effect of rapamycin on the life span of a mouse model of type 2 diabetes, db/db mice. At 4 months of age, male and female C57BLKSJ-lepr (db/db) mice (db/db) were placed on either a control diet, lacking rapamycin or a diet containing rapamycin and maintained on these diets over their life span. Rapamycin was found to reduce the life span of the db/db mice. The median survival of male db/db mice fed the control and rapamycin diets was 349 and 302 days, respectively, and the median survival of female db/db mice fed the control and rapamycin diets was 487 and 411 days, respectively. Adjusting for gender differences, rapamycin increased the mortality risk 1.7-fold in both male and female db/db mice. End-of-life pathological data showed that suppurative inflammation was the main cause of death in the db/db mice, which is enhanced slightly by rapamycin treatment.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Inflammation/pathology , Longevity , Sirolimus , Animals , Cause of Death , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/mortality , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/mortality , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Female , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Longevity/drug effects , Longevity/physiology , Male , Mice , Mice, Inbred C57BL , Mortality , Sex Factors , Sirolimus/metabolism , Sirolimus/pharmacology , Treatment Outcome
11.
J Biol Chem ; 289(47): 32703-16, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25288788

ABSTRACT

High glucose-induced Akt acts as a signaling hub for mesangial cell hypertrophy and matrix expansion, which are recognized as cardinal signatures for the development of diabetic nephropathy. How mesangial cells sustain the activated state of Akt is not clearly understood. Here we show Akt-dependent phosphorylation of the transcription factor FoxO1 by high glucose. Phosphorylation-deficient, constitutively active FoxO1 inhibited the high glucose-induced phosphorylation of Akt to suppress the phosphorylation/inactivation of PRAS40 and mTORC1 activity. In contrast, dominant negative FoxO1 increased the phosphorylation of Akt, resulting in increased mTORC1 activity similar to high glucose treatment. Notably, FoxO1 regulates high glucose-induced protein synthesis, hypertrophy, and expression of fibronectin and PAI-1. High glucose paves the way for complications of diabetic nephropathy through the production of reactive oxygen species (ROS). We considered whether the FoxO1 target antioxidant enzyme catalase contributes to sustained activation of Akt. High glucose-inactivated FoxO1 decreases the expression of catalase to increase the production of ROS. Moreover, we show that catalase blocks high glucose-stimulated Akt phosphorylation to attenuate the inactivation of FoxO1 and PRAS40, resulting in the inhibition of mTORC1 and mesangial cell hypertrophy and fibronectin and PAI-1 expression. Finally, using kidney cortices from type 1 diabetic OVE26 mice, we show that increased FoxO1 phosphorylation is associated with decreased catalase expression and increased fibronectin and PAI-1 expression. Together, our results provide the first evidence for the presence of a positive feedback loop for the sustained activation of Akt involving inactivated FoxO1 and a decrease in catalase expression, leading to increased ROS and mesangial cell hypertrophy and matrix protein expression.


Subject(s)
Feedback, Physiological/drug effects , Forkhead Transcription Factors/metabolism , Glucose/pharmacology , Mesangial Cells/drug effects , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Catalase/genetics , Catalase/metabolism , Cell Size/drug effects , Cells, Cultured , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Fibronectins/metabolism , Gene Expression/drug effects , Immunoblotting , Kidney Cortex/metabolism , Kidney Cortex/pathology , Mechanistic Target of Rapamycin Complex 1 , Mesangial Cells/metabolism , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/metabolism , Rats , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
Am J Physiol Cell Physiol ; 306(11): C1089-100, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24740537

ABSTRACT

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.


Subject(s)
Cell Proliferation , Feedback, Physiological/physiology , Mesangial Cells/metabolism , Mitogen-Activated Protein Kinase 7/biosynthesis , Oncogene Protein v-akt/biosynthesis , Platelet-Derived Growth Factor/pharmacology , Animals , Cell Proliferation/drug effects , Cells, Cultured , Feedback, Physiological/drug effects , Mesangial Cells/drug effects , Rats
13.
J Biol Chem ; 288(40): 28668-86, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23940049

ABSTRACT

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.


