Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Am J Physiol Heart Circ Physiol ; 297(6): H2109-19, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19820199

ABSTRACT

The aim of this paper is to study the myocardial damage secondary to long-term streptozotocin-induced type 1 diabetes mellitus (DM1). Normotensive and spontaneously hypertensive rats (SHR) received either streptozotocin injections or vehicle. After 22 or 6 wk, DM1, SHR, DM1/SHR, and control rats were killed, and the left ventricles studied by histology, quantitative PCR, Western blot, ELISA, and electromobility shift assay. Cardiomyocyte cultures were also performed. The expression of profibrotic factors, transforming growth factor-beta (TGF-beta1), connective tissue growth factor, and matrix proteins was increased, and the TGF-beta1-linked transcription factors phospho-Smad3/4 and activator protein-1 were activated in the DM1 myocardium. Proapoptotic molecules FasL, Fas, Bax, and cleaved caspase-3 were also augmented. Myocardial injury in long-term hypertension shared these features. In addition, hypertension was associated with activation of NF-kappaB, increased inflammatory cell infiltrate, and expression of the mediators [interleukin-1beta (IL-1beta), tumor necrosis factor-alpha, monocyte chemoattractant protein 1, vascular cell adhesion molecule 1, angiotensinogen, and oxidants], which were absent in long-term DM1. At this stage, the combination of DM1 and hypertension resulted in nonsignificant additive effects. Moreover, the coexistence of DM1 blunted the inflammatory response to hypertension. Anti-inflammatory IL-10 and antioxidants were induced in long-term DM1 and DM1/SHR hearts. Myocardial inflammation was, however, observed in the short-term model. In cultured cardiomyocytes, IL-10, TGF-beta1, and catalase blocked the glucose-stimulated expression of proinflammatory genes. Fibrosis and apoptosis are features of long-term myocardial damage in experimental DM1. Associated hypertension does not induce additional changes. Myocardial inflammation is present in hypertension and short-term DM1, but is not a key feature in long-term DM1. Local reduction of proinflammatory factors and expression of anti-inflammatory and antioxidant molecules may underlie this effect.


Subject(s)
Apoptosis , Cardiomyopathies/etiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 1/complications , Hypertension/complications , Inflammation/prevention & control , Myocardium/pathology , Animals , Antioxidants/metabolism , Apoptosis Regulatory Proteins/metabolism , Biomarkers/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Extracellular Matrix Proteins/metabolism , Fibrosis , Hypertension/metabolism , Hypertension/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Myocardium/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Time Factors
2.
Nefrologia ; 28 Suppl 6: 23-6, 2008.
Article in English | MEDLINE | ID: mdl-18957008

ABSTRACT

A high glucose concentration is shared by peritoneal dialysis (PD) and diabetes mellitus (DM). High glucose leads to tissue injury in diabetes. Peritoneal dialysis research has emphasized the role of glucose degradation products in tissue injury. Apoptosis induction is one of the mechanisms of tissue injury induced both by glucose and glucose degradation products. We now review the role of apoptosis and its regulation by glucose degradation products in antibacterial defense and loss of renal function in diabetes mellitus and peritoneal dialysis. The pathogenic role of the recently identified glucose degradation product 3,4-di-deoxyglucosone- 3-ene (3,4-DGE) is detailed. Available therapeutic strategies include the use of peritoneal dialysis solutions containing a low concentration of glucose degradation products. Based on preclinical results, specific targeting of apoptosis regulatory factor should be explored in the clinical setting.


