Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Am J Physiol Renal Physiol ; 311(6): F1230-F1242, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27335372

ABSTRACT

Pericytes are tissue-resident mesenchymal progenitor cells anatomically associated with the vasculature that have been shown to participate in tissue regeneration. Here, we tested the hypothesis that kidney pericytes, derived from FoxD1+ mesodermal progenitors during embryogenesis, are necessary for postnatal kidney homeostasis. Diphtheria toxin delivery to FoxD1Cre::RsDTR transgenic mice resulted in selective ablation of >90% of kidney pericytes but not other cell lineages. Abrupt increases in plasma creatinine, blood urea nitrogen, and albuminuria within 96 h indicated acute kidney injury in pericyte-ablated mice. Loss of pericytes led to a rapid accumulation of neutral lipid vacuoles, swollen mitochondria, and apoptosis in tubular epithelial cells. Pericyte ablation led to endothelial cell swelling, reduced expression of vascular homeostasis markers, and peritubular capillary loss. Despite the observed injury, no signs of the acute inflammatory response were observed. Pathway enrichment analysis of genes expressed in kidney pericytes in vivo identified basement membrane proteins, angiogenic factors, and factors regulating vascular tone as major regulators of vascular function. Using novel microphysiological devices, we recapitulated human kidney peritubular capillaries coated with pericytes and showed that pericytes regulate permeability, basement membrane deposition, and microvascular tone. These findings suggest that through the active support of the microvasculature, pericytes are essential to adult kidney homeostasis.


Subject(s)
Acute Kidney Injury/metabolism , Capillaries/metabolism , Endothelium, Vascular/metabolism , Kidney/blood supply , Pericytes/metabolism , Animals , Kidney/metabolism , Mice , Mice, Transgenic , Microvessels/metabolism , Permeability
2.
Am J Physiol Cell Physiol ; 304(7): C591-603, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23325411

ABSTRACT

Chronic kidney disease, defined as loss of kidney function for more than three months, is characterized pathologically by glomerulosclerosis, interstitial fibrosis, tubular atrophy, peritubular capillary rarefaction, and inflammation. Recent studies have identified a previously poorly appreciated, yet extensive population of mesenchymal cells, called either pericytes when attached to peritubular capillaries or resident fibroblasts when embedded in matrix, as the progenitors of scar-forming cells known as myofibroblasts. In response to sustained kidney injury, pericytes detach from the vasculature and differentiate into myofibroblasts, a process not only causing fibrosis, but also directly contributing to capillary rarefaction and inflammation. The interrelationship of these three detrimental processes makes myofibroblasts and their pericyte progenitors an attractive target in chronic kidney disease. In this review, we describe current understanding of the mechanisms of pericyte-to-myofibroblast differentiation during chronic kidney disease, draw parallels with disease processes in the glomerulus, and highlight promising new therapeutic strategies that target pericytes or myofibroblasts. In addition, we describe the critical paracrine roles of epithelial, endothelial, and innate immune cells in the fibrogenic process.


Subject(s)
Fibrosis/pathology , Kidney/pathology , Renal Insufficiency, Chronic/pathology , Animals , Kidney/cytology , Mesenchymal Stem Cells/pathology , Myofibroblasts/pathology , Pericytes/pathology
3.
Blood ; 118(7): 1934-42, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21685370