Subject(s)
Angiotensin II/pharmacology , Fibronectins/metabolism , Mitochondria/metabolism , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Peroxynitrous Acid/pharmacology , Reactive Oxygen Species/metabolism , Animals , Biological Availability , Fibrosis , Gene Silencing/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Mesangial Cells/drug effects , Mesangial Cells/enzymology , Mesangial Cells/pathology , Mitochondria/drug effects , Mitochondria/enzymology , Models, Biological , NADPH Oxidase 4 , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Oxidative Stress/drug effects , Rats , Superoxide Dismutase/metabolism , Superoxides/metabolism , Up-Regulation/drug effects
14.
J Biol Chem ; 288(34): 24503-17, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23821550

ABSTRACT

Skeletal remodeling consists of timely formation and resorption of bone by osteoblasts and osteoclasts in a quantitative manner. Patients with chronic myeloid leukemia receiving inhibitors of c-Abl tyrosine kinase often show reduced bone remodeling due to impaired osteoblast and osteoclast function. BMP-2 plays a significant role in bone generation and resorption by contributing to the formation of mature osteoblasts and osteoclasts. The effects of c-Abl on BMP-2-induced bone remodeling and the underlying mechanisms are not well studied. Using a pharmacological inhibitor and expression of a dominant negative mutant of c-Abl, we show an essential role of this tyrosine kinase in the development of bone nodules containing mature osteoblasts and formation of multinucleated osteoclasts in response to BMP-2. Calvarial osteoblasts prepared from c-Abl null mice showed the absolute requirement of this tyrosine kinase in maturation of osteoblasts and osteoclasts. Activation of phosphatidylinositol 3-kinase (PI 3-kinase)/Akt signaling by BMP-2 leads to osteoblast differentiation. Remarkably, inhibition of c-Abl significantly suppressed BMP-2-stimulated PI 3-kinase activity and its downstream Akt phosphorylation. Interestingly, c-Abl regulated BMP-2-induced osteoclastogenic CSF-1 expression. More importantly, we identified the requirements of c-Abl in BMP-2 autoregulation and the expressions of alkaline phosphatase and osterix that are necessary for osteoblast differentiation. c-Abl contributed to BMP receptor-specific Smad-dependent transcription of CSF-1, osterix, and BMP-2. Finally, c-Abl associates with BMP receptor IA and regulates phosphorylation of Smad in response to BMP-2. We propose that activation of c-Abl is an important step, which induces into two signaling pathways involving noncanonical PI 3-kinase and canonical Smads to integrate BMP-2-induced osteogenesis.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Smad5 Protein/metabolism , Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/genetics , Animals , Bone Morphogenetic Protein 2/genetics , Cell Line , Gene Expression Regulation/physiology , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/genetics , Mice , Osteoblasts/cytology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Skull/cytology , Skull/metabolism , Smad5 Protein/genetics , Sp7 Transcription Factor , Transcription Factors/biosynthesis , Transcription Factors/genetics
15.
Biochem J ; 433(2): 393-402, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21029048

ABSTRACT

BMP-2 (bone morphogenetic protein-2) promotes differentiation of osteoblast precursor cells to mature osteoblasts that form healthy bone. In the present study, we demonstrate a novel mechanism of BMP-2-induced osteoblast differentiation. The antioxidant NAC (N-acetyl-L-cysteine) and the flavoprotein enzyme NAD(P)H oxidase inhibitor DPI (diphenyleneiodonium) prevented BMP-2-stimulated alkaline phosphatase expression and mineralized bone nodule formation in mouse 2T3 pre-osteoblasts. BMP-2 elicited a rapid generation of ROS (reactive oxygen species) concomitant with increased activation of NAD(P)H oxidase. NAC and DPI inhibited BMP-2-induced ROS production and NAD(P)H oxidase activity respectively. NAD(P)H oxidases display structurally similar catalytic subunits (Nox1-5) with differential expression in various cells. We demonstrate that 2T3 pre-osteoblasts predominantly express the Nox4 isotype of NAD(P)H oxidase. To extend this finding, we tested the functional effects of Nox4. Adenovirus-mediated expression of dominant-negative Nox4 inhibited BMP-2-induced alkaline phosphatase expression. BMP-2 promotes expression of BMP-2 for maintenance of the osteoblast phenotype. NAC and DPI significantly blocked BMP-2-stimulated expression of BMP2 mRNA and protein due to a decrease in BMP2 gene transcription. Dominant-negative Nox4 also mimicked this effect of NAC and DPI. Our results provide the first evidence for a new signalling pathway linking BMP-2-stimulated Nox4-derived physiological ROS to BMP-2 expression and osteoblast differentiation.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Cell Differentiation , NADPH Oxidases/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Reactive Oxygen Species/metabolism , Transcription, Genetic , Alkaline Phosphatase/metabolism , Animals , Cell Line , Mice , NADPH Oxidase 4 , RNA, Messenger/genetics
16.
J Biol Chem ; 285(48): 37503-12, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20861022