Subject(s)
Apoptosis/physiology , Diabetes Mellitus/drug therapy , Diabetes Mellitus/immunology , Peritoneal Dialysis , Apoptosis/drug effects , Bacterial Infections/immunology , Diabetes Mellitus/microbiology , Humans
3.
Curr Med Chem ; 13(4): 449-54, 2006.
Article in English | MEDLINE | ID: mdl-16475932

ABSTRACT

Tubular cell loss is prominent both in acute and chronic renal failure. Apoptosis and its regulatory mechanisms contribute to cell number regulation in the kidney. The potential role of apoptosis ranges from induction and progression to repair of renal injury. However, therapeutic interest has focused in preventing the apoptotic loss of tubular cells that leads to acute and chronic renal failure. Death ligands and receptors, such as tumor necrosis factor and Fas ligand, proapoptotic and antiapoptotic Bcl2 family members and caspases have all been shown to participate in apoptosis regulation in the course of renal cell injury. Nevertheless, the precise role of these proteins is unclear, and the participation of most known apoptosis regulatory proteins has not been studied. We now review the role of apoptosis in renal injury, the potential molecular targets of therapeutic intervention, the therapeutic weapons to modulate the activity of these targets and the few examples of therapeutic intervention on apoptosis, with emphasis in acute renal failure.


Subject(s)
Kidney Tubules/cytology , Acute Kidney Injury/pathology , Apoptosis/physiology , Cell Survival , Endoplasmic Reticulum/physiology , Humans , Mitochondria/physiology , Receptors, Tumor Necrosis Factor/physiology , bcl-X Protein/physiology
4.
Cell Death Dis ; 7: e2305, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27441657

ABSTRACT

Vascular calcification (VC) is associated with increased cardiovascular mortality in aging, chronic kidney disease (CKD), type 2 diabetes mellitus (T2DM) and atherosclerosis. TNF-like weak inducer of apoptosis (TWEAK) recently emerged as a new biomarker for the diagnosis and prognosis of cardiovascular diseases. TWEAK binding to its functional receptor Fn14 was reported to promote several steps of atherosclerotic plaque progression. However, no information is currently available on the role of TWEAK/Fn14 on the development of medial calcification, which is highly prevalent in aging, CKD and T2DM. This study explored the involvement of TWEAK in human vascular smooth muscle cells (h-VSMCs) calcification in vitro. We report that TWEAK binding to Fn14 promotes inorganic phosphate-induced h-VSMCs calcification, favors h-VSMCs osteogenic transition, decreasing acta2 and myh11 and increasing bmp2 mRNA and tissue non-specific alkaline phosphatase (TNAP), and increases MMP9 activity. Blockade of the canonical NFκB pathway reduced by 80% TWEAK pro-calcific properties and decreased osteogenic transition, TNAP and MMP9 activity. Blockade of non-canonical NFκB signaling by a siRNA targeting RelB reduced by 20% TWEAK pro-calcific effects and decreased TWEAK-induced loss of h-VSMCs contractile phenotype and MMP9 activity, without modulating bmp2 mRNA or TNAP activity. Inhibition of ERK1/2 activation by a MAPK kinase inhibitor did not influence TWEAK pro-calcific properties. Our results suggest that TWEAK/Fn14 directly favors inorganic phosphate-induced h-VSMCs calcification by activation of both canonical and non-canonical NFκB pathways. Given the availability of neutralizing anti-TWEAK strategies, our study sheds light on the TWEAK/Fn14 axis as a novel therapeutic target in the prevention of VC.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phosphates/pharmacology , Receptors, Tumor Necrosis Factor/genetics , Transcription Factor RelB/genetics , Tumor Necrosis Factors/genetics , Actins/genetics , Actins/metabolism , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cytokine TWEAK , Gene Expression Regulation , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Phosphates/metabolism , Primary Cell Culture , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , TWEAK Receptor , Transcription Factor RelB/antagonists & inhibitors , Transcription Factor RelB/metabolism , Tumor Necrosis Factors/metabolism , Vascular Calcification/genetics , Vascular Calcification/metabolism , Vascular Calcification/pathology
5.
Cell Death Dis ; 6: e1644, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25675304