ABSTRACT

Ischemia/reperfusion (I/R) injury in the kidney is a major cause of acute kidney injury (AKI) in humans and is associated with significantly high mortality. To identify genes that modulate kidney injury and repair, we conducted genome-wide expression analysis in the rat kidneys after I/R and found that the mRNA levels of fibrinogen (Fg)α, Fgß, and Fgγ chains significantly increase in the kidney and remain elevated throughout the regeneration process. Cellular characterization of Fgα and Fgγ chain immunoreactive proteins shows a predominant expression in renal tubular cells and the localization of immunoreactive Fgß chain protein is primarily in the renal interstitium in healthy and regenerating kidney. We also show that urinary excretion of Fg is massively increased after kidney damage and is capable of distinguishing human patients with acute or chronic kidney injury (n = 25) from healthy volunteers (n = 25) with high sensitivity and specificity (area under the receiver operating characteristic of 0.98). Furthermore, we demonstrate that Fgß-derived Bß(15-42) peptide administration protects mice from I/R-induced kidney injury by aiding in epithelial cell proliferation and tissue repair. Given that kidney regeneration is a major determinant of outcome for patients with kidney damage, these results provide new opportunities for the use of Fg in diagnosis, prevention, and therapeutic interventions in kidney disease.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/etiology , Fibrin Fibrinogen Degradation Products/therapeutic use , Peptide Fragments/therapeutic use , Reperfusion Injury/complications , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Aged , Amino Acid Sequence , Animals , Apoptosis/drug effects , Female , Fibrinogen/genetics , Fibrinogen/immunology , Fibrinogen/urine , Humans , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Molecular Sequence Data , RNA, Messenger/genetics , Rats , Rats, Wistar , Up-Regulation
4.
Kidney Int ; 82(2): 172-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22437410

ABSTRACT

Chronic kidney disease (CKD) remains one of the leading causes of death in the developed world, and acute kidney injury (AKI) is now recognized as a major risk factor in its development. Understanding the factors leading to CKD after acute injury are limited by current animal models of AKI, which concurrently target various kidney cell types including epithelial, endothelial, and inflammatory cells. Here, we developed a mouse model of kidney injury using the Six2-Cre-LoxP technology to selectively activate expression of the simian diphtheria toxin (DT) receptor in renal epithelia derived from the metanephric mesenchyme. By adjusting the timing and dose of DT, a highly selective model of tubular injury was created to define the acute and chronic consequences of isolated epithelial injury. The DT-induced sublethal tubular epithelial injury was confined to the S1 and S2 segments of the proximal tubule rather than being widespread in the metanephric mesenchyme-derived epithelial lineage. Acute injury was promptly followed by inflammatory cell infiltration and robust tubular cell proliferation, leading to complete recovery after a single toxin insult. In striking contrast, three insults to renal epithelial cells at 1-week intervals resulted in maladaptive repair with interstitial capillary loss, fibrosis, and glomerulosclerosis, which was highly correlated with the degree of interstitial fibrosis. Thus, selective epithelial injury can drive the formation of interstitial fibrosis, capillary rarefaction, and potentially glomerulosclerosis, substantiating a direct role for damaged tubule epithelium in the pathogenesis of CKD.


Subject(s)
Acute Kidney Injury/complications , Epithelial Cells/pathology , Glomerulonephritis/etiology , Kidney Tubules, Proximal/pathology , Acute Kidney Injury/genetics , Acute Kidney Injury/immunology , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Apoptosis , Cell Proliferation , Disease Models, Animal , Disease Progression , Epithelial Cells/immunology , Epithelial Cells/metabolism , Fibrosis , Glomerulonephritis/genetics , Glomerulonephritis/immunology , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Tubules, Proximal/immunology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/physiopathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Regeneration , Time Factors
5.
Cell Physiol Biochem ; 29(3-4): 523-32, 2012.
Article in English | MEDLINE | ID: mdl-22508059

ABSTRACT

The Th1/Th2 balance represents an important factor in the pathogenesis of renal ischemia-reperfusion injury (IRI). In addition, IRI causes a systemic inflammation that can affect other tissues, such as the lungs. To investigate the ability of renal IRI to modulate pulmonary function in a specific model of allergic inflammation, C57Bl/6 mice were immunized with ovalbumin/albumen on days 0 and 7 and challenged with an ovalbumin (OA) aerosol on days 14 and 21. After 24 h of the second antigen challenge, the animals were subjected to 45 minutes of ischemia. After 24 h of reperfusion, the bronchoalveolar lavage (BAL) fluid, blood and lung tissue were collected for analysis. Serum creatinine levels increased in both allergic and non-immunized animals subjected to IRI. However, BAL analysis showed a reduction in the total cells (46%) and neutrophils (58%) compared with control allergic animals not submitted to IRI. In addition, OA challenge induced the phosphorylation of ERK and Akt and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in lung homogenates. After renal IRI, the phosphorylation of ERK and expression of COX-2 and iNOS were markedly reduced; however, there was no difference in the phosphorylation of Akt between sham and ischemic OA-challenged animals. Mucus production was also reduced in allergic mice after renal IRI. IL-4, IL-5 and IL-13 were markedly down-regulated in immunized/challenged mice subjected to IRI. These results suggest that renal IRI can modulate lung allergic inflammation, probably by altering the Th1/Th2 balance and, at least in part, by changing cellular signal transduction factors.


Subject(s)
Kidney/injuries , Lung/immunology , Reperfusion Injury/immunology , Th1-Th2 Balance , Animals , Blood Cell Count , Bronchoalveolar Lavage Fluid/immunology , Creatinine/blood , Cyclooxygenase 2/metabolism , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interleukins/immunology , Interleukins/metabolism , Kidney/immunology , Kidney/pathology , Lung/metabolism , Lung/pathology , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mucus/immunology , Neutrophils/metabolism , Nitric Oxide Synthase Type II/metabolism , Ovalbumin/administration & dosage , Ovalbumin/immunology , Phosphorylation
6.
PLoS One ; 12(7): e0181861, 2017.
Article in English | MEDLINE | ID: mdl-28746409

ABSTRACT

ZSF1 rats exhibit spontaneous nephropathy secondary to obesity, hypertension, and diabetes, and have gained interest as a model system with potentially high translational value to progressive human disease. To thoroughly characterize this model, and to better understand how closely it recapitulates human disease, we performed a high resolution longitudinal analysis of renal disease progression in ZSF1 rats spanning from early disease to end stage renal disease. Analyses included metabolic endpoints, renal histology and ultrastructure, evaluation of a urinary biomarker of fibrosis, and transcriptome analysis of glomerular-enriched tissue over the course of disease. Our findings support the translational value of the ZSF1 rat model, and are provided here to assist researchers in the determination of the model's suitability for testing a particular mechanism of interest, the design of therapeutic intervention studies, and the identification of new targets and biomarkers for type 2 diabetic nephropathy.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/genetics , Kidney Failure, Chronic/complications , Kidney/metabolism , Animals , Cluster Analysis , Collagen/genetics , Collagen/metabolism , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Immunohistochemistry , Kidney/pathology , Kidney/ultrastructure , Kidney Failure, Chronic/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Male , Microscopy, Electron, Transmission , Obesity/complications , Rats , Reverse Transcriptase Polymerase Chain Reaction
7.
PLoS One ; 11(5): e0155368, 2016.
Article in English | MEDLINE | ID: mdl-27171494

ABSTRACT

The cytokine TWEAK and its cognate receptor Fn14 are members of the TNF/TNFR superfamily and are upregulated in tissue injury to mediate local tissue responses including inflammation and tissue remodeling. We found that in various models of kidney disease, Fn14 expression (mRNA and protein) is upregulated in the kidney. These models include: lupus nephritis mouse models (Nephrotoxic serum Transfer Nephritis and MRL.Faslpr/lpr), acute kidney injury models (Ischemia reperfusion injury and Folic acid injury), and a ZSF-1 diabetic nephropathy rat model. Fn14 expression levels correlate with disease severity as measured by disease histology. We have also shown for the first time the detection of soluble Fn14 (sFn14) in the urine and serum of mice. Importantly, we found the sFn14 levels are markedly increased in the diseased mice and are correlated with disease biomarkers including proteinuria and MCP-1. We have also detected sFn14 in human plasma and urine. Moreover, sFn14 levels, in urine are significantly increased in DN patients and correlated with proteinuria and MCP-1 levels. Thus our data not only confirm the up-regulation of Fn14/TWEAK pathway in kidney diseases, but also suggest a novel mechanism for its regulation by the generation of sFn14. The correlation of sFn14 levels and disease severity suggest that sFn14 may serve as a potential biomarker for both acute and chronic kidney diseases.


Subject(s)
Kidney Diseases/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Acute Kidney Injury/blood , Acute Kidney Injury/pathology , Acute Kidney Injury/urine , Adult , Animals , Chromatography, Liquid , Disease Models, Animal , Folic Acid/metabolism , Humans , Kidney/metabolism , Kidney/pathology , Lupus Nephritis/blood , Lupus Nephritis/pathology , Lupus Nephritis/urine , Male , Mice , Receptors, Tumor Necrosis Factor/blood , Reperfusion Injury/blood , Reperfusion Injury/pathology , Reperfusion Injury/urine , Solubility , TWEAK Receptor , Tandem Mass Spectrometry , Up-Regulation
8.
EBioMedicine ; 2(9): 1090-101, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26501107

ABSTRACT

Global or local ischemia contributes to the pathogenesis of acute kidney injury (AKI). Currently there are no specific therapies to prevent AKI. Potentiation of glycolytic metabolism and attenuation of mitochondrial respiration may decrease cell injury and reduce reactive oxygen species generation from the mitochondria. Meclizine, an over-the-counter anti-nausea and -dizziness drug, was identified in a 'nutrient-sensitized' chemical screen. Pretreatment with 100 mg/kg of meclizine, 17 h prior to ischemia protected mice from IRI. Serum creatinine levels at 24 h after IRI were 0.13 ± 0.06 mg/dl (sham, n = 3), 1.59 ± 0.10 mg/dl (vehicle, n = 8) and 0.89 ± 0.11 mg/dl (meclizine, n = 8). Kidney injury was significantly decreased in meclizine treated mice compared with vehicle group (p < 0.001). Protection was also seen when meclizine was administered 24 h prior to ischemia. Meclizine reduced inflammation, mitochondrial oxygen consumption, oxidative stress, mitochondrial fragmentation, and tubular injury. Meclizine preconditioned kidney tubular epithelial cells, exposed to blockade of glycolytic and oxidative metabolism with 2-deoxyglucose and NaCN, had reduced LDH and cytochrome c release. Meclizine upregulated glycolysis in glucose-containing media and reduced cellular ATP levels in galactose-containing media. Meclizine inhibited the Kennedy pathway and caused rapid accumulation of phosphoethanolamine. Phosphoethanolamine recapitulated meclizine-induced protection both in vitro and in vivo.


Subject(s)
Ischemic Preconditioning , Kidney/blood supply , Kidney/pathology , Meclizine/therapeutic use , Protective Agents/therapeutic use , Reperfusion Injury/drug therapy , Acute Kidney Injury/complications , Acute Kidney Injury/drug therapy , Acute Kidney Injury/pathology , Adenosine Triphosphate/metabolism , Animals , Cell Respiration/drug effects , Cytochromes c/metabolism , Deoxyglucose/pharmacology , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Ethanolamines/metabolism , Galactose/pharmacology , Glycolysis/drug effects , Humans , Inflammation/complications , Inflammation/pathology , Kidney/drug effects , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/pathology , L-Lactate Dehydrogenase/metabolism , LLC-PK1 Cells , Male , Meclizine/pharmacology , Meclizine/toxicity , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Protective Agents/pharmacology , Reperfusion Injury/complications , Reperfusion Injury/pathology , Sodium Cyanide/pharmacology , Swine , Up-Regulation/drug effects
9.
PLoS One ; 8(7): e68640, 2013.
Article in English | MEDLINE | ID: mdl-23844229