ABSTRACT

Diabetes and high glucose (HG) increase the generation of NADPH oxidase-derived reactive oxygen species and induce apoptosis of glomerular epithelial cells (podocytes). Loss of podocytes contributes to albuminuria, a major risk factor for progression of kidney disease. Here, we show that HG inactivates AMP-activated protein kinase (AMPK), up-regulates Nox4, enhances NADPH oxidase activity, and induces podocyte apoptosis. Activation of AMPK blocked HG-induced expression of Nox4, NADPH oxidase activity, and apoptosis. We also identified the tumor suppressor protein p53 as a mediator of podocyte apoptosis in cells exposed to HG. Inactivation of AMPK by HG up-regulated the expression and phosphorylation of p53, and p53 acted downstream of Nox4. To investigate the mechanism of podocyte apoptosis in vivo, we used OVE26 mice, a model of type 1 diabetes. Glomeruli isolated from these mice showed decreased phosphorylation of AMPK and enhanced expression of Nox4 and p53. Pharmacologic activation of AMPK by 5-aminoimidazole-4-carboxamide-1-riboside in OVE26 mice attenuated Nox4 and p53 expression. Administration of 5-aminoimidazole-4-carboxamide-1-riboside also prevented renal hypertrophy, glomerular basement thickening, foot process effacement, and podocyte loss, resulting in marked reduction in albuminuria. Our results uncover a novel function of AMPK that integrates metabolic input to Nox4 and provide new insight for activation of p53 to induce podocyte apoptosis. The data indicate the potential therapeutic utility of AMPK activators to block Nox4 and reactive oxygen species generation and to reduce urinary albumin excretion in type 1 diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis , Diabetes Mellitus/genetics , Down-Regulation , Podocytes/cytology , Tumor Suppressor Protein p53/genetics , AMP-Activated Protein Kinases/genetics , Animals , Diabetes Mellitus/enzymology , Diabetes Mellitus/metabolism , Diabetes Mellitus/physiopathology , Disease Models, Animal , Female , Gene Expression Regulation , Glucose/genetics , Humans , Male , Mice , NADPH Oxidase 4 , NADPH Oxidases , Phosphorylation , Podocytes/metabolism , Tumor Suppressor Protein p53/metabolism
17.
J Cell Physiol ; 225(1): 27-41, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20629086

ABSTRACT

Diabetic nephropathy manifests aberrant activation of TORC1, which senses key signals to modulate protein synthesis and renal hypertrophy. PRAS40 has recently been identified as a raptor-interacting protein and is a component and a constitutive inhibitor of TORC1. The mechanism by which high glucose stimulates TORC1 activity is not known. PRAS40 was identified in the mesangial cells in renal glomeruli and in tubulointerstitium of rat kidney. Streptozotocin-induced diabetic renal hypertrophy was associated with phosphorylation of PRAS40 in the cortex and glomeruli. In vitro, high glucose concentration increased PRAS40 phosphorylation in a PI 3 kinase- and Akt-dependent manner, resulting in dissociation of raptor-PRAS40 complex in mesangial cells. High glucose augmented the inactivating and activating phosphorylation of 4EBP-1 and S6 kinase, respectively, with concomitant induction of protein synthesis and hypertrophy. Expression of TORC1-nonphosphorylatable mutant of 4EBP-1 and dominant-negative S6 kinase significantly inhibited high glucose-induced protein synthesis and hypertrophy. PRAS40 knockdown mimicked the effect of high glucose on phosphorylation of 4EBP-1 and S6 kinase, protein synthesis, and hypertrophy. To elucidate the role of PRAS40 phosphorylation, we used phosphorylation-deficient mutant of PRAS40, which in contrast to PRAS40 knockdown inhibited phosphorylation of 4EBP-1 and S6 kinase, leading to reduced mesangial cell hypertrophy. Thus, our data identify high glucose-induced phosphorylation and inactivation of PRAS40 as a central node for mesangial cell hypertrophy in diabetic nephropathy.


Subject(s)
Glucose/metabolism , Mesangial Cells/metabolism , Mesangial Cells/pathology , Phosphoproteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Enzyme Activation , Intracellular Signaling Peptides and Proteins , Male , Mesangial Cells/cytology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases/genetics , Ribosomal Protein S6 Kinases/metabolism , Transcription Factors/genetics
18.
Cell Signal ; 22(5): 749-58, 2010 May.
Article in English | MEDLINE | ID: mdl-20060890

