Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 7(10): e48652, 2012.
Article in English | MEDLINE | ID: mdl-23119081

ABSTRACT

Decreases in circulating 25,hydroxyl-vitamin D3 (25 OH D3) and 1,25,dihydroxyl-vitamin D3 (1,25 (OH)2 D3) have been extensively documented in patients with type 2 diabetes. Nevertheless, the molecular reasons behind this drop, and whether it is a cause or an effect of disease progression is still poorly understood. With the skin and the liver, the kidney is one of the most important sites for vitamin D metabolism. Previous studies have also shown that CYP24A1 (an enzyme implicated in vitamin D metabolism), might play an important role in furthering the progression of kidney lesions during diabetic nephropathy. In this study we show a link between CYP24A1 increase and senescence followed by apoptosis induction in the renal proximal tubules of diabetic kidneys. We show that CYP24A1 expression was increased during diabetic nephropathy progression. This increase derived from protein kinase C activation and increased H(2)O(2) cellular production. CYP24A1 increase had a major impact on cellular phenotype, by pushing cells into senescence, and later into apoptosis. Our data suggest that control of CYP24A1 increase during diabetes has a beneficial effect on senescence induction and caspase-3 increased expression. We concluded that diabetes induces an increase in CYP24A1 expression, destabilizing vitamin D metabolism in the renal proximal tubules, leading to cellular instability and apoptosis, and thereby accelerating tubular injury progression during diabetic nephropathy.


Subject(s)
Apoptosis , Caspase 3/metabolism , Diabetes Mellitus, Type 2/metabolism , Kidney Tubules/metabolism , Steroid Hydroxylases/metabolism , Animals , Blotting, Western , Caspase 3/genetics , Cells, Cultured , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Diet, High-Fat , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression , Glucose/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidants/pharmacology , RNA Interference , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Steroid Hydroxylases/genetics , Up-Regulation/drug effects , Vitamin D3 24-Hydroxylase
2.
Diabetes ; 61(2): 474-84, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22210314

ABSTRACT

This study investigated the mechanisms underlying tubular apoptosis in diabetes by identifying proapoptotic genes that are differentially upregulated by reactive oxygen species in renal proximal tubular cells (RPTCs) in models of diabetes. Total RNAs isolated from renal proximal tubules (RPTs) of 20-week-old heterozygous db/m+, db/db, and db/db catalase (CAT)-transgenic (Tg) mice were used for DNA chip microarray analysis. Real-time quantitative PCR assays, immunohistochemistry, and mice rendered diabetic with streptozotocin were used to validate the proapoptotic gene expression in RPTs. Cultured rat RPTCs were used to confirm the apoptotic activity and regulation of proapoptotic gene expression. Additionally, studies in kidney tissues from patients with and without diabetes were used to confirm enhanced proapoptotic gene expression in RPTs. Bcl-2-modifying factor (Bmf) was differentially upregulated (P<0.01) in RPTs of db/db mice compared with db/m+ and db/db CAT-Tg mice and in RPTs of streptozotocin-induced diabetic mice in which insulin reversed this finding. In vitro, Bmf cDNA overexpression in rat RPTCs coimmunoprecipated with Bcl-2, enhanced caspase-3 activity, and promoted apoptosis. High glucose (25 mmol/L) induced Bmf mRNA expression in RPTCs, whereas rotenone, catalase, diphenylene iodinium, and apocynin decreased it. Knockdown of Bmf with small interfering RNA reduced high glucose-induced apoptosis in RPTCs. More important, enhanced Bmf expression was detected in RPTs of kidneys from patients with diabetes. These data demonstrate differential upregulation of Bmf in diabetic RPTs and suggest a potential role for Bmf in regulating RPTC apoptosis and tubular atrophy in diabetes.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis , Diabetes Mellitus, Experimental/pathology , Kidney Tubules, Proximal/pathology , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Animals , Caspase 3/metabolism , Cells, Cultured , Humans , Immunohistochemistry , Kidney Tubules, Proximal/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/analysis , Rats , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
3.
Kidney Int ; 77(12): 1086-97, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20237455

ABSTRACT

Transgenic mice that overexpress angiotensinogen, the sole precursor of angiotensins, in their renal proximal tubular cells develop hypertension, albuminuria, and tubular apoptosis. These pathological changes are due to enhanced generation of reactive oxygen species in the proximal tubule cells. Here, we determined whether overexpression of catalase to decrease oxidant injury in the proximal tubular cells could reverse these abnormalities. Double-transgenic mice specifically overexpressing angiotensinogen and catalase in their renal proximal tubular cells were created by cross-breeding the single transgenics. Non-transgenic littermates served as controls. Overexpression of catalase prevented hypertension, albuminuria, tubulointerstitial fibrosis, and tubular apoptosis in the angiotensinogen transgenic mice. Furthermore, the double transgenics had lower reactive oxygen species generation and reduced pro-fibrotic and apoptotic gene expression in the renal proximal tubular cells. Renal angiotensin converting enzyme-2 expression and urinary angiotensin 1-7 levels were downregulated in the single but normal in the double-transgenic mice. Thus, we suggest that the intrarenal renin-angiotensin system and reactive oxygen species generation have an important role in the development of hypertension and renal injury.