ABSTRACT

Inflammatory macrophages are abundant in kidney disease, stimulating repair, or driving chronic inflammation and fibrosis. Damage associated molecules (DAMPs), released from injured cells engage pattern recognition receptors (PRRs) on macrophages, contributing to activation. Understanding mechanisms of macrophage activation during kidney injury may lead to strategies to alleviate chronic disease. We identified Triggering-Receptor-in-Myeloid-cells (TREM)-1, a regulator of TLR signaling, as highly upregulated in kidney inflammatory macrophages and tested the roles of these receptors in macrophage activation and kidney disease. Kidney DAMPs activated macrophages in vitro, independently of TREM-1, but partially dependent on TLR-2/-4, MyD88. In two models of progressive interstitial kidney disease, TREM-1 blockade had no impact on disease or macrophage activation in vivo, but TLR-2/-4, or MyD88 deficiency was anti-inflammatory and anti-fibrotic. When MyD88 was mutated only in the myeloid lineage, however, there was no bearing on macrophage activation or disease progression. Instead, TLR-2/-4 or MyD88 deficiency reduced activation of mesenchyme lineage cells resulting in reduced inflammation and fibrosis, indicating that these pathways play dominant roles in activation of myofibroblasts but not macrophages. To conclude, TREM-1, TLR2/4 and MyD88 signaling pathways are redundant in myeloid cell activation in kidney injury, but the latter appear to regulate activation of mesenchymal cells.


Subject(s)
Membrane Glycoproteins/metabolism , Myeloid Cells/metabolism , Receptors, Immunologic/metabolism , Renal Insufficiency, Chronic/metabolism , Signal Transduction , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Disease Models, Animal , Female , Fibrosis , Gene Expression Regulation , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Membrane Glycoproteins/genetics , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88 , Receptors, Immunologic/genetics , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/immunology , Renal Insufficiency, Chronic/pathology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Triggering Receptor Expressed on Myeloid Cells-1
10.
Eur J Pharmacol ; 634(1-3): 132-7, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20153312

ABSTRACT

Lipopolysaccharides from gram-negative bacteria are amongst the most common causative agents of acute lung injury, which is characterized by an inflammatory response, with cellular infiltration and the release of mediators/cytokines. There is evidence that bradykinin plays a role in lung inflammation in asthma but in other types of lung inflammation its role is less clear. In the present study we evaluated the role of the bradykinin B1 receptor in acute lung injury caused by lipopolysaccharide inhalation and the mechanisms behind bradykinin actions participating in the inflammatory response. We found that in C57Bl/6 mice, the bradykinin B1 receptor expression was up-regulated 24h after lipopolysaccharide inhalation. At this time, the number of cells and protein concentration were significantly increased in the bronchoalveolar lavage fluid and the mice developed airway hyperreactivity to methacholine. In addition, there was an increased expression of tumor necrosis factor-alpha, interleukin-1 beta and interferon-gamma and chemokines (monocytes chemotactic protein-1 and KC) in the bronchoalveolar lavage fluid and in the lung tissue. We then treated the mice with a bradykinin B1 receptor antagonist, R-954 (Ac-Orn-[Oic2, alpha-MePhe5, D-betaNal7, Ile8]desArg9-bradykinin), 30 min after lipopolysaccharide administration. We observed that this treatment prevented the airway hyperreactivity as well as the increased cellular infiltration and protein content in the bronchoalveolar lavage fluid. Moreover, R-954 inhibited the expression of cytokines/chemokines. These results implicate bradykinin, acting through B1 receptor, in the development of acute lung injury caused by lipopolysaccharide inhalation.


Subject(s)
Acute Lung Injury/metabolism , Disease Models, Animal , Lipopolysaccharides/toxicity , Receptor, Bradykinin B1/biosynthesis , Receptor, Bradykinin B1/physiology , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Administration, Inhalation , Animals , Bradykinin/administration & dosage , Bradykinin/analogs & derivatives , Bradykinin/therapeutic use , Bradykinin B1 Receptor Antagonists , Bronchoalveolar Lavage Fluid , Cytokines/analysis , Cytokines/biosynthesis , Inflammation Mediators/administration & dosage , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Inbred C57BL , Receptor, Bradykinin B1/administration & dosage
11.
Int Immunopharmacol ; 9(6): 653-7, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19010452

ABSTRACT

The Kallikrein-kinin system works through activation of two receptors. One constitutive, named B2 receptor (B2R) and another inducible, denominated B1 receptor (B1R). In renal fibrosis, B2R receptor activation appears to be protective, however B1R participation is unveiled. The aim of this study was to analyze how the deletion of the B1R would modify tissue responses after unilateral ureteral obstruction (UUO). For that, B1R knockout (B1KO) and wild-type mice (B1B2WT) were subjected to UUO and sacrificed at days 1, 5 and 14. Renal dysfunction was assayed by urine proteinuria/creatinine ratio and percentage of tubulointerstitial fibrosis. Kidneys were harvested at day 5 to analyze anti and pro-inflammatory molecules expression by real-time PCR. We demonstrated that at all time points, B1KO mice presented lower proteinuria/creatinine ratio from bladder urine. B1KO protection was reinforced by its lower tubular interstitial fibrosis percentage at day 14 (B1B2WT: 12.16+/-1.53% vs. B1KO: 6.73+/-1.07%, p<0.02). UUO was able to induce B1R expression and its highest transcription was achieved at day 5. At this day, B1KO had significant lower expression of pro-inflammatory molecules such as TGF-beta, MCP-1, OPN and IL-6 and higher anti-inflammatory components, as IL-10 and HO-1. Herein, we observed that B1R deletion may be an important component in renal fibrosis prevention.


Subject(s)
Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney/pathology , Receptor, Bradykinin B1/genetics , Animals , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Creatinine/blood , Creatinine/urine , Fibrosis , Gene Deletion , Heme Oxygenase-1/immunology , Heme Oxygenase-1/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Kallikrein-Kinin System/genetics , Kidney Diseases/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteinuria/urine , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Ureteral Obstruction/complications
12.
PLoS One ; 3(8): e3050, 2008 Aug 25.
Article in English | MEDLINE | ID: mdl-18725957

ABSTRACT

Previously we have demonstrated that bradykinin B1 receptor deficient mice (B1KO) were protected against renal ischemia and reperfusion injury (IRI). Here, we aimed to analyze the effect of B1 antagonism on renal IRI and to study whether B1R knockout or antagonism could modulate the renal expression of pro and anti-inflammatory molecules. To this end, mice were subjected to 45 minutes ischemia and reperfused at 4, 24, 48 and 120 hours. Wild-type mice were treated intra-peritoneally with antagonists of either B1 (R-954, 200 microg/kg) or B2 receptor (HOE140, 200 microg/kg) 30 minutes prior to ischemia. Blood samples were collected to ascertain serum creatinine level, and kidneys were harvested for gene transcript analyses by real-time PCR. Herein, B1R antagonism (R-954) was able to decrease serum creatinine levels, whereas B2R antagonism had no effect. The protection seen under B1R deletion or antagonism was associated with an increased expression of GATA-3, IL-4 and IL-10 and a decreased T-bet and IL-1beta transcription. Moreover, treatment with R-954 resulted in lower MCP-1, and higher HO-1 expression. Our results demonstrated that bradykinin B1R antagonism is beneficial in renal IRI.


Subject(s)
Bradykinin B1 Receptor Antagonists , Kidney Diseases/drug therapy , Reperfusion Injury/drug therapy , Animals , Bradykinin/analogs & derivatives , Bradykinin/therapeutic use , Caspase 3/metabolism , Cell Death , DNA Primers , GATA3 Transcription Factor/therapeutic use , Gene Deletion , Gene Expression Regulation , Humans , Interleukin-10/therapeutic use , Interleukin-4/therapeutic use , Kidney/physiology , Kidney/physiopathology , Kidney Diseases/mortality , Kidney Diseases/pathology , Male , Mice , Mice, Knockout , Receptor, Bradykinin B1/deficiency , Receptor, Bradykinin B1/genetics , Reperfusion Injury/mortality , Reperfusion Injury/pathology , Survivors
SELECTION OF CITATIONS
SEARCH DETAIL