ABSTRACT

Sustained activation of Akt kinase acts as a focal regulator to increase cell growth and survival, which causes tumorigenesis including breast cancer. Statins, potent inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase, display anticancer activity. The molecular mechanisms by which statins block cancer cell growth are poorly understood. We demonstrate that in the tumors derived from MDA-MB-231 human breast cancer cell xenografts, simvastatin significantly inhibited phosphorylation of Akt with concomitant attenuation of the expression of the anti-apoptotic protein Bcl(XL). In many cancer cells, Bcl(XL) is a target of NFkappaB. Simvastatin inhibited the DNA binding and transcriptional activities of NFkappaB resulting in marked reduction in transcription of Bcl(XL). Signals transmitted by anti-neoplastic mechanism implanted in the cancer cells serve to obstruct the initial outgrowth of tumors. One such mechanism represents the action of the tumor suppressor protein PTEN, which negatively regulates Akt kinase activity. We provide the first evidence for significantly increased levels of PTEN in the tumors of simvastatin-administered mice. Importantly, simvastatin markedly prevented binding of NFkappaB to the two canonical recognition elements, NFRE-1 and NFRE-2 present in the PTEN promoter. Contrary to the transcriptional suppression of Bcl(XL), simvastatin significantly increased the transcription of PTEN. Furthermore, expression of NFkappaB p65 subunit inhibited transcription of PTEN, resulting in reduced protein expression, which leads to enhanced phosphorylation of Akt. Taken together, our data present a novel bifaceted mechanism where simvastatin acts on a nodal transcription factor NFkappaB, which attenuates the expression of anti-apoptotic Bcl(XL) and simultaneously derepresses the expression of anti-proliferative/proapoptotic tumor suppressor PTEN to prevent breast cancer cell growth.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , NF-kappa B/metabolism , PTEN Phosphohydrolase/genetics , Simvastatin/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Female , Humans , Mice , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Response Elements/genetics , Transcription, Genetic/drug effects , Xenograft Model Antitumor Assays , bcl-X Protein/genetics
19.
Endocrinology ; 150(11): 4989-98, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19819979

ABSTRACT

Murine spleen cells produce mature osteoclasts when cocultured with osteoblastic cells. Colony-stimulating factor (CSF)-1 is the growth factor required for differentiating the monocyte-macrophage precursor cells into preosteoclasts. Bone morphogenic protein (BMP) signaling in osteoblasts regulates bone mass in mice, suggesting a role of BMP in osteoclastogenesis along with osteoblast activity. The intracellular signal transduction cross talk regulating the osteoblastic production of CSF-1 as a mechanism of BMP-induced osteoclastogenesis is described in this report. We have recently described the involvement of Smad 1/5 in BMP-2-induced CSF-1 expression and osteoclast formation. In this study, using the pharmacological inhibitors and the adenovirus (Ad) vectors expressing dominant-negative (DN) phosphatidylinositol 3 kinase (PI3K), the PI3K-signaling inhibitor, phosphatase and tensin homolog deleted in chromosome 10 (PTEN) or DN Akt kinase in the in vitro coculture assay, we show an essential role of the lipid kinase cascade in BMP-2-mediated multinucleated osteoclast formation and CSF-1 mRNA expression, transcription, and secretion. Inhibition of PI3K/Akt signaling blocked the binding of Smads 1/5 to the CSF-1 BMP-responsive element present in the CSF-1 promoter, resulting in attenuation of Smad-dependent CSF-1 transcription. Furthermore, PI3K inhibition and DN Akt prevented association of the transcriptional coactivator, CREB (cAMP response element binding protein) binding protein (CBP), with Smads 1/5. Together, these data for the first time demonstrate that PI3K-dependent Akt activation regulates BMP-2-induced CSF-1 expression and provides a mechanism for osteoblastic cell-assisted osteoclast differentiation.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation , Macrophage Colony-Stimulating Factor/genetics , Osteoblasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Bone Morphogenetic Protein 2/genetics , Cell Line , Gene Expression , Macrophage Colony-Stimulating Factor/metabolism , Mice , Osteoblasts/cytology , Phosphatidylinositol 3-Kinases/genetics , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism
20.
J Biol Chem ; 284(41): 27790-27798, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19648120

ABSTRACT

Substantial evidence suggests roles of TSC2 and PTEN in the development of cancer predisposition syndromes. Loss of TSC2 results in benign tumors, neurological disorders, and angiomyolipomas. We found that PTEN mRNA and protein levels are elevated in Tsc2(-/-) mouse embryo fibroblasts with concomitant reduction in Akt phosphorylation. Reconstitution of TSC2 in Tsc2(-/-) mouse embryo fibroblasts decreases PTEN levels. Interestingly, increased HIF1alpha activity present in Tsc2 null cells is required for PTEN transcription and protein expression. We identified a canonical hypoxia-responsive element in the PTEN promoter, which regulates the transcription of this tumor suppressor protein in a TSC2-dependent manner. Finally, we demonstrate a positive correlation between expression of HIF1alpha and PTEN in renal angiomyolipomas from TSC patients. Our results reveal a unique function of HIF1alpha in up-regulation of PTEN and provide a new mechanism of reduced Akt phosphorylation in Tsc2 null cells. These data suggest that PTEN may safeguard against developing malignant tumors in patients with TSC deficiency.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , PTEN Phosphohydrolase/metabolism , Tumor Suppressor Proteins/metabolism , Angiomyolipoma/genetics , Angiomyolipoma/metabolism , Angiomyolipoma/pathology , Animals , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...