ABSTRACT

Albuminuria promotes tubular injury and cell death, and is associated with faster progression of chronic kidney disease (CKD) to end-stage renal disease. However, the molecular mechanisms regulating tubular cell death in response to albuminuria are not fully understood. Brain abundant signal protein 1 (BASP1) was recently shown to mediate glucose-induced apoptosis in tubular cells. We have studied the role of BASP1 in albumin-induced tubular cell death. BASP1 expression was studied in experimental puromycin aminonucleoside-induced nephrotic syndrome in rats and in human nephrotic syndrome. The role of BASP1 in albumin-induced apoptosis was studied in cultured human HK2 proximal tubular epithelial cells. Puromycin aminonucleoside induced proteinuria and increased total kidney BASP1 mRNA and protein expression. Immunohistochemistry localized the increased BASP1 to tubular cells. BASP1 expression colocalized with deoxynucleotidyl-transferase-mediated dUTP nick-end labeling staining for apoptotic cells. Increased tubular BASP1 expression was observed in human proteinuric nephropathy by immunohistochemistry, providing evidence for potential clinical relevance. In cultured tubular cells, albumin induced apoptosis and increased BASP1 mRNA and protein expression at 6-48 h. Confocal microscopy localized the increased BASP1 expression in albumin-treated cells mainly to the perinuclear area. A peripheral location near the cell membrane was more conspicuous in albumin-treated apoptotic cells, where it colocalized with actin. Inhibition of BASP1 expression by a BASP1 siRNA protected from albumin-induced apoptosis. In conclusion, albumin-induced apoptosis in tubular cells is BASP1-dependent. This information may be used to design novel therapeutic approaches to slow CKD progression based on protection of tubular cells from the adverse consequences of albuminuria.


Subject(s)
Albumins/pharmacology , Calmodulin-Binding Proteins/metabolism , Cytoskeletal Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Calmodulin-Binding Proteins/genetics , Cell Line , Cytoskeletal Proteins/genetics , Female , Humans , Immunohistochemistry , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Male , Membrane Proteins/genetics , Microscopy, Confocal , Nerve Tissue Proteins/genetics , RNA, Small Interfering/genetics , Rats , Repressor Proteins/genetics
6.
Article in English | MEDLINE | ID: mdl-12476791

ABSTRACT

Cell number abnormalities are frequent in renal diseases, and range from the hypercellularity of postinfectious glomerulonephritis to the cell depletion of chronic renal atrophy. Recent research has shown that apoptosis and its regulatory mechanisms contribute to cell number regulation in the kidney. The potential role of apoptosis ranges from induction and progression to repair of renal injury. Death ligands and receptors, such as tumor necrosis factor and Fas ligand, proapoptotic and antiapoptotic Bcl2 family members and caspases have all been shown to participate in apoptosis regulation in the course of renal cell injury. However, the precise role of these proteins is unclear, and the participation of most known apoptosis regulatory proteins has not been studied. We now review the role of apoptosis in renal injury, the potential molecular targets of therapeutic intervention, the therapeutic weapons to modulate the activity of these targets and the few examples of therapeutic intervention on apoptosis, with emphasis on the acute tubular necrosis.


Subject(s)
Apoptosis , Kidney Diseases/pathology , Animals , Caspases/metabolism , Cytokines/metabolism , Humans , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/therapy , Kidney Tubular Necrosis, Acute/metabolism , Kidney Tubular Necrosis, Acute/pathology , Kidney Tubular Necrosis, Acute/therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Transcription Factors/metabolism
8.
Cytokine Growth Factor Rev ; 24(1): 23-40, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22959722

ABSTRACT

Macrophage migration inhibitory factor (MIF) is increased in kidney and urine during kidney disease. MIF binds to and activates CD74 and chemokine receptors CXCR2 and CXCR4. CD74 is a protein trafficking regulator and a cell membrane receptor for MIF, D-dopachrome tautomerase (D-DT/MIF-2) and bacterial proteins. MIF signaling through CD74 requires CD44. CD74, CD44 and CXCR4 are upregulated in renal cells in diseased kidneys and MIF activation of CD74 in kidney cells promotes an inflammatory response. MIF or CXCR2 targeting protects from experimental kidney injury, CD44 deficiency modulates kidney injury and CXCR4 activation promotes glomerular injury. However, the contribution of MIF or MIF-2 to these actions of MIF receptors has not been explored. The safety and efficacy of strategies targeting MIF, CD74, CD44 and CXCR4 are under study in humans.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/metabolism , Histocompatibility Antigens Class II/metabolism , Intramolecular Oxidoreductases/metabolism , Kidney Diseases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Animals , Humans , Hyaluronan Receptors/metabolism , Inflammation , Intramolecular Oxidoreductases/urine , Macrophage Migration-Inhibitory Factors/urine , Mice , Receptors, CXCR4/metabolism , Receptors, Cell Surface/metabolism , Receptors, Interleukin-8B/metabolism
9.
Kidney Int ; 69(12): 2205-11, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16688118