Subject(s)
Angiotensinogen/genetics , Catalase/administration & dosage , Hypertension/prevention & control , Kidney Tubules, Proximal/pathology , Animals , Apoptosis/drug effects , Catalase/genetics , Catalase/therapeutic use , Gene Expression , Hypertension/etiology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Mice , Mice, Transgenic , Reactive Oxygen Species/metabolism , Renin-Angiotensin System/physiology
4.
J Am Soc Nephrol ; 21(6): 943-54, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20299359

ABSTRACT

Apoptosis of tubular epithelial cells contributes to the tubular atrophy that accompanies diabetic nephropathy. Reactive oxygen species (ROS) promote tubular apoptosis, but the mechanisms by which this occurs are incompletely understood. Here, we sought proapoptotic genes that ROS differentially upregulate in renal proximal tubular cells of diabetic (db/db) mice. We performed microarray analysis using total RNA from freshly isolated renal proximal tubules of nondiabetic, diabetic, and diabetic transgenic mice overexpressing catalase in the proximal tubule (thereby attenuating ROS). We observed greater expression of caspase-12 in the proximal tubules of the diabetic mice compared with the nondiabetic and diabetic transgenic mice. Quantitative PCR and immunohistochemistry confirmed the enhanced expression of caspase-12, as well as members of the endoplasmic reticulum stress-induced apoptotic pathway. Ex vivo, albumin induced caspase-12 activity and expression (protein and mRNA) and mRNA expression of the CCAT/enhancer-binding protein homologous protein in freshly isolated wild-type proximal tubules but not in catalase-overexpressing proximal tubules. In vitro, albumin stimulated activity of both caspase-12 and caspase-3 as well as expression of caspase-12 and CCAT/enhancer-binding protein homologous protein in a human proximal tubule cell line (HK-2). The free radical scavenger tiron inhibited these effects. Furthermore, knockdown of caspase-12 with small interfering RNA reduced albumin-induced apoptosis in HK-2 cells. Taken together, these studies demonstrate that albuminuria may induce tubular apoptosis through generation of ROS and the subsequent expression and activation of endoplasmic reticulum stress genes in the diabetic kidney.


Subject(s)
Apoptosis , Caspase 12/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Reactive Oxygen Species/metabolism , Albumins/pharmacology , Animals , Apoptosis/drug effects , Caspase 12/genetics , Cell Line , Disease Models, Animal , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation/drug effects , Heat-Shock Proteins/metabolism , Humans , Male , Mice , Mice, Mutant Strains , Mice, Transgenic , RNA, Small Interfering/pharmacology , Transcription Factor CHOP/metabolism
5.
Hepatology ; 50(4): 1121-9, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19637194

ABSTRACT

UNLABELLED: We identified the far upstream element binding protein 1 (FBP1), an activator of transcription of the proto-oncogene c-myc, in a functional yeast survival screen for tumor-related antiapoptotic proteins and demonstrated strong overexpression of FBP1 in human hepatocellular carcinoma (HCC). Knockdown of the protein in HCC cells resulted in increased sensitivity to apoptotic stimuli, reduced cell proliferation, and impaired tumor formation in a mouse xenograft transplantation model. Interestingly, analysis of gene regulation in these cells revealed that c-myc levels were not influenced by FBP1 in HCC cells. Instead, we identified the cell cycle inhibitor p21 as a direct target gene repressed by FBP1, and in addition, expression levels of the proapoptotic genes tumor necrosis factor alpha, tumor necrosis factor-related apoptosis-inducing ligand, Noxa, and Bik were elevated in the absence of FBP1. CONCLUSION: Our data establish FBP1 as an important oncoprotein overexpressed in HCC that induces tumor propagation through direct or indirect repression of cell cycle inhibitors and proapoptotic target genes.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Animals , Apoptosis/physiology , Cell Cycle/physiology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Humans , Mice , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins , Signal Transduction/physiology , Transplantation, Heterologous
6.
J Am Soc Nephrol ; 19(2): 269-80, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18057217

ABSTRACT

The intrarenal renin-angiotensin system (RAS) plays an important role in the progression of diabetic nephropathy. We have previously reported that mice overexpressing angiotensinogen in renal proximal tubular cells (RPTC) develop hypertension, albuminuria, and renal injury. Here, we investigated whether activation of the intrarenal RAS contributes to apoptosis of RPTC in diabetes. Induction of diabetes with streptozotocin in these transgenic mice led to significant increases in BP, albuminuria, RPTC apoptosis, and proapoptotic gene expression compared with diabetic nontransgenic littermates. Insulin and/or RAS blockers markedly attenuated these changes. Hydralazine prevented hypertension but not albuminuria, RPTC apoptosis, or proapoptotic gene expression. In vitro, high-glucose medium significantly increased apoptosis and caspase-3 activity in rat immortalized RPTC overexpressing angiotensinogen compared with control cells, and these changes were prevented by insulin and/or RAS blockers. In conclusion, intrarenal RAS activation and high glucose may act in concert to increase tubular apoptosis in diabetes, independent of systemic hypertension.


Subject(s)
Angiotensinogen/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Kidney Tubules, Proximal/pathology , Kidney Tubules, Proximal/physiology , Albuminuria/drug therapy , Albuminuria/pathology , Albuminuria/physiopathology , Angiotensinogen/physiology , Animals , Antihypertensive Agents/pharmacology , Apoptosis/physiology , Blood Glucose/metabolism , Cell Line, Transformed , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Nephropathies/drug therapy , Hydralazine/pharmacology , Hypertension, Renal/drug therapy , Hypertension, Renal/pathology , Hypertension, Renal/physiopathology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Losartan/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Perindopril/pharmacology , Rats , Renin-Angiotensin System/physiology , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
7.
Diabetes ; 57(2): 451-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17977949

ABSTRACT

OBJECTIVE: The present study investigated the relationships between reactive oxygen species (ROS), interstitial fibrosis, and renal proximal tubular cell (RPTC) apoptosis in type 2 diabetic db/db mice and in db/db transgenic (Tg) mice overexpressing rat catalase (rCAT) in their RPTCs (db/db rCAT-Tg). RESEARCH DESIGN AND METHODS: Blood pressure, blood glucose, and albuminuria were monitored for up to 5 months. Kidneys were processed for histology and apoptosis studies (terminal transferase-mediated dUTP nick-end labeling or immunostaining for active caspase-3 and Bax). Real-time quantitative PCR assays were used to quantify angiotensinogen (ANG), p53, and Bax mRNA levels. RESULTS: db/db mice developed obesity, hyperglycemia, hypertension, and albuminuria. In contrast, db/db rCAT-Tg mice became obese and hyperglycemic but had normal blood pressure and attenuated albuminuria compared with db/db mice. Kidneys from db/db mice displayed progressive glomerular hypertrophy, glomerulosclerosis, interstitial fibrosis, and tubular apoptosis and increased expression of collagen type IV, Bax, and active caspase-3, as well as increased ROS production. These changes, except glomerular hypertrophy, were markedly attenuated in kidneys of db/db rCAT-Tg mice. Furthermore, ANG, p53, and Bax mRNA expression was increased in renal proximal tubules of db/db mice but not of db/db rCAT-Tg mice. CONCLUSIONS: Our results indicate a crucial role for intra-renal ROS in the progression of hypertension, albuminuria, interstitial fibrosis, and tubular apoptosis in type 2 diabetes and demonstrate the beneficial effects of suppressing ROS formation.


Subject(s)
Catalase/genetics , Diabetes Mellitus, Type 2/physiopathology , Kidney Tubules, Proximal/enzymology , Nephritis, Interstitial/genetics , Animals , Apoptosis , Catalase/metabolism , Cattle , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetic Nephropathies , In Situ Nick-End Labeling , Kidney Tubules, Proximal/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nephritis, Interstitial/enzymology , Obesity/genetics , Reactive Oxygen Species/metabolism
8.
FASEB J ; 17(10): 1295-7, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12759333

ABSTRACT

Apoptosis is a fundamental biological process used to eliminate unwanted cells in a multicellular organism. An increasing number of regulatory proteins have been identified that either promote or inhibit apoptosis. For tumors to arise, apoptosis must be blocked in the transformed cells, for example by mutational overexpression of anti-apoptotic proteins, which represent attractive target proteins for molecular therapy strategies. In a functional yeast survival screen designed to select new anti-apoptotic mammalian genes, we have identified the chromosomal high-mobility group box-1 protein (HMGB1) as an inhibitor of yeast cell death induced by the pro-apoptotic Bcl-2 family member Bak. The C-terminal 33 amino acids of HMGB1 are dispensable for this inhibitory function. HMGB1 is also able to protect mammalian cells against different death stimuli including ultraviolet radiation, CD95-, TRAIL-, Casp-8-, and Bax-induced apoptosis. We found high HMGB1 protein levels in human primary breast carcinoma. Hmgb1 RNA levels are changing during different stages of mouse mammary gland development and are particularly low during lactation and involution. These data suggest that HMGB1 may participate in the regulation of mammary gland apoptosis and that its high expression level promotes tumor growth because of its anti-apoptotic properties.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Carcinoma/metabolism , HMGB1 Protein/metabolism , HMGB1 Protein/physiology , Animals , Cell Line , Cytoprotection , Female , Gene Expression Regulation, Developmental , HMGB1 Protein/genetics , Humans , Mammary Glands, Animal/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Models, Biological , RNA, Messenger/metabolism , Rats , Schizosaccharomyces/cytology , Schizosaccharomyces/genetics , Transformation, Genetic , Tumor Cells, Cultured , bcl-2 Homologous Antagonist-Killer Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...