Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Am J Pathol ; 183(1): 119-30, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23707238

ABSTRACT

In nondiabetic rat models of renal disease, angiotensin II (Ang II) perpetuates podocyte injury and promotes progression to end-stage kidney disease. Herein, we wanted to explore the role of Ang II in diabetic nephropathy by a translational approach spanning from in vitro to in vivo rat and human studies, and to dissect the intracellular pathways involved. In isolated perfused rat kidneys and in cultured human podocytes, Ang II down-regulated nephrin expression via Notch1 activation and nuclear translocation of Snail. Hairy enhancer of split-1 was a Notch1-downstream gene effector that activated Snail in cultured podocytes. In vitro changes of the Snail/nephrin axis were similar to those in renal biopsy specimens of Zucker diabetic fatty rats and patients with advanced diabetic nephropathy, and were normalized by pharmacological inhibition of the renin-angiotensin system. Collectively, the present studies provide evidence that Ang II plays a relevant role in perpetuating glomerular injury in experimental and human diabetic nephropathy via persistent activation of Notch1 and Snail signaling in podocytes, eventually resulting in down-regulation of nephrin expression, the integrity of which is crucial for the glomerular filtration barrier.


Subject(s)
Angiotensin II/metabolism , Diabetic Nephropathies/metabolism , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Transcription Factors/metabolism , Aged , Animals , Case-Control Studies , Cells, Cultured , Diabetes Mellitus, Type 2/complications , Down-Regulation , Female , Humans , Immunohistochemistry , Kidney/metabolism , Kidney/pathology , Linear Models , Male , Microscopy, Electron, Transmission , Middle Aged , Rats , Rats, Sprague-Dawley , Rats, Zucker , Real-Time Polymerase Chain Reaction , Snail Family Transcription Factors
2.
Am J Physiol Renal Physiol ; 304(6): F808-19, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23136004

ABSTRACT

Bardoxolone methyl is an antioxidant inflammation modulator acting through induction of Keap1-Nrf2 pathway. Results from a recent phase IIb clinical trial reported that bardoxolone methyl was associated with improvement in the estimated glomerular filtration rate in patients with advanced chronic kidney disease and Type 2 diabetes. However, increases in albuminuria, serum transaminase, and frequency of adverse events were noted. We studied the effect of 3-mo treatment with RTA 405, a synthetic triterpenoid analog of bardoxolone methyl in Zucker diabetic fatty rats with overt Type 2 diabetes. Rats were treated from 3 mo of age with vehicle, RTA 405, ramipril, or RTA 405 plus ramipril. RTA 405 caused severe changes in food intake and diuresis with decline in body weight, worsening of dyslipidemia, and increase in blood pressure. Early elevation in serum transaminase was followed by liver injury. RTA 405 worsened proteinuria, glomerulosclerosis, and tubular damage. Ramipril was renoprotective, but when given with RTA 405 it was not able to limit its worsening effects. These data could be due to degradation products in the drug substance used, as disclosed by the company once the study was concluded. To overcome such a drawback, the company offered to test dh404, a variant of RTA 405, in Zucker diabetic fatty rats. The dh404 did not display beneficial effects on proteinuria, glomerulosclerosis, and interstitial inflammation. Rather, kidneys from three rats receiving dh404 showed the presence of a granulomatous and inflammatory process reminiscent of a pseudotumor. Altogether these data raise serious concerns on the use of bardoxolone analogs in Type 2 diabetic nephropathy.


Subject(s)
Diabetic Nephropathies/drug therapy , Kidney/drug effects , Oleanolic Acid/analogs & derivatives , Triterpenes/adverse effects , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Blood Pressure/drug effects , Body Weight/drug effects , Chromatography, Liquid , Diabetic Nephropathies/pathology , Disease Models, Animal , Diuresis/drug effects , Drinking/drug effects , Hemodynamics/drug effects , Kidney/pathology , Liver/drug effects , Liver/pathology , Male , Mass Spectrometry , Oleanolic Acid/adverse effects , Ramipril/therapeutic use , Rats , Rats, Zucker , Renal Circulation/drug effects
3.
Am J Physiol Renal Physiol ; 303(9): F1370-81, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22952284

ABSTRACT

We previously reported that in a model of spontaneously progressive glomerular injury with early podocyte loss, abnormal migration, and proliferation of glomerular parietal epithelial progenitor cells contributed to the formation of synechiae and crescentic lesions. Here we first investigated whether a similar sequence of events could be extended to rats with adriamycin (ADR)-induced nephropathy. As a second aim, the regenerative potential of therapy with bone marrow-derived mesenchymal stem cells (MSCs) on glomerular resident cells was evaluated. In ADR-treated rats, decrease of WT1(+) podocyte number due to apoptosis was associated with reduced glomerular expression of nephrin and CD2AP. As a consequence of podocyte injury, glomerular adhesions of the capillary tuft to the Bowman's capsule were observed, followed by crescent-like lesions and glomerulosclerosis. Cellular components of synechiae were either NCAM(+) parietal progenitor cells or nestin(+) podocytes. In ADR rats, repeated injections of MSCs limited podocyte loss and apoptosis and partially preserved nephrin and CD2AP. MSCs attenuated the formation of glomerular podocyte-parietal epithelial cell bridges and normalized the distribution of NCAM(+) progenitor cells along the Bowman's capsule, thereby reducing glomerulosclerosis. Finding that MSCs increased glomerular VEGF expression and limited microvascular rarefaction may explain the prosurvival effect by stem cell therapy. MSCs also displayed anti-inflammatory activity. Coculture of MSCs with ADR-damaged podocytes showed a functional role of stem cell-derived VEGF on prosurvival pathways. These data suggest that MSCs by virtue of their tropism for damaged kidney and ability to provide a local prosurvival environment may represent a useful strategy to preserve podocyte viability and reduce glomerular inflammation and sclerosis.


Subject(s)
Kidney Diseases/pathology , Kidney Diseases/physiopathology , Kidney/physiology , Mesenchymal Stem Cell Transplantation , Podocytes/physiology , Regeneration/physiology , Stem Cells/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis/physiology , Cell Count , Cell Movement/physiology , Coculture Techniques , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Doxorubicin/adverse effects , Kidney/pathology , Kidney Diseases/chemically induced , Male , Membrane Proteins/metabolism , Podocytes/pathology , Rats , Rats, Inbred Lew , Stem Cells/pathology , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
4.
Am J Physiol Renal Physiol ; 301(5): F1114-23, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21816757

ABSTRACT

Diabetic nephropathy is associated with cardiovascular morbidity. Angiotensin-converting enzyme (ACE) inhibitors provide imperfect renoprotection in advanced type 2 diabetes, and cardiovascular risk remains elevated. Endothelin (ET)-1 has a role in renal and cardiac dysfunction in diabetes. Here, we assessed whether combination therapy with an ACE inhibitor and ET(A) receptor antagonist provided reno- and cardioprotection in rats with overt type 2 diabetes. Four groups of Zucker diabetic fatty (ZDF) rats were treated orally from 4 (when proteinuric) to 8 mo with vehicle, ramipril (1 mg/kg), sitaxsentan (60 mg/kg), and ramipril plus sitaxsentan. Lean rats served as controls. Combined therapy ameliorated proteinuria and glomerulosclerosis mostly as a result of the action of ramipril. Simultaneous blockade of ANG II and ET-1 pathways normalized renal monocyte chemoattractant protein-1 and interstitial inflammation. Cardiomyocyte loss, volume enlargement, and capillary rarefaction were prominent abnormalities of ZDF myocardium. Myocyte volume was reduced by ramipril and sitaxsentan, which also ameliorated heart capillary density. Drug combination restored myocardial structure and reestablished an adequate capillary network in the presence of increased cardiac expression of VEGF/VEGFR-1, and significant reduction of oxidative stress. In conclusion, in type 2 diabetes concomitant blockade of ANG II synthesis and ET-1 biological activity through an ET(A) receptor antagonist led to substantial albeit not complete renoprotection, almost due to the ACE inhibitor. The drug combination also showed cardioprotective properties, which however, were mainly dependent on the contribution of the ET(A) receptor antagonist through the action of VEGF.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Diabetes Mellitus, Type 2/pathology , Endothelin A Receptor Antagonists , Heart/drug effects , Kidney/pathology , Myocardium/pathology , Animals , Blood Glucose/metabolism , Body Weight/physiology , Cell Count , Collagen Type III/metabolism , Diabetes Mellitus, Type 2/genetics , Endothelin-1/metabolism , Hemodynamics/drug effects , Immunohistochemistry , Kidney/drug effects , Kidney/metabolism , Kidney Function Tests , Male , Muscle Cells/drug effects , Myocardium/metabolism , Rats , Rats, Zucker , Real-Time Polymerase Chain Reaction , Receptors, Vascular Endothelial Growth Factor/metabolism , Survival , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
Am J Physiol Renal Physiol ; 300(6): F1291-300, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21454255

ABSTRACT

Despite the central role of tubular plasma proteins that characterize progressive kidney diseases, protein concentrations along the nephron in pathological conditions have not been quantified so far. We combined experimental techniques and theoretical analysis to estimate glomerular and tubular levels of albumin in the experimental model of 5/6 nephrectomy (Nx) in the rat, with or without angiotensin-converting enzyme (ACE) inhibition. We measured glomerular permselectivity by clearance of fluorescent Ficoll and albumin and used theoretical analysis to estimate tubular albumin. As expected, 5/6 Nx induced an elevation of the fractional clearance of the largest Ficoll molecules (radii >56 Å, P < 0.05), increasing the importance of the shunt pathway of the glomerular membrane and the albumin excretion rate (119 ± 41 vs. 0.6 ± 0.2 mg/24 h, P < 0.01). ACE inhibition normalized glomerular permselectivity and urinary albumin (0.5 ± 0.3 mg/24 h). Theoretical analysis indicates that with 5/6 Nx, an increased albumin filtration overcomes proximal tubule reabsorption, with a massive increase in average albumin concentration along the tubule, reaching the highest value of >2,500 µg/ml at the end of the collecting duct. ACE inhibition improved glomerular permselectivity, limiting albumin filtration under proximal tubule reabsorption capacity, with low albumin concentration along the entire nephron, averaging <13 µg/ml at the end of the collecting duct. These results reinforce our understanding of the mechanisms of renal disease progression and the effects of angiotensin II antagonism. They also suggest that evaluation of tubular protein concentration levels could help to identify patients at risk of kidney disease progression and to improve clinical management.


Subject(s)
Albumins/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Kidney Diseases/metabolism , Kidney Glomerulus/drug effects , Albuminuria/metabolism , Albuminuria/pathology , Analysis of Variance , Animals , Glomerular Filtration Rate/drug effects , Kidney Diseases/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Nephrectomy , Proteinuria/metabolism , Proteinuria/pathology , Rats , Rats, Sprague-Dawley
6.
Am J Physiol Renal Physiol ; 299(5): F1203-11, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20719975

ABSTRACT

The capacity of renin-angiotensin system (RAS) inhibitors to delay progression of diabetic nephropathy depends on the time at which therapy is started. A multimodal intervention is required to afford renoprotection in overt diabetic nephropathy. Here we assessed the effects of maximal RAS inhibition by angiotensin-converting enzyme (ACE) inhibitor plus angiotensin II type 1 receptor blocker (ARB) in combination with statin in rats with overt diabetic nephropathy. Uninephrectomized rats made diabetic by streptozotocin were orally treated from 4 (when proteinuria and renal lesions had developed) to 8 mo with vehicle, lisinopril plus candesartan, lisinopril plus candesartan plus rosuvastatin, or rosuvastatin alone. Systolic blood pressure increased in diabetic rats and was significantly lowered by combined therapies. Dual RAS blockade significantly reduced proteinuria compared with vehicle. Addition of statin further lowered proteinuria to control levels. Glomerulosclerosis was ameliorated by RAS inhibitors or statin, and regression was achieved by the addition of statin. Loss of podocytes of diabetic rats was limited by ACE inhibitor plus ARB while normalized by the three drugs. Defective nephrin expression of diabetes was increased by dual RAS blockade or statin and restored by the triple therapy. Tubular damage, interstitial inflammation, and expression of the fibrotic markers transforming growth factor (TGF)-ß1 and phosphorylated Smad 2/3 in tubuli were significantly reduced by the triple regimen. These data suggest a strategy to target proteinuria to try to achieve regression of renal disease in diabetic patients who do not fully benefit from RAS inhibition alone.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Diabetic Nephropathies/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Proteinuria/drug therapy , Animals , Blood Pressure/drug effects , Cholesterol/blood , Cholesterol Esters/blood , Diabetic Nephropathies/urine , Fibrosis , Immunohistochemistry , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Function Tests , Kidney Glomerulus/pathology , Lipid Metabolism/drug effects , Male , Membrane Proteins/biosynthesis , Nephritis, Interstitial/pathology , Podocytes/pathology , Proteinuria/etiology , Rats , Rats, Sprague-Dawley , Renin-Angiotensin System/drug effects
7.
Pediatr Nephrol ; 25(11): 2231-40, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20424866

ABSTRACT

Shiga toxin (Stx)-producing enterohemorrhagic Escherichia coli O157:H7 has become a global threat to public health, as a primary cause of a worldwide spread of hemorrhagic colitis complicated by diarrhea-associated hemolytic uremic syndrome (HUS), a disorder of thrombocytopenia, microangiopathic hemolytic anemia, and acute renal failure that mainly affects early childhood. Endothelial dysfunction has been recognized as the trigger event in the development of microangiopathic processes. Endothelial cells, mainly those located in the renal microvasculature, are primary targets of the toxic effects of Stx1 and 2. Stxs bound to their specific globotriaosylceramide (Gb3Cer) receptor on the cell surface trigger a cascade of signaling events, involving NF-κB activation, that induce expression of genes encoding for adhesion molecules and chemokines, and culminate in the adhesion of leukocytes to endothelial cells, thereby increasing the endothelial susceptibility to leukocyte-mediated injury. Activated endothelial cells in response to Stxs lose the normal thromboresistance phenotype and become thrombogenic, initiating microvascular thrombus formation. Evidence is emerging that complement activation in response to Stxs favors platelet thrombus formation on endothelial cells, which may play a role in amplifying the inflammation-thrombosis circuit in Stx-associated HUS.


Subject(s)
Endothelium, Vascular/physiopathology , Hemolytic-Uremic Syndrome/metabolism , Shiga Toxin/metabolism , Acute Kidney Injury/metabolism , Capillaries/metabolism , Chemokines/metabolism , Child, Preschool , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Escherichia coli O157/metabolism , Hemolytic-Uremic Syndrome/etiology , Hemolytic-Uremic Syndrome/physiopathology , Humans , Leukocytes/metabolism , Models, Biological , NF-kappa B/metabolism , Shiga Toxin/pharmacology , Trihexosylceramides/metabolism
8.
Stem Cells ; 28(3): 513-22, 2010 Mar 31.
Article in English | MEDLINE | ID: mdl-20049901

ABSTRACT

In search for new sources of mesenchymal stem cells (MSCs) for renal repair in acute kidney injury (AKI), we investigated the potential of human cord blood (CB)-MSCs to cure mice with AKI. Infusion of CB-MSCs in immunodeficient mice with cisplatin-induced AKI ameliorated both renal function and tubular cell injury, and prolonged survival. Transplanted CB-MSCs localized in peritubular areas, limited capillary alterations and neutrophil infiltration. Apoptosis reduced and tubular cell proliferation increased by virtue of stem cell capacity to produce growth factors. The reno-protective effect of CB-MSCs was further confirmed by their ability to inhibit oxidative damage and to induce the prosurvival factor Akt in tubular cells. The evidence that CB-MSCs in vitro increased the production of growth factors and inhibited IL-1 beta and TNFalpha synthesis when cocultured with damaged proximal tubular cells indicates a regenerative and anti-inflammatory action of stem cell treatment. Altogether these results highlight the potential of human CB-MSCs as future cell therapy for testing in human AKI.


Subject(s)
Cell Differentiation/physiology , Cord Blood Stem Cell Transplantation/methods , Graft Survival/physiology , Kidney Diseases/surgery , Kidney/surgery , Mesenchymal Stem Cell Transplantation/methods , Acute Disease , Animals , Anti-Inflammatory Agents/metabolism , Apoptosis/physiology , Cell Culture Techniques , Cell Proliferation , Coculture Techniques , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/physiopathology , Kidney Tubules/cytology , Kidney Tubules/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, SCID , Oxidative Stress/physiology , Proto-Oncogene Proteins c-akt/metabolism
9.
Am J Physiol Renal Physiol ; 297(5): F1448-56, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19675181

ABSTRACT

In the present study, we evaluated the effect of simultaneously blocking angiotensin II synthesis and endothelin (ET)-1 activity as a multimodal intervention to implement renoprotection in overt diabetic nephropathy. Mechanisms underlying combined therapy effectiveness were addressed by investigating podocyte structure and function and glomerular barrier size-selective properties. Uninephrectomized rats made diabetic by streptozotocin received orally placebo, lisinopril (12.5 mg/l), the ET(A) receptor antagonist avosentan (30 mg/kg), or their combination from 4 (when animals had proteinuria) to 8 mo. Proteinuria, renal damage, podocyte number, nephrin expression, and glomerular size selectivity by graded-size Ficoll molecule fractional clearance were assessed. Combined therapy normalized proteinuria, provided complete protection from tubulointerstitial damage, and induced regression of glomerular lesions, while only a partial renoprotection was achieved by each drug alone. Lisinopril plus avosentan restored to normal values the number of podocytes. Single therapies only limited podocyte depletion. Defective nephrin expression of diabetes was prevented by each drug. Altered glomerular size selectivity to large macromolecules of diabetic rats was remarkably improved by lisinopril and the combined treatment. Avosentan ameliorated peritubular capillary architecture and reduced interstitial inflammation and fibrosis. The ACE inhibitor and ET(A) receptor antagonist induced regression of glomerular lesions in overt diabetic nephropathy. Regression of renal disease was conceivably the result of the synergistic effect of the ACE inhibitor of preserving glomerular permselective properties and the ET(A) antagonist in improving tubulointerstitial changes. These findings provide mechanistic insights to explain the antiproteinuric effect of this combined therapy in diabetes.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Diabetic Nephropathies/prevention & control , Lisinopril/therapeutic use , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Animals , Blood Pressure/drug effects , Capillaries/pathology , Cell Count , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/mortality , Diabetic Nephropathies/pathology , Drug Therapy, Combination , Endothelin A Receptor Antagonists , Immunohistochemistry , Kidney/pathology , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Lipids/blood , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Podocytes/drug effects , Podocytes/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
10.
N Engl J Med ; 361(4): 345-57, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19625716

ABSTRACT

BACKGROUND: The hemolytic-uremic syndrome consists of the triad of microangiopathic hemolytic anemia, thrombocytopenia, and renal failure. The common form of the syndrome is triggered by infection with Shiga toxin-producing bacteria and has a favorable outcome. The less common form of the syndrome, called atypical hemolytic-uremic syndrome, accounts for about 10% of cases, and patients with this form of the syndrome have a poor prognosis. Approximately half of the patients with atypical hemolytic-uremic syndrome have mutations in genes that regulate the complement system. Genetic factors in the remaining cases are unknown. We studied the role of thrombomodulin, an endothelial glycoprotein with anticoagulant, antiinflammatory, and cytoprotective properties, in atypical hemolytic-uremic syndrome. METHODS: We sequenced the entire thrombomodulin gene (THBD) in 152 patients with atypical hemolytic-uremic syndrome and in 380 controls. Using purified proteins and cell-expression systems, we investigated whether thrombomodulin regulates the complement system, and we characterized the mechanisms. We evaluated the effects of thrombomodulin missense mutations associated with atypical hemolytic-uremic syndrome on complement activation by expressing thrombomodulin variants in cultured cells. RESULTS: Of 152 patients with atypical hemolytic-uremic syndrome, 7 unrelated patients had six different heterozygous missense THBD mutations. In vitro, thrombomodulin binds to C3b and factor H (CFH) and negatively regulates complement by accelerating factor I-mediated inactivation of C3b in the presence of cofactors, CFH or C4b binding protein. By promoting activation of the plasma procarboxypeptidase B, thrombomodulin also accelerates the inactivation of anaphylatoxins C3a and C5a. Cultured cells expressing thrombomodulin variants associated with atypical hemolytic-uremic syndrome had diminished capacity to inactivate C3b and to activate procarboxypeptidase B and were thus less protected from activated complement. CONCLUSIONS: Mutations that impair the function of thrombomodulin occur in about 5% of patients with atypical hemolytic-uremic syndrome.


Subject(s)
Complement Activation/genetics , Hemolytic-Uremic Syndrome/genetics , Mutation, Missense , Thrombomodulin/genetics , Adolescent , Adult , Child , Complement C3b , Complement Factor I , Complement Pathway, Alternative/physiology , DNA Mutational Analysis , Hemolytic-Uremic Syndrome/immunology , Heterozygote , Humans , Middle Aged , Pedigree , Polymorphism, Single Nucleotide , Thrombomodulin/metabolism , Young Adult
11.
Kidney Int ; 76(9): 960-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19625993

ABSTRACT

Blockade of the renin-angiotensin system (RAS), the standard treatment for chronic proteinuric nephropathy, slows but may not halt progression of the disease, particularly when therapy is started late. Because vasopressin may also play a role in the progression of renal disease, we measured the effect of a dual V(1a) and V(2) vasopressin receptor antagonist (RWJ-676070) alone or combined with angiotensin-converting enzyme inhibition or angiotensin II type 1 receptor blockade on proteinuria and renal disease progression during overt nephropathy. Twenty-one days after renal mass reduction, a time of established injury, rats were given vehicle, RWJ-676070, enalapril, losartan, RWJ-676070 plus enalapril, or losartan in drinking water for an additional 39 days. RWJ-676070 returned the blood pressure to pre-treatment levels, which were significantly lower than those in vehicle-treated rats. Enalapril, losartan, and the combined therapies reduced blood pressure to a greater extent. RWJ-676070 afforded a partial antiproteinuric effect, which was enhanced by the addition of enalapril or losartan. Renal functional impairment, and glomerular and tubular changes were partially ameliorated by RWJ-676070; parameters significantly improved with either enalapril or losartan alone and improved to a greater extent with the combined therapies. Our findings suggest that vasopressin receptor antagonists could be of additional therapeutic value in the treatment of chronic proteinuric nephropathy.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Antidiuretic Hormone Receptor Antagonists , Benzazepines/pharmacology , Enalapril/pharmacology , Hormone Antagonists/pharmacology , Kidney Diseases/drug therapy , Kidney/drug effects , Losartan/pharmacology , Renin-Angiotensin System/drug effects , Spiro Compounds/pharmacology , Animals , Benzazepines/blood , Benzazepines/pharmacokinetics , Biomarkers/blood , Biomarkers/urine , Blood Pressure/drug effects , Body Weight/drug effects , Chronic Disease , Disease Models, Animal , Disease Progression , Diuresis/drug effects , Drinking/drug effects , Drug Therapy, Combination , Eating/drug effects , Hormone Antagonists/blood , Hormone Antagonists/pharmacokinetics , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Male , Nephrectomy , Proteinuria/drug therapy , Proteinuria/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Vasopressin/metabolism , Spiro Compounds/blood , Spiro Compounds/pharmacokinetics , Time Factors
12.
Front Biosci (Landmark Ed) ; 14(5): 1815-22, 2009 01 01.
Article in English | MEDLINE | ID: mdl-19273165

ABSTRACT

Tubulointerstitial damage followed by scarring and progressive loss of renal function is common to many forms of chronic proteinuric nephropathies. The severity of tubulointerstitial injury and in particular interstitial macrophage infiltration strongly correlate with the risk of renal failure. Proteins filtered through the glomerular capillary in excessive amount activate proximal tubular cells to upregulate chemokines mainly via activation of NF-kappaB-dependent pathway. Chemokines secreted toward the basolateral compartment of tubular epithelial cells incite local recruitment of mononuclear cells, that in turn interact with resident renal cells and extracellular matrix to create a proinflammatory microenvironment that amplifies tubulointerstitial inflammation and promotes renal scarring. The association between proteinuria and interstitial accumulation of inflammatory cells via activation of transcription factors and overexpression of chemokines has been established both experimentally and in human proteinuric nephropathies. Blocking leukocyte recruitment by interfering with transcription factor activity or chemokines and their receptors is envisioned as a strategy to retard kidney disease progression.


Subject(s)
Chemokines/physiology , Kidney Diseases/physiopathology , Chemokines/genetics , Chemokines/metabolism , Dietary Proteins/administration & dosage , Disease Progression , Humans , Kidney Diseases/metabolism , Kidney Tubules, Proximal/metabolism , Proteinuria/metabolism , Proteinuria/physiopathology , Up-Regulation
13.
J Clin Invest ; 119(3): 524-30, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19197138

ABSTRACT

The renin-angiotensin system plays a role in the etiology of hypertension and the pathophysiology of cardiac and renal diseases in humans. Ang II is the central product of this system and is involved in regulating immune responses, inflammation, cell growth, and proliferation by acting through Ang II type 1 receptors (AT1 and AT2). Here, we show that targeted disruption of the Agtr1a gene that encodes AT1A results in marked prolongation of life span in mice. Agtr1a-/- mice developed less cardiac and vascular injury, and multiple organs from these mice displayed less oxidative damage than wild-type mice. The longevity phenotype was associated with an increased number of mitochondria and upregulation of the prosurvival genes nicotinamide phosphoribosyltransferase (Nampt) and sirtuin 3 (Sirt3) in the kidney. In cultured tubular epithelial cells, Ang II downregulated Sirt3 mRNA, and this effect was inhibited by an AT1 antagonist. These results demonstrate that disruption of AT1 promotes longevity in mice, possibly through the attenuation of oxidative stress and overexpression of prosurvival genes, and suggests that the Ang II/AT1 pathway may be targeted to influence life span in mammals.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Longevity/genetics , Mice, Knockout/genetics , Animals , Blood Glucose , Body Weight , Cytokines/genetics , Down-Regulation , Energy Intake , Heart Diseases/genetics , Heart Diseases/prevention & control , Mice , Mitochondrial Proteins/genetics , Nicotinamide Phosphoribosyltransferase/genetics , Oxidative Stress/genetics , Phenotype , Rotarod Performance Test , Sirtuin 3 , Sirtuins/genetics , Up-Regulation , Vascular Diseases/genetics , Vascular Diseases/prevention & control
14.
Kidney Int ; 75(10): 1050-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19242507

ABSTRACT

Intrarenal complement activation plays an important role in the progression of chronic kidney disease. A key target of the activated complement cascade is the proximal tubule, a site where abnormally filtered plasma proteins and complement factors combine to promote injury. This study determined whether protein overloading of human proximal tubular cells (HK-2) in culture enhances complement activation by impairing complement regulation. Addition of albumin or transferrin to the cells incubated with diluted human serum as a source of complement caused increased apical C3 deposition. Soluble complement receptor-1 (an inhibitor of all 3 activation pathways) blocked complement deposition while the classical and lectin pathway inhibitor, magnesium chloride-EGTA, was, ineffective. Media containing albumin as well as complement had additive proinflammatory effects as shown by increased fractalkine and transforming growth factor-beta mRNA expression. This paralleled active C3 and C5b-9 generations, effects not shared by transferrin. Factor H, one of the main natural inhibitors of the alternative pathway, binds to heparan sulfate proteoglycans. Both the density of heparan sulfate and factor H binding were reduced with protein loading, thereby enhancing the albumin- and serum-dependent complement activation potential. Thus, protein overload reduces the ability of the tubule cell to bind factor H and counteract complement activation, effects instrumental to renal disease progression.


Subject(s)
Complement Factor H/metabolism , Complement System Proteins/metabolism , Kidney Tubules, Proximal/pathology , Proteins/pharmacology , Cell Line , Complement Activation , Disease Progression , Humans , Kidney Diseases/etiology , Kidney Diseases/pathology , Protein Binding , Serum Albumin/pharmacology , Transferrin/pharmacology
15.
J Am Soc Nephrol ; 20(1): 123-30, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19092126

ABSTRACT

The role of dendritic cells (DC) that accumulate in the renal parenchyma of non-immune-mediated proteinuric nephropathies is not well understood. Under certain circumstances, DC capture immunologically ignored antigens, including self-antigens, and present them within MHC class I, initiating an autoimmune response. We studied whether DC could generate antigenic peptides from the self-protein albumin. Exposure of rat proximal tubular cells to autologous albumin resulted in its proteolytic cleavage to form an N-terminal 24-amino acid peptide (ALB1-24). This peptide was further processed by the DC proteasome into antigenic peptides that had binding motifs for MHC class I and were capable of activating syngeneic CD8+ T cells. In vivo, the rat five-sixths nephrectomy model allowed the localization and activation of renal DC. Accumulation of DC in the renal parenchyma peaked 1 wk after surgery and decreased at 4 wk, concomitant with their appearance in the renal draining lymph nodes. DC from renal lymph nodes, loaded with ALB1-24, activated syngeneic CD8+ T cells in primary culture. The response of CD8+ T cells of five-sixths nephrectomized rats was amplified with secondary stimulation. In contrast, DC from renal lymph nodes of five-sixths nephrectomized rats treated with the proteasomal inhibitor bortezomib lost their capacity to stimulate CD8+ T cells in primary and secondary cultures. These data suggest that albumin can be a source of potentially antigenic peptides upon renal injury and that renal DC play a role in processing self-proteins through a proteasome-dependent pathway.


Subject(s)
Albumins/metabolism , Antigen Presentation , Dendritic Cells/physiology , Kidney/immunology , Proteasome Endopeptidase Complex/physiology , Animals , CD11c Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/metabolism , Immune Tolerance , Kidney Tubules, Proximal/metabolism , Proteasome Inhibitors , Proteinuria/immunology , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
16.
J Immunol ; 181(2): 1460-9, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18606701

ABSTRACT

Shiga toxins (Stx) are the virulence factors of enterohemorrhagic Escherichia coli O157:H7, a worldwide emerging diarrheal pathogen, which precipitates postdiarrheal hemolytic uremic syndrome, the leading cause of acute renal failure in children. In this study, we show that Stx2 triggered expression of fractalkine (FKN), a CX3C transmembrane chemokine, acting as both adhesion counterreceptor on endothelial cells and soluble chemoattractant. Stx2 caused in HUVEC expression of FKN mRNA and protein, which promoted leukocyte capture, ablated by Abs to either endothelial FKN or leukocyte CX3CR1 receptor. Exposure of human glomerular endothelial cells to Stx2 recapitulated its FKN-inducing activity and FKN-mediated leukocyte adhesion. Both processes required phosphorylation of Src-family protein tyrosine kinase and p38 MAPK in endothelial cells. Furthermore, they depended on nuclear import of NF-kappaB and other stress-responsive transcription factors. Inhibition of their nuclear import with the cell-penetrating SN50 peptide reduced FKN mRNA levels and FKN-mediated leukocyte capture by endothelial cells. Adenoviral overexpression of IkappaBalpha inhibited FKN mRNA up-regulation. The FKN-mediated responses to Stx2 were also dependent on AP-1. In mice, both virulence factors of Stx-producing E. coli, Stx and LPS, are required to elicit hemolytic uremic syndrome. In this study, FKN was detected within glomeruli of C57BL/6 mice injected with Stx2, and further increased after Stx2 plus LPS coadministration. This was associated with recruitment of CX3CR1-positive cells. Thus, in response to Stx2, FKN is induced playing an essential role in the promotion of leukocyte-endothelial cell interaction thereby potentially contributing to the renal microvascular dysfunction and thrombotic microangiopathy that underlie hemolytic uremic syndrome due to enterohemorrhagic E. coli O157:H7 infection.


Subject(s)
Chemokine CX3CL1/metabolism , Endothelial Cells/immunology , Hemolytic-Uremic Syndrome/immunology , Kidney Glomerulus/immunology , Leukocytes/immunology , Receptors, Chemokine/metabolism , Shiga Toxin 2/immunology , Animals , CX3C Chemokine Receptor 1 , Cell Adhesion , Cells, Cultured , Chemokine CX3CL1/immunology , Disease Models, Animal , Endothelial Cells/metabolism , Escherichia coli O157/immunology , Hemolytic-Uremic Syndrome/metabolism , Hemolytic-Uremic Syndrome/microbiology , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/immunology , NF-kappa B/metabolism , Receptors, Chemokine/immunology , Shiga Toxin 2/metabolism , Signal Transduction , Transcription Factor AP-1/immunology , Transcription Factor AP-1/metabolism , Up-Regulation , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Stem Cells ; 26(8): 2075-82, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18499895

ABSTRACT

Transplantation of bone marrow mesenchymal stem cells (BM-MSC) or stromal cells from rodents has been identified as a strategy for renal repair in experimental models of acute kidney injury (AKI), a highly life-threatening clinical setting. The therapeutic potential of BM-MSC of human origin has not been reported so far. Here, we investigated whether human BM-MSC treatment could prevent AKI induced by cisplatin and prolong survival in an immunodeficient mouse model. Results showed that human BM-MSC infusion decreased proximal tubular epithelial cell injury and ameliorated the deficit in renal function, resulting in reduced recipient mortality. Infused BM-MSC became localized predominantly in peritubular areas and acted to reduce renal cell apoptosis and to increase proliferation. BM-MSC also induced protection against AKI-related peritubular capillary changes consisting of endothelial cell abnormalities, leukocyte infiltration, and low endothelial cell and lumen volume density as assessed by morphometric analysis. These findings indicate that human MSC of bone marrow origin hold potential to prolong survival in AKI and should be considered for testing in a clinical trial. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Bone Marrow Cells/cytology , Kidney/injuries , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Animals , Apoptosis , Body Weight , Cell Differentiation , Cell Proliferation , Cisplatin/pharmacology , Female , Humans , Kidney/metabolism , Kidney/pathology , Mice , Mice, SCID , Phenotype
18.
J Am Soc Nephrol ; 19(6): 1158-67, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18354030

ABSTRACT

Intrarenal complement activation leads to chronic tubulointerstitial injury in animal models of proteinuric nephropathies, making this process a potential target for therapy. This study investigated whether a C3-mediated pathway promotes renal injury in the protein overload model and whether the abnormal exposure of proximal tubular cells to filtered complement could trigger the resulting inflammatory response. Mice with C3 deficiency were protected to a significant degree against the protein overload-induced interstitial inflammatory response and tissue damage, and they had less severe podocyte injury and less proteinuria. When the same injury was induced in wild-type (WT) mice, antiproteinuric treatment with the angiotensin-converting enzyme inhibitor lisinopril reduced the amount of plasma protein filtered, decreased the accumulation of C3 by proximal tubular cells, and protected against interstitial inflammation and damage. For determination of the injurious role of plasma-derived C3, as opposed to tubular cell-derived C3, C3-deficient kidneys were transplanted into WT mice. Protein overload led to the development of glomerular injury, accumulation of C3 in podocytes and proximal tubules, and tubulointerstitial changes. Conversely, when WT kidneys were transplanted into C3-deficient mice, protein overload led to a more mild disease and abnormal C3 deposition was not observed. These data suggest that the presence of C3 increases the glomerular filtration barrier's susceptibility to injury, ultrafiltered C3 contributes more to tubulointerstitial damage induced by protein overload than locally synthesized C3, and local C3 synthesis is irrelevant to the development of proteinuria. It is speculated that therapies targeting complement combined with interventions to minimize proteinuria would more effectively prevent the progression of renal disease.


Subject(s)
Complement C3/deficiency , Renal Insufficiency/etiology , Animals , Complement C3/physiology , Disease Progression , Glomerular Filtration Rate , Male , Mice
19.
J Am Soc Nephrol ; 18(11): 2921-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17942965

ABSTRACT

In mice with cisplatin-induced acute kidney injury, administration of bone marrow-derived mesenchymal stem cells (MSC) restores renal tubular structure and improves renal function, but the underlying mechanism is unclear. Here, we examined the process of kidney cell repair in co-culture experiments with MSC and cisplatin-injured proximal tubular epithelial cells (PTEC). Exposure of PTEC to cisplatin markedly reduced cell viability at 4 days, but co-culture with MSC provided a protective effect by promoting tubular cell proliferation. This effect was mediated by insulin-like growth factor-1 (IGF-1), highly expressed by MSC as mRNA and protein, since blocking the growth factor's function with a specific antibody attenuated cell proliferation of PTEC. Confirming this, knocking down IGF-1 expression in MSC by small interfering-RNA also resulted in a significant decrease in PTEC proliferation and increased apoptosis. Furthermore, in the murine model of cisplatin-induced kidney injury, administering IGF-1 gene-silenced MSC limited their protective effect on renal function and tubular structure. These findings indicate that MSC exert beneficial effects on tubular cell repair in acute kidney injury by producing the mitogenic and pro-survival factor IGF-1.


Subject(s)
Epithelial Cells/physiology , Insulin-Like Growth Factor I/physiology , Kidney Diseases/therapy , Kidney Tubules, Proximal/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Animals , Cell Culture Techniques , Cell Proliferation , Cell Survival/physiology , Cisplatin , Coculture Techniques , Female , Male , Mice , Mice, Inbred C57BL
20.
Am J Nephrol ; 27(6): 630-8, 2007.
Article in English | MEDLINE | ID: mdl-17851231

ABSTRACT

BACKGROUND: Evidence is accumulating that statins can reduce proteinuria and disease progression in chronic kidney disease. However, some safety concerns have been recently raised on the use of these agents, mainly due to transient episodes of proteinuria observed in patients receiving high doses of rosuvastatin. METHODS: We investigated in rats with renal mass ablation (RMR) whether rosuvastatin (5 or 20 mg/day) worsens proteinuria as compared to untreated RMR animals. Moreover, we also examined whether rosuvastatin-induced changes in proteinuria would be due to the effect of the drug on permselective properties of glomerular capillary barrier, measured by the fractional clearance of graded-size Ficoll molecules and/or by proximal tubular mechanisms, by assessing urinary excretion of beta(2)-microglobulin. RESULTS: RMR rats given rosuvastatin for 28 days showed a progressive increase in proteinuria, with values numerically but not significantly higher than those in RMR animals given the vehicle. In RMR rats, rosuvastatin did not significantly affect the fractional clearance of Ficoll as compared to vehicle-treated RMR animals. A significant correlation was found between urinary protein and beta(2)-microglobulin excretion in rats treated with rosuvastatin (r = 0.936, p < 0.001), but not in those given vehicle. Renal function, glomerular and tubulointerstitial injury were comparable in rosuvastatin-treated and untreated RMR rats at the end of the 28-day follow-up. CONCLUSION: In rats with RMR, rosuvastatin mildly enhances urinary protein excretion rate. This, however, was not the result of further changes in the size-permselective function of glomerular capillary barrier.


Subject(s)
Capillaries/drug effects , Capillary Permeability/drug effects , Fluorobenzenes/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Kidney Glomerulus/blood supply , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Animals , Capillaries/physiopathology , Disease Models, Animal , Kidney Glomerulus/drug effects , Proteinuria/chemically induced , Proteinuria/physiopathology , Rats , Rats, Sprague-Dawley , Rosuvastatin Calcium
SELECTION OF CITATIONS
SEARCH DETAIL
...