ABSTRACT

Fas-associated death domain (FADD) is an adaptor protein that is required for the transmission of the death signal from lethal receptors of the tumor necrosis factor superfamily. FADD contains a death domain (DD) and a death effector domain (DED). As death receptors contribute to renal tubular injury and tubular cell FADD increases in acute renal failure, we have studied the function of FADD in tubular epithelium. FADD expression was studied in kidney samples from mice. In order to study the contribution of FADD to renal tubular cell survival, FADD or FADD-DD were overexpressed in murine tubular epithelium. FADD is expressed in renal tubules of the healthy kidney. Both FADD and FADD-DD induce apoptosis in primary cultures of murine tubular epithelium and in the murine cortical tubular cell line. Death induced by FADD-DD has apoptotic morphology, but differs from death receptor-induced apoptosis in that it is not blocked by inhibitors of caspases. Neither an inhibitor of serine proteases nor overexpression of antiapoptotic BclxL prevented cell death. However, the combination of caspase and serine protease inhibition was protective. FADD and FADD-DD overexpression decreased nuclear factor kappa B activity. These data suggest that FADD has a death regulatory function in renal tubular cells that is independent of death receptors. FADD-DD is sufficient to induce apoptosis in these cells. This information is relevant to understanding the role of FADD in tubular injury.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis/physiology , Epithelial Cells/chemistry , Kidney Tubules/chemistry , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/genetics , Caspase Inhibitors , Caspases/metabolism , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/physiology , Fas-Associated Death Domain Protein , Gene Expression Regulation , Kidney Tubules/drug effects , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Mice , Mice, Inbred Strains , NF-kappa B/physiology , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Receptors, Tumor Necrosis Factor/physiology , Serine Proteinase Inhibitors
10.
Kidney Int ; 70(10): 1750-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17003819

ABSTRACT

Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK, TNFSF12) is a member of the TNF superfamily. TWEAK activates the Fn14 receptor, and may regulate apoptosis, proliferation, and inflammation, processes that play a significant role in pathological conditions. However, there is little information on the function and regulation of this system in the kidney. Therefore, TWEAK and Fn14 expression were studied in cultured murine tubular epithelial MCT cells and in mice in vivo. The effect of TWEAK on cell death was determined. We found that TWEAK and Fn14 expression was increased in experimental acute renal failure induced by folic acid. Cultured tubular cells express both TWEAK and the Fn14 receptor. TWEAK did not induce cell death in non-stimulated tubular cells. However, in cells costimulated with TNFalpha/interferon-gamma, TWEAK induced apoptosis through the activation of the Fn14 receptor. Apoptosis was associated with activation of caspase-8, caspase-9, and caspase-3, Bid cleavage, and evidence of mitochondrial injury. There was no evidence of endoplasmic reticulum stress. A pan-caspase inhibitor benzyloxycarbonyl-Val-Ala-DL-Asp prevented TWEAK-induced apoptosis, but it sensitized cells to necrosis via generation of reactive oxygen species. In conclusion, cooperation between inflammatory cytokines results in tubular cell death. TWEAK and Fn14 may play a role in renal tubular cell injury.


Subject(s)
Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factors/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspases/genetics , Caspases/metabolism , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Cytokine TWEAK , Cytokines , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Folic Acid , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Interferon-gamma/pharmacology , Kidney Tubules, Proximal/drug effects , Mice , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor/genetics , TWEAK Receptor , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL