151
|
Kim J, Kil IS, Seok YM, Yang ES, Kim DK, Lim DG, Park JW, Bonventre JV, Park KM. Orchiectomy attenuates post-ischemic oxidative stress and ischemia/reperfusion injury in mice. A role for manganese superoxide dismutase. J Biol Chem 2006; 281:20349-56. [PMID: 16682413 DOI: 10.1074/jbc.m512740200] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Males are much more susceptible to ischemia/reperfusion (I/R)-induced kidney injury when compared with females. Recently we reported that the presence of testosterone, rather than the absence of estrogen, plays a critical role in gender differences in kidney susceptibility to I/R injury in mice. Although reactive oxygen species and antioxidant defenses have been implicated in I/R injury, their roles remain to be defined. Here we report that the orchiectomized animal had significantly less lipid peroxidation and lower hydrogen peroxide levels in the kidney 4 and 24 h after 30 min of bilateral renal ischemia when compared with intact or dihydrotestosterone-treated orchiectomized males. The post-ischemic kidney expression and activity of manganese superoxide dismutase (MnSOD) in orchiectomized mice was much greater than in intact or dihydrotestosterone-administered orchiectomized mice. Four hours after 30 min of bilateral ischemia, superoxide formation was significantly lower in orchiectomized mice than in intact mice. In Madin-Darby canine kidney cells, a kidney epithelial cell line, 1 mm H(2)O(2) decreased MnSOD activity, an effect that was potentiated by pretreatment with dihydrotestosterone. Orchiectomy prevented the post-ischemic decrease of catalase activity. Treatment of male mice with manganese(III) tetrakis(1-methyl-4-pyridyl)porphyrin (MnTMPyP), a SOD mimetic, reduced the post-ischemic increase of plasma creatinine, lipid peroxidation, and tissue hydrogen peroxide. These results suggest that orchiectomy accelerates the post-ischemic activation of MnSOD and reduces reactive oxygen species and lipid peroxidation, resulting in reduced kidney susceptibility to I/R injury.
Collapse
Affiliation(s)
- Jinu Kim
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
152
|
Chander V, Chopra K. Protective effect of nitric oxide pathway in resveratrol renal ischemia-reperfusion injury in rats. Arch Med Res 2006; 37:19-26. [PMID: 16314181 DOI: 10.1016/j.arcmed.2005.05.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2004] [Accepted: 05/11/2005] [Indexed: 11/22/2022]
Abstract
BACKGROUND Nitric oxide (NO), synthesized from L-arginine by the enzyme nitric oxide synthase (NOS), seems to play an ambiguous role during tissue ischemia-reperfusion (I/R) injury. This study was designed to investigate the effects of resveratrol, a polyphenolic phytoalexin, in renal ischemia reperfusion (RIR) injury in rats. METHODS Forty-eight rats were randomized into six groups. Group 1: sham operated (C); group 2: right nephrectomy (UNI); group 3: UNI + 45 min of ischemia and 24 h of reperfusion in the contralateral kidney; group 4: UNI + RIR + L-NAME (10 mg/kg, i.p.); group 5: UNI + RIR + resveratrol (5 mg/kg, p.o.); group 6: UNI + RIR + resveratrol + L-NAME. At the end of the reperfusion period, rats were sacrificed. Thiobarbituric acid reactive substances (TBARS), reduced glutathione (GSH) levels, catalase (CAT), and superoxide dismutase (SOD) activities were determined in renal tissue. Serum creatinine and blood urea nitrogen (BUN) were measured for the evaluation of renal function. Tissue and urine nitrite levels were measured to assess total nitric oxide levels. RESULTS Ischemic control animals demonstrated severe deterioration of renal function, altered renal morphology, reduced total nitric oxide levels and a marked renal oxidative stress. CONCLUSIONS Pretreatment of animals with resveratrol markedly attenuated renal dysfunction, morphological alterations, improved nitric oxide levels, reduced elevated TBARS levels and restored the depleted renal antioxidant enzymes, However, treatment with L-NAME attenuated this protection afforded by resveratrol indicating that resveratrol exerts its protective effect through NO release.
Collapse
Affiliation(s)
- Vikas Chander
- Pharmacology Division, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | | |
Collapse
|
153
|
Abstract
Acute renal failure has 50-80% mortality. Currently, treatment options for this life-threatening disease are limited. Stem cells offer an exciting potential for kidney regeneration. This review discusses pathogenesis of acute renal failure resulting from ischemia-reperfusion injury and the role of stem cells in reversing or mitigating this disorder. Specifically, the issues of differentiation of kidney cells from embryonic stem cells and bone marrow stem cells, and whether adult kidney stem/progenitor cells exist in the postnatal kidney are discussed. Evidence to support the conclusion that intra-renal cells, including surviving tubular epithelial cells and potential renal stem/progenitor cells, are the main source for renal regeneration is provided. Future research in selecting the type(s) of stem cells and optimizing the dose, frequency and route of administration of the cells will be fundamental in successful cell replacement therapy in acute renal failure. Methods for enhancing endogenous renal cell proliferation and differentiation for renal repair continue to be important research directions.
Collapse
Affiliation(s)
- Fangming Lin
- Department of Pediatrics, University of Texas Southwestern Medical Center at Dallas, 75390, USA.
| |
Collapse
|
154
|
Foglieni C, Fulgenzi A, Ticozzi P, Pellegatta F, Sciorati C, Belloni D, Ferrero E, Ferrero ME. Protective effect of EDTA preadministration on renal ischemia. BMC Nephrol 2006; 7:5. [PMID: 16536881 PMCID: PMC1501003 DOI: 10.1186/1471-2369-7-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Accepted: 03/15/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chelation therapy with sodium edetate (EDTA) improved renal function and slowed the progression of renal insufficiency in patients subjected to lead intoxication. This study was performed to identify the underlying mechanism of the ability of EDTA treatment to protect kidneys from damage. METHODS The effects of EDTA administration were studied in a rat model of acute renal failure induced by 60 minutes ischemia followed or not by 60 minutes reperfusion. Renal ischemic damage was evaluated by histological studies and by functional studies, namely serum creatinine and blood urea nitrogen levels. Treatment with EDTA was performed 30 minutes before the induction of ischemia. Polymorphonuclear cell (PMN) adhesion capability, plasmatic nitric oxide (NO) levels and endothelial NO synthase (eNOS) renal expression were studied as well as the EDTA protection from the TNFalpha-induced vascular leakage in the kidneys. Data was compared by two-way analysis of variance followed by a post hoc test. RESULTS EDTA administration resulted in the preservation of both functional and histological parameters of rat kidneys. PMN obtained from peripheral blood of EDTA-treated ischemized rats, displayed a significant reduction in the expression of the adhesion molecule Mac-1 with respect to controls. NO was significantly increased by EDTA administration and eNOS expression was higher and more diffuse in kidneys of rats treated with EDTA than in the controls. Finally, EDTA administration was able to prevent in vivo the TNFalpha-induced vascular leakage in the kidneys. CONCLUSION This data provides evidence that EDTA treatment is able to protect rat kidneys from ischemic damage possibly through the stimulation of NO production.
Collapse
Affiliation(s)
- Chiara Foglieni
- Cardiovascular Department, Istituto Scientifico San Raffaele, via Olgettina, 60 Milan, Italy
| | - Alessandro Fulgenzi
- Istituto di Patologia Generale, Università degli Studi di Milano, via Mangiagalli 31, Milan, Italy
| | - Paolo Ticozzi
- Istituto di Patologia Generale, Università degli Studi di Milano, via Mangiagalli 31, Milan, Italy
| | - Fabio Pellegatta
- Istituto di Scienze Farmacologiche Università degli Studi di Milano, Via Balzaretti 22, Milan, Italy
| | - Clara Sciorati
- Laboratory of Tumor Immunology, Istituto Scientifico San Raffaele, Via Olgettina 60, Milan, Italy
| | - Daniela Belloni
- Istituto di Patologia Generale, Università degli Studi di Milano, via Mangiagalli 31, Milan, Italy
| | - Elisabetta Ferrero
- Laboratory of Tumor Immunology, Istituto Scientifico San Raffaele, Via Olgettina 60, Milan, Italy
| | - Maria Elena Ferrero
- Istituto di Patologia Generale, Università degli Studi di Milano, via Mangiagalli 31, Milan, Italy
| |
Collapse
|
155
|
Kang DG, Sohn EJ, Moon MK, Mun YJ, Woo WH, Kim MK, Lee HS. Yukmijihwang-tang ameliorates ischemia/reperfusion-induced renal injury in rats. JOURNAL OF ETHNOPHARMACOLOGY 2006; 104:47-53. [PMID: 16183223 DOI: 10.1016/j.jep.2005.08.044] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2004] [Revised: 07/29/2005] [Accepted: 08/15/2005] [Indexed: 05/04/2023]
Abstract
The present study was designed to examine whether Yukmijihwang-tang (YJT), which is a Korean decoction for the treatment of renal disease, has an effect on renal functional parameters in association with the expression of aquaporin 2 (AQP 2), Na,K-ATPase, heme oxygenase-1 (HO-1) in rats with ischemia/reperfusion-induced acute renal failure (ARF). Polyuria caused by down-regulation of renal AQP 2 in the ischemia/reperfusion-induced ARF rats was markedly restored by administration of YJT (100 or 200 mg/kg, p.o.) with restoring expression of AQP 2 in the kidney. The expressions of Na,K-ATPase alpha1 and beta1 subunits in the renal medulla and cortex of the ARF rats were also restored in them by the administration of YJT. Administration of YJT lowered the expression of renal HO-1, which was up-regulated in rats with ischemia/reperfusion-induced ARF. The renal functional parameters including creatinine clearance, urinary sodium excretion, urinary osmolality, and solute-free reabsorption were also markedly restored in ischemia-ARF rats by administration of YJT. Histological study also showed that renal damages in the ARF rats were abrogated by administration of YJT. Taken together, these data indicate that YJT ameliorates renal defects in rats with ischemia/reperfusion-induced ARF.
Collapse
Affiliation(s)
- Dae Gill Kang
- Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
156
|
Antunes N, Martinusso CA, Takiya CM, da Silva AJR, de Ornellas JFR, Elias PR, Leite M, Cardoso LR. Fructose-1,6 diphosphate as a protective agent for experimental ischemic acute renal failure. Kidney Int 2006; 69:68-72. [PMID: 16374425 DOI: 10.1038/sj.ki.5000013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cold ischemia time is a risk factor for the development of acute renal failure in the immediate post-transplant period. In this study, we aimed to determine if intravenous fructose-1,6-diphosphate (FDP), given before nephrectomy, attenuates renal cell injury in a cold ischemia model. Male adult Wistar rats were subjected to infusion of either FDP 350 mg/kg (group F, n=6), an equal volume of 0.9% NaCl (group S, n=6), an equal volume/osmolality of mannitol (group M, n=6) or no infusion (group C, n=7). Kidneys were then perfused in situ with Collins solution and nephrectomy was performed. Other kidney slices were stored in Collins solution at 4 degrees C. Adenosine triphosphate (ATP) levels and lactate dehydrogenase (LDH) release were examined at 0, 24, 48 and 72 h. Other slices, obtained after 50 min immersion in Collins solution at 37 degrees C, were frozen for characterization of cytoskeletal preservation using phalloidin-FITC staining. Apical fluorescence intensity of proximal tubule cells, indicative of the F-actin concentration, was measured in a fluorescence microscope interfaced with computer image analysis system. Adenosine triphosphate levels, after up to 72 h of tissue incubation, were higher (P<0.05) in the FDP group when compared to other groups. In addition, LDH release was smaller (P<0.0001) in the FDP group. The F-actin concentration of proximal tubule cells cells was greater in the FDP group (P<0.0001). Results indicate that FDP is a useful tool to increase tissue viability in a rat kidney subjected to cold ischemia, by maintaining ATP cell content, decreasing LDH release and preventing microfilament disruption of proximal tubule cells.
Collapse
Affiliation(s)
- N Antunes
- Universidade Federal do Rio de Janeiro: Nefrologica, HUCFF, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
157
|
Takahashi T, Shimizu H, Akagi R, Morita K, Sassa S. Heme oxygenase-1: a new drug target in oxidative tissue injuries in critically ill conditions. Drug Dev Res 2006. [DOI: 10.1002/ddr.20073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
158
|
Cheng CW, Rifai A, Ka SM, Shui HA, Lin YF, Lee WH, Chen A. Calcium-binding proteins annexin A2 and S100A6 are sensors of tubular injury and recovery in acute renal failure. Kidney Int 2006; 68:2694-703. [PMID: 16316344 DOI: 10.1111/j.1523-1755.2005.00740.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Rise in cellular calcium is associated with acute tubular necrosis, the most common cause of acute renal failure (ARF). The mechanisms that calcium signaling induce in the quiescent tubular cells to proliferate and differentiate during acute tubular necrosis have not been elucidated. METHODS Acute tubular necrosis induced in mice by single intravenous injection of uranyl nitrate and examined after 1, 3, 7, and 14 days. Renal function was monitored and kidneys were evaluated by histology, immunohistochemistry, Western blotting, in situ hybridization, and real-time reverse transcription-polymerase chain reaction (RT-PCR). Models of folic acid induced-ARF and ischemic/reperfusion (I/R) injury were similarly investigated. RESULTS Analysis of mRNA expression of intracellular calcium and phospholipid-binding proteins demonstrated selective expression of S100A6 and Annexin A2 (Anxa2) in the renal cortex with marked elevation on day 3, and gradually decline on day 7 and further attenuation on day 14. Similarly, the expression of both proteins, as demonstrated by immunohistochemistry and Western blot analysis, was increased and reached the peak level on day 7 and then gradually declined by day 14. Vimentin, a marker of dedifferentiated cells, was highly expressed during the recovery phase. Combined in situ hybridization immunohistochemistry revealed colocalization of both S100A6 and Anxa2 with proliferating cell nuclear antigen (PCNA). The universality of this phenomenon was confirmed in two other mouse acute tubular necrosis models, the ischemic-reperfusion injury and folic acid-induced ARF. CONCLUSION Collectively, these findings demonstrate that S100A6 and Anxa2 expression, initiated in response to tubular injury, persist in parallel throughout the recovery process of tubular cells in acute renal failure.
Collapse
Affiliation(s)
- Chao-Wen Cheng
- Graduate Institute of Life Sciences, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
159
|
Zahedi K, Revelo MP, Barone S, Wang Z, Tehrani K, Citron DP, Bissler JJ, Rabb H, Soleimani M. Stathmin-deficient mice develop fibrosis and show delayed recovery from ischemic-reperfusion injury. Am J Physiol Renal Physiol 2006; 290:F1559-67. [PMID: 16434570 DOI: 10.1152/ajprenal.00424.2005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In kidneys subjected to ischemic reperfusion injury (IRI) stathmin, a tubulin-binding protein involved in the regulation of mitosis, is expressed in dedifferentiated and proliferating renal tubule cells during the recovery phase. To ascertain the role of stathmin in the recovery from ischemic kidney injury, stathmin-deficient (OP18-/-) and wild-type (WT) animals were subjected to experimental IRI. At 3, 7, and 14 days after reperfusion serum samples and kidneys were collected for the examination of parameters of renal function, morphology, and recovery. Our studies indicate that on day 14 after reperfusion OP18-/- mice have significant renal failure, whereas the creatinine levels of WT animals have returned to baseline. Compared with WT animals OP18-/- mice had more extensive tubular fibrosis. The examination of proliferating cell nuclear antigen expression indicated that OP18-/- animals have increased proliferative or DNA repair activity for a more prolonged duration. The OP18-/- animals also had an increased number of tubules with apoptotic cells. These results suggest that in stathmin-deficient mice subjected to IRI, the aberrant regulation of cell cycle progression, not observed under normal conditions, impairs or at least delays the process of tubular repair and recovery after acute renal injury.
Collapse
Affiliation(s)
- Kamyar Zahedi
- Division of Nephrology and Hypertension, Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Hong ME, Hong JC, Stepkowski S, Kahan BD. Correlation Between Cyclosporine-Induced Nephrotoxicity in Reduced Nephron Mass and Expression of Kidney Injury Molecule-1 and Aquaporin-2 Gene. Transplant Proc 2005; 37:4254-8. [PMID: 16387092 DOI: 10.1016/j.transproceed.2005.10.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Indexed: 02/07/2023]
Abstract
Sirolimus (SRL) has been shown to exacerbate cyclosporine (CsA)-induced nephrotoxicity. The expression of the kidney injury molecule-1 (KIM-1) is markedly upregulated in the postischemic rat kidney. We sought to correlate drug-induced nephrotoxicity and the expression of KIM-1 and aquaporin-2 (AQP-2) in male PVG rats with 2 kidneys (2K), 1 kidney (1K), and half a kidney (1/2K) treated with SRL alone, CsA alone, or a combination of both (SRL-CsA). After 7 days of treatment, the 2K group treated with SRL-CsA showed a significant decrease in creatinine clearance compared with the 2K SRL alone and 2K CsA alone groups (1.2 vs 2.47 vs 2.46 mL/min; P < .001). There was a trend toward deterioration of creatinine clearance in the 1K and 1/2K groups treated with SRL-CsA. The KIM-1 expression in the 2K SRL-CsA group was significantly upregulated compared with that in the 2K SRL alone and 2K CsA alone groups (P = .02). The AQP-2 expression was comparable in the 3 groups. After 1 week of treatment washout, the 2K, 1K, and 1/2K groups treated with SRL alone demonstrated a significantly higher creatinine clearance rate than did the groups treated with SRL-CsA (P = .04, P = .02, and P = .004). The expression of KIM-1 and AQP-2 was similar among the treatment groups. SRL-CsA-induced nephrotoxicity resulted in overexpression of KIM-1, suggesting injury to the proximal tubule. Treatment with SRL alone may enable earlier reversal of tubular injury.
Collapse
Affiliation(s)
- M E Hong
- Division of Pediatric Renal Diseases and Hypertension, Department of Pediatrics, The University of Texas Medical School at Houston, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
161
|
Mizutani A, Murakami K, Okajima K, Kira SI, Mizutani S, Kudo K, Takatani J, Goto K, Hattori S, Noguchi T. Olprinone reduces ischemia/reperfusion-induced acute renal injury in rats through enhancement of cAMP. Shock 2005; 24:281-7. [PMID: 16135969 DOI: 10.1097/01.shk.0000175555.95676.34] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Activated leukocytes are implicated in development of ischemia/reperfusion (I/R)-induced organ injuries. Phosphodiesterase 3 inhibitors have anti-inflammatory effects by preventing cyclic adenosine monophosphate (cAMP) degradation. We examined the effects of olprinone, a specific phosphodiesterase 3 inhibitor, on I/R-induced acute renal injury model in rats. Forty-five minute renal I/R was induced in uni-nephrectomized rats. Rats were divided into a vehicle group, an olprinone group, and a dibutyril (DB) cAMP group. Olprinone (0.2 microg/kg/minute) infusion began 30 min after reperfusion and continued for 3 h. DBcAMP (5 mg/kg), a stable analog of cAMP, was intraperitoneally administered 5 min after reperfusion to clarify the effect of cAMP in our model. Olprinone reduced the I/R-induced increases in serum levels of blood urea nitrogen and creatinine, and improved histological changes, including acute tubular necrosis in the outer medulla. Hemodynamic status was not affected by olprinone. I/R-induced a decrease in renal tissue blood flow, an increase in renal vascular permeability, and an enhancement of leukocyte activation, reflected by renal tissue levels of myeloperoxidase activity, and the tissue levels of cytokine-induced neutrophil chemoattractant (an equivalent of human interleukin 8) and tumor necrosis factor-alpha were all significantly decreased by olprinone. Olprinone also increased the renal tissue and plasma levels of cAMP in rats subjected to renal I/R. DBcAMP showed similar effects. Our results indicated that olprinone reduced the I/R-induced acute renal injury, probably by inhibiting leukocyte activation. The effects of olprinone could be explained through its action on cAMP levels.
Collapse
Affiliation(s)
- Akio Mizutani
- Division of Intensive Care Unit, Oita University Faculty of Medicine Hospital, Hasama-machi, Oita, 879-5593, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Thakar CV, Zahedi K, Revelo MP, Wang Z, Burnham CE, Barone S, Bevans S, Lentsch AB, Rabb H, Soleimani M. Identification of thrombospondin 1 (TSP-1) as a novel mediator of cell injury in kidney ischemia. J Clin Invest 2005; 115:3451-9. [PMID: 16294224 PMCID: PMC1283940 DOI: 10.1172/jci25461] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2005] [Accepted: 09/13/2005] [Indexed: 12/31/2022] Open
Abstract
Thrombospondin 1 (TSP-1) is a matricellular protein that inhibits angiogenesis and causes apoptosis in vivo and in vitro in several cancerous cells and tissues. Here we identify TSP-1 as the molecule with the highest induction level at 3 hours of IR injury in rat and mouse kidneys subjected to ischemia/reperfusion (IR) injury using the DNA microarray approach. Northern hybridizations demonstrated that TSP-1 expression was undetectable at baseline, induced at 3 and 12 hours, and returned to baseline levels at 48 hours of reperfusion. Immunocytochemical staining identified the injured proximal tubules as the predominant sites of expression of TSP-1 in IR injury and showed colocalization of TSP-1 with activated caspase-3. Addition of purified TSP-1 to normal kidney proximal tubule cells or cells subjected to ATP depletion in vitro induced injury as demonstrated by cytochrome c immunocytochemical staining and caspase-3 activity. The deleterious role of TSP-1 in ischemic kidney injury was demonstrated directly in TSP-1 null mice, which showed significant protection against IR injury-induced renal failure and tubular damage. We propose that TSP-1 is a novel regulator of ischemic damage in the kidney and may play an important role in the pathophysiology of ischemic kidney failure.
Collapse
Affiliation(s)
- Charuhas V Thakar
- Department of Medicine, University of Cincinnati School of Medicine, Cincinnati, Ohio 45267-0585, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Barone S, Okaya T, Rudich S, Petrovic S, Tenrani K, Wang Z, Zahedi K, Casero RA, Lentsch AB, Soleimani M. Distinct and sequential upregulation of genes regulating cell growth and cell cycle progression during hepatic ischemia-reperfusion injury. Am J Physiol Cell Physiol 2005; 289:C826-35. [PMID: 15888550 DOI: 10.1152/ajpcell.00629.2004] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ischemia-reperfusion injury (IRI) in liver and other organs is manifested as an injury phase followed by recovery and resolution. Control of cell growth and proliferation is essential for recovery from the injury. We examined the expression of three related regulators of cell cycle progression in liver IRI: spermidine/spermine N-acetyltransferase (SSAT), p21 (a cyclin-dependent kinase inhibitor), and stathmin. Mice were subjected to hepatic IRI, and liver tissues were harvested at timed intervals. The expression of SSAT, the rate-limiting enzyme in the polyamine catabolic pathway, had increased fivefold 6 h after IRI and correlated with increased putrescine levels in the liver, consistent with increased SSAT enzymatic activity in IRI. The expression of p21, which is transactivated by p53, was undetectable in sham-operated animals but was heavily induced at 12 and 24 h of reperfusion and declined to undetectable baseline levels at 72 h of reperfusion. The interaction of the polyamine pathway with the p53-p21 pathway was shown in vitro, where activation of SSAT with polyamine analog or the addition of putrescine to cultured hepatocytes induced the expression of p53 and p21 and decreased cell viability. The expression of stathmin, which is under negative transcriptional regulation by p21 and controls cell proliferation and progression through mitosis, remained undetectable at 6, 12, and 24 h of reperfusion and was progressively and heavily induced at 48 and 72 h of reperfusion. Double-immunofluorescence labeling with antibodies against stathmin and PCNA, a marker of cell proliferation, demonstrated colocalization of stathmin and PCNA at 48 and 72 h of reperfusion in hepatocytes, indicating the initiation of cell proliferation. The distinct and sequential upregulation of SSAT, p21, and stathmin, along with biochemical activation of the polyamine catabolic pathway in IRI in vivo and the demonstration of p53-p21 upregulation by SSAT and putrescine in vitro, points to the important role of regulators of cell growth and cell cycle progression in the pathophysiology and/or recovery in liver IRI. The data further suggest that SSAT may play a role in the initiation of injury, whereas p21 and stathmin may be involved in the resolution and recovery after liver IRI.
Collapse
Affiliation(s)
- Sharon Barone
- Department of Medicine, University of Cincinnati, 231 Albert Sabin Way, MSB 259G, Cincinnati, Ohio 45267-0585, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Zhu MD, Fang CH, Shi XS. Renal cells apoptosis and Bax, Bcl-2 expression in rats with severe acute pancreatitis. Shijie Huaren Xiaohua Zazhi 2005; 13:2103-2107. [DOI: 10.11569/wcjd.v13.i17.2103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the roles of renal cell apoptosis and expression of Bax and Bcl-2 in renal injury in rats with severe acute pancreatitis (SAP).
METHODS: Thirty rats were randomized into sham operation (SO, n = 15) and SAP group (n = 15). The model of SAP was established by retrograde injection of 50 g/L sodium taurocholate into the biliopancreatic duct in the rats. Serum creatinine (Cr) and blood urea nitrogen (BUN) were determined 6, 12, and 18 h after injection. The pathological changes were observed by light and electron microscopy. The apoptosis of renal cells was determined by TUNEL method, and the expression of Bax and Bcl-2 was detected by immunohistochemical staining (SABC assay).
RESULTS: As compared with those in SO group at the same phase, serum Cr and BUN in SAP group increased markedly. Significant injuries of renal tissue were observed under light and electron microscope.The apoptotic index of renal glomerular cells increased significantly 6, 12, 18 h after injection (3.2±1.1 vs 0.8±0.1, P <0.05; 3.9±1.8 vs 0.9±0.1, P <0.05; 4.8±1.7 vs 0.8±0.2, P <0.01, respectively). The apoptotic index of renal tubule cells also increased significantly (9.8±1.1 vs 1.3±0.5, P <0.01; 12.6±2.3 vs 1.2±0.3, P <0.01; 15.7±2.6 vs 1.3±0.3, P <0.01, respectively). The expression of Bax in renal glomerulus and tubule increased significantly at 6, 12, 18 h, respectively (glomerulus: 3.05±0.86 vs 1.01±0.04, P <0.05; 3.37±1.13 vs 1.06±0.05, P <0.05; 4.11±1.24 vs 1.09±0.03, P <0.05; tubule: 8.17±2.16 vs 1.53±0.07, P <0.01; 12.92±1.81 vs 1.61±0.05, P <0.01; 13.08±2.53 vs 1.59±0.05, P <0.01), and increased continuously. Bcl-2 was mainly expressed in the regions of renal tubule, and it also increased in SAP group at 6, 12, 18 h, respectively (3.38±1.07 vs 1.58±0.02, P <0.05; 4.26±1.13 vs 1.59±0.06, P <0.05; 7.21±1.04 vs 1.61±0.02, P <0.01). The level of Bcl-2 expression in renal glomerulus was higher in SAP group than that in SO group at 6, 12, 18 h, respectively (2.18±0.65 vs 1.04±0.02; 2.42±0.63 vs 1.03±0.05, P <0.05; 2.69±0.47 vs 1.08±0.04, P <0.05), but decreased with the duration of the injuries.
CONCLUSION: Renal cell apoptosis, which relates to the up-regulated expression of Bax and downed regulated expression of Bcl-2, may be involved in the pathogenesis of renal injury in patients with SAP.
Collapse
|
165
|
Lin F, Moran A, Igarashi P. Intrarenal cells, not bone marrow-derived cells, are the major source for regeneration in postischemic kidney. J Clin Invest 2005; 115:1756-64. [PMID: 16007252 PMCID: PMC1159127 DOI: 10.1172/jci23015] [Citation(s) in RCA: 350] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Accepted: 05/17/2005] [Indexed: 11/17/2022] Open
Abstract
Ischemic injury to the kidney produces acute tubular necrosis and apoptosis followed by tubular regeneration and recovery of renal function. Although mitotic cells are present in the tubules of postischemic kidneys, the origins of the proliferating cells are not known. Bone marrow cells (BMCs) can differentiate across lineages to repair injured organs, including the kidney. However, the relative contribution of intrarenal cells and extrarenal cells to kidney regeneration is not clear. We produced transgenic mice that expressed enhanced GFP (EGFP) specifically and permanently in mature renal tubular epithelial cells. Following ischemia/reperfusion injury (IRI), EGFP-positive cells showed incorporation of BrdU and expression of vimentin, which provides direct evidence that the cells composing regenerating tubules are derived from renal tubular epithelial cells. In BMC-transplanted mice, 89% of proliferating epithelial cells originated from host cells, and 11% originated from donor BMCs. Twenty-eight days after IRI, the kidneys contained 8% donor-derived cells, of which 8.4% were epithelial cells, 10.6% were glomerular cells, and 81% were interstitial cells. No renal functional improvement was observed in mice that were transplanted with exogenous BMCs. These results show that intrarenal cells are the main source of renal repair, and a single injection of BMCs does not make a significant contribution to renal functional or structural recovery.
Collapse
Affiliation(s)
- Fangming Lin
- Department of Pediatrics and Department of Internal Medicine and Division of Basic Sciences, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 5390-9063, USA.
| | | | | |
Collapse
|
166
|
Devalaraja-Narashimha K, Singaravelu K, Padanilam BJ. Poly(ADP-ribose) polymerase-mediated cell injury in acute renal failure. Pharmacol Res 2005; 52:44-59. [PMID: 15911333 DOI: 10.1016/j.phrs.2005.02.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Accepted: 02/01/2005] [Indexed: 01/12/2023]
Abstract
Acute Renal Failure (ARF) is the most costly kidney disease in hospitalized patients and remains as a serious problem in clinical medicine. The mortality rate among ARF patients remains around 50% and no pharmaceutical agents are currently available to improve its clinical outcome. Although several successful therapeutic approaches have been developed in animal models of the disease, translation of the results to clinical ARF remains elusive. Understanding the cellular and molecular mechanisms of vascular and tubular dysfunction in ARF is important for developing acceptable therapeutic interventions. Following an ischemic episode, cells of the affected nephron undergo necrotic and/or apoptotic cell death. Necrotic cell death is widely considered to be a futile process that cannot be modulated by pharmacological means as opposed to apoptosis. However, recent reports from various laboratories including ours indicate that inhibition or absence of poly(ADP)-ribose polymerase (PARP), one of the molecules involved in cell death, provides remarkable protection in disease models such as stroke, myocardial infarction and renal ischemia which are characterized predominantly by necrotic type of cell death. Overactivation of PARP in conditions such as ischemic renal injury leads to cellular depletion of its substrate NAD+ and consequently ATP. The severely compromised cellular energetic state induces acute cell injury and diminishes renal functions. PARP activation also enhances the expression of proinflammatory agents and adhesion molecules in ischemic kidneys. Pharmacological inhibition and gene ablation of PARP-1 decreased energy depletion, inflammatory response and improved renal functions in the setting renal ischemia/reperfusion injury. The biochemical pathways and the cellular and molecular mechanisms mediated by PARP-1 activation in eliciting the energy depletion and inflammatory responses in ischemic kidney are not fully elucidated. Dissecting the molecular mechanisms by which PARP activation contributes to oxidant-induced cell death will provide new strategies to interfere in those pathways to modulate cell death in renal ischemia. The current review evaluates the experimental evidences in animal and cell culture models implicating PARP as a pathophysiological modulator of acute renal failure with particular emphasis on ischemic renal injury.
Collapse
|
167
|
Tanaka H, Terada Y, Okado T, Inoshita S, Kuwahara M, Sasaki S. Role of the E2F1-p19-p53 pathway in ischemic acute renal failure. Nephron Clin Pract 2005; 101:p27-34. [PMID: 15990448 DOI: 10.1159/000086642] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2003] [Accepted: 03/22/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cell cycle progression and arrest of renal tubular cells after acute injury is a reactive process of renal regeneration. The p16(INK4a)/p19(ARF) (alternative reading frame) locus encodes two proteins involved in cell cycle regulation. We investigated the transcriptional regulation and tissue distribution of p19(ARF) in ischemic acute renal failure (ARF). METHODS We examined the time course and immunohistochemistry of p19(ARF) in rat kidneys following the induction of ischemic ARF. We also examined the effect of p19(ARF) overexpression on p53 levels and cell cycle progression in MDCK cells. RESULTS The protein expression of p19(ARF) strongly increased 72 h after the ischemia. Immunohistochemical studies showed that the renal tubular cells in the outer medulla expressed p19(ARF) protein 72 h after ischemic injury. The time course of E2F1 induction was observed at 6-24 h, and it was found to precede p19(ARF) expression. In MDCK cells, the overexpression of E2F1 increased promoter activity and the protein level of p19(ARF) and induced apoptosis. Transfection of the p19(ARF) expression vector caused an increase in p53 protein, cell cycle arrest and apoptosis. CONCLUSIONS These data support the hypothesis that the E2F1-p19(ARF)-p53 pathway forms a negative feedback loop to regulate the cell cycle of renal tubular cells in the ischemic ARF.
Collapse
Affiliation(s)
- Hiroyuki Tanaka
- Department of Nephrology, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
168
|
Thurman JM, Lucia MS, Ljubanovic D, Holers VM. Acute tubular necrosis is characterized by activation of the alternative pathway of complement. Kidney Int 2005; 67:524-30. [PMID: 15673300 DOI: 10.1111/j.1523-1755.2005.67109.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Studies in animal models have shown that the alternative pathway of complement is activated in the kidney after ischemia/reperfusion. In addition, mice deficient in complement factor B, a necessary component of the alternative pathway, are protected from ischemic acute renal failure. The purpose of this study was to determine whether alternative pathway activation also occurs during the development of ischemic acute tubular necrosis in the human kidney. METHODS Biopsies were identified from nine patients with morphologically normal kidneys and seven patients with evidence of acute tubular necrosis by light microscopy. Immunofluorescence microscopy was used to quantify and localize the complement activation products C3d and C4d. The results were correlated with available clinical data. RESULTS Similar to mice, small amounts of activated C3d were present along the tubular basement membrane in normal kidneys. However, kidneys from patients with acute tubular necrosis had C3d complement deposition along a significantly greater number of tubules, and many of the tubules were completely circumscribed. In contrast, C4d was not detectable, indicating that complement activation occurred primarily via alternative pathway activation. CONCLUSION Complement activation occurs in human ischemic acute tubular necrosis. As in rodents, complement activation along the tubular basement membrane after ischemia appears to occur principally via the alternative complement pathway. Because of this, an inhibitor of the alternative pathway might limit complement activation and inflammation after ischemia/reperfusion, thereby protecting the kidney from ischemic acute renal failure.
Collapse
Affiliation(s)
- Joshua M Thurman
- Department of Internal Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| | | | | | | |
Collapse
|
169
|
Pompermayer K, Souza DG, Lara GG, Silveira KD, Cassali GD, Andrade AA, Bonjardim CA, Passaglio KT, Assreuy J, Cunha FQ, Vieira MAR, Teixeira MM. The ATP-sensitive potassium channel blocker glibenclamide prevents renal ischemia/reperfusion injury in rats. Kidney Int 2005; 67:1785-96. [PMID: 15840025 DOI: 10.1111/j.1523-1755.2005.00276.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Renal ischemia/reperfusion (I/R) is a complex neutrophil-mediated syndrome. Adenosine-triphosphate (ATP)-sensitive potassium (K(ATP)) channels are involved in neutrophil migration in vivo. In the present study, we have investigated the effects of glibenclamide, a K(ATP) channel blocker, in renal I/R injury in rats. METHODS The left kidney of the rats was excised through a flank incision and ischemia was performed in the contralateral kidney by total interruption of renal artery flow for 45 minutes. Renal perfusion was reestablished, and the kidney and lungs were removed for analysis of vascular permeability, neutrophil accumulation, and content of cytokines [tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1beta, and IL-10] 4 and 24 hours later. Renal function was assessed by measuring creatinine, Na(+), and K(+) levels in the plasma and by determination of creatinine clearance. Drugs were administered subcutaneously after the onset of ischemia. RESULTS Reperfusion of the ischemic kidney induced local (kidney) and remote (lung) inflammatory injury and marked renal dysfunction. Glibenclamide (20 mg/kg) significantly inhibited the reperfusion-associated increase in vascular permeability, neutrophil accumulation, increase in TNF-alpha levels and nuclear factor-kappaB (NF-kappaB) translocation. These inhibitory effects were noticed in the kidney and lungs. Moreover, glibenclamide markedly ameliorated the renal dysfunction at 4 and 24 hours. CONCLUSION Treatment with glibenclamide is associated with inhibition of neutrophil recruitment and amelioration of renal dysfunction following renal I/R. Glibenclamide may have a therapeutic role in the treatment of renal I/R injury, such as after renal transplantation.
Collapse
Affiliation(s)
- Kenia Pompermayer
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas-Universidade Federal de Minas Gerais, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Chen W, Langer RM, Janczewska S, Furian L, Geary R, Qu X, Wang M, Verani R, Condon T, Stecker K, Bennett CF, Stepkowski SM. Methoxyethyl-modified intercellular adhesion molecule-1 antisense phosphorothiateoligonucleotides inhibit allograft rejection, ischemic-reperfusion injury, and cyclosporine-induced nephrotoxicity. Transplantation 2005; 79:401-8. [PMID: 15729165 DOI: 10.1097/01.tp.0000149505.53886.27] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The addition of phosphorothioate (PS) groups to natural phosphodiester (PD) antisense oligodeoxynucleotides (oligo) prevents their in vivo hydrolysis by nucleases allowing an RNase-dependent elimination of targeted mRNA. To further improve oligo function 2'-methoxyethyl (ME) groups were attached to selected nucleotides at the 3'-end because ME groups block RNase activity. METHODS/RESULTS ME modification of PS- or PD/PS-oligo targeting human intracellular adhesion molecule (ICAM)-1 mRNA significantly increased the degree and duration of the in vitro inhibitory effects without compromising selectivity and specificity. A 7-day intravenous or oral therapy with rat ME/PS-modified ICAM-1 antisense oligo extended the survivals of kidney allografts. In addition, ME/PS-modified ICAM-1 antisense oligo reduced ischemic-reperfusion injury in kidneys, as measured by glomerular filtration rate, creatinine levels, and infiltration with leukocytes. Finally, a 14-day treatment with cyclosporine (CsA)-induced nephrotoxicity in syngeneic kidney transplants correlated with both increased ICAM-1 protein expression and infiltration with leukocytes. Graft perfusion and treatment of recipients with ICAM-1 antisense ME/PS-oligo alleviated the nephrotoxic effect and decreased ICAM-1 expression and leukocyte infiltration. CONCLUSIONS ME/PS-modified ICAM-1 antisense oligo is very effective in inhibiting the ICAM-1-dependent mechanism of graft infiltration and tissue damage involved in allograft rejection, ischemic-reperfusion injury, and CsA-induced nephrotoxicity.
Collapse
Affiliation(s)
- Wenhau Chen
- Division of Immunology and Organ Transplantation, The University of Texas Medical School-Houston, 6431 Fannin, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
171
|
Affiliation(s)
- M-R Losser
- Département d'anesthésie-réanimation, hôpital Lariboisière AP-HP, 2, rue Ambroise-Paré, 75475 Paris cedex 10, France.
| |
Collapse
|
172
|
Erkan E, Devarajan P, Schwartz GJ. Apoptotic response to albumin overload: proximal vs. distal/collecting tubule cells. Am J Nephrol 2005; 25:121-31. [PMID: 15812145 DOI: 10.1159/000084888] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2004] [Accepted: 02/28/2005] [Indexed: 11/19/2022]
Abstract
End-stage renal disease due to proteinuric states has a great impact on the quality of life by necessitating renal replacement therapy. Understanding the pathophysiologic consequences of proteinuria is crucial in order to develop treatment strategies to halt the progression. We have previously reported that cultured porcine proximal tubule cells respond to albumin overload by undergoing apoptosis. In this study, we investigated the differential apoptotic response to albumin in HKC-8 (proximal tubule) and MDCK (collecting/distal tubule) cells under high concentrations of albumin simulating the nephrotic milieu. Our results are consistent with marked cytotoxicity and apoptosis within 24 h of albumin incubation in HKC-8 cells that was closely related to the fatty acid content of the albumin. In contrast, in MDCK cells, albumin stimulated cell turnover by stimulating proliferation and late onset apoptosis regardless of the fatty acid content. Another important result of this study is the direct demonstration of albumin uptake by MDCK cells mediated by endocytosis via clathrin-coated pits. A comparison of albumin uptake between proximal and distal/collecting tubule cells revealed faster uptake in proximal tubule cells within 15 min but almost 100% albumin uptake of both cell types in 1 h. In summary, our findings demonstrate that both proximal and distal nephron segments are affected in proteinuric states, but the degrees of susceptibility to albumin and associated lipid moieties are distinct in the different nephron segments.
Collapse
Affiliation(s)
- Elif Erkan
- Golisano Children's Hospital at Strong, University of Rochester, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
173
|
Spurgeon KR, Donohoe DL, Basile DP. Transforming growth factor-β in acute renal failure: receptor expression, effects on proliferation, cellularity, and vascularization after recovery from injury. Am J Physiol Renal Physiol 2005; 288:F568-77. [PMID: 15536165 DOI: 10.1152/ajprenal.00330.2004] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transforming growth factor (TGF)-β1and a number of TGF-β-responsive genes are transiently enhanced following induction of ischemic acute renal failure (ARF) in the rat. The mRNA and protein expression of TGF-β receptors were analyzed in postischemic rat kidneys by ribonuclease protection, in situ hybridization, and immunohistochemistry. TGF-βRI and -RII were enhanced within 3 days of ischemia-reperfusion (I/R) injury and remained elevated for up 7 days post-I/R; TGF-β receptor expression was localized primarily in regenerating tubules within the outer medulla. A neutralizing TGF-β antibody exacerbated cellular proliferation observed on day 3 postischemia but had no effect on day 1 or 2. TGF-β antibody treatment had no measurable effect on loss of renal function or the restoration of renal function during the recovery response for up to 35 days postsurgery. However, ischemic injury resulted in modest renal hypertrophy that is due, in part, to in an increase in the number of interstitial cells in the postischemic kidney. Immunohistochemistry showed that several of these cells stained positively for the fibroblast-specific marker, S100A4 positive. Anti-TGF-β treatment substantially attenuated the renal hypertrophy, interstitial cellularity, and S100A4-positive cells present at 35 days post-I/R. Finally, TGF-β immunoneutralization attenuated the loss of renal vascular density following recovery from I/R injury. These data suggest that the TGF-β/TβR system is enhanced in the postischemic kidney. However, the current study failed to identify a prominent role for this system in the repair of proximal tubules following ARF. In contrast, the activation of this system may play an important role in the long-term structure of the postischemic kidney by influencing microvascular structure and interstitial cellularity.
Collapse
Affiliation(s)
- Kimberly R Spurgeon
- Dept. of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | | | |
Collapse
|
174
|
Lameire N. [Which are the therapeutic interventions allowing to ensure a protection of the renal function?]. ANNALES FRANCAISES D'ANESTHESIE ET DE REANIMATION 2005; 24:206-21. [PMID: 15737508 DOI: 10.1016/j.annfar.2004.12.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Affiliation(s)
- N Lameire
- Service de néphrologie, faculté de médecine, hôpital universitaire Gand-De-Pintelaan, 185, 9000 Gent, Belgique.
| |
Collapse
|
175
|
Bellomo R, Ronco C. Renal Failure and Support. Crit Care 2005. [DOI: 10.1016/b978-0-323-02262-0.50033-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
176
|
Kang DG, Sohn EJ, Moon MK, Lee YM, Lee HS. Rehmannia glutinose Ameliorates Renal Function in the Ischemia/Reperfusion-Induced Acute Renal Failure Rats. Biol Pharm Bull 2005; 28:1662-7. [PMID: 16141536 DOI: 10.1248/bpb.28.1662] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present study was designed to examine whether aqueous extract of steamed root of Rehmannia glutinose (ARR) has an ameliorative effect on renal functional parameters in association with the expressions of aquaporin 2 (AQP 2), Na,K-ATPase, and heme oxygenase-1 (HO-1) in the ischemia-reperfusion induced acute renal failure (ARF) rats. Polyuria caused by down-regulation of renal AQP 2 in the ischemia-induced ARF rats was markedly restored by administration of ARR (200 mg/kg, p.o.) with restoring expression of AQP 2 in the kidney. The expressions of Na,K-ATPase alpha1 and beta1 subunits in the renal medullar and cortex of the ARF rats were also restored in the ARF rats by administration of ARR. On the other hand, administration of ARR lowered the renal expression of HO-1 up-regulated in rats with ischemia-induced ARF. The renal functional parameters including creatinine clearance, urinary sodium excretion, urinary osmolality, and solute-free reabsorption were also markedly restored in ischemia-ARF rats by administration of ARR. Taken together, these data indicate that RSR ameliorates renal defects in rats with ischemia-induced ARF.
Collapse
Affiliation(s)
- Dae Gill Kang
- Professional Graduate School of Oriental Medicine and Medicinal Resources Research Institute (MeRRI), Wonkwang University, Jeonbuk, Republic of Korea
| | | | | | | | | |
Collapse
|
177
|
Basile DP, Donohoe D, Cao X, Van Why SK. Resistance to ischemic acute renal failure in the Brown Norway rat: a new model to study cytoprotection. Kidney Int 2004; 65:2201-11. [PMID: 15149333 DOI: 10.1111/j.1523-1755.2004.00637.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND An in vivo model of intrinsic resistance to ischemia could be invaluable to define how specific pathways to injury or putative protectors from injury affect the severity of acute renal failure (ARF). The purpose of this study was to determine whether separate rat strains had differential sensitivity to renal ischemia, characterize the extent of protection, and begin to define differences in gene expression that might impact on the severity of ARF. METHODS The sensitivity to 45 minutes of renal ischemia in Sprague-Dawley rat (SD) was compared with 2 lines of Brown-Norway rats (BN/Mcw, BN/Hsd). Constitutive and inducible stress protein expression was compared between strains. RESULTS At 24 hours' reperfusion, SD rats had higher creatinine (3.4 mg/dL), elevated Na and water excretion, and proximal tubule necrosis. Both strains of BN rats were resistant to loss of renal function (Scr = 0.9 mg/dL at 24 hours' reflow) and had preserved renal morphology. BN rats had no redistribution of Na,K-ATPase into detergent-soluble cortical extracts found early (15 minutes) after ischemia in SD rats. Hsc73 expression did not differ between strains and was not induced by ischemia. Compared with SD, induction of Hsp25 and 72 by renal ischemia was blunted in both BN strains. Constitutive Hsp25 was higher in both BN-Mcw and BN-Hsd compared with SD rat kidney. Constitutive Hsp72 was significantly higher only in BN-Mcw kidneys. Immunohistochemistry showed baseline Hsp72 and 25 expression was increased in proximal tubules of BN-Mcw versus SD. CONCLUSION BN rat kidney is resistant to ischemic injury and provides a new model for studying cytoprotective mechanisms. Initial study of strain-specific gene expression suggests particular stress proteins are among the potential mechanisms contributing to protection against ARF.
Collapse
Affiliation(s)
- David P Basile
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | | | | | |
Collapse
|
178
|
Molina A, Ubeda M, Escribese MM, García-Bermejo L, Sancho D, Pérez de Lema G, Liaño F, Cabañas C, Sánchez-Madrid F, Mampaso F. Renal ischemia/reperfusion injury: functional tissue preservation by anti-activated {beta}1 integrin therapy. J Am Soc Nephrol 2004; 16:374-82. [PMID: 15590758 DOI: 10.1681/asn.2004070528] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Renal ischemia/reperfusion injury (IRI) is an important cause of acute renal failure. Cellular and molecular responses of the kidney to IRI are complex and not fully understood. beta1 integrins localize to the basal surface of tubular epithelium interacting with extracellular matrix components of the basal membrane, including collagen IV. Whether preservation of tubular epithelium integrity could be a therapeutic approach for IRI was assessed. The effects of HUTS-21 mAb administration, which recognizes an activation-dependent epitope of beta1 integrins, in a rat model of IRI were investigated. Preischemic HUTS-21 administration resulted in the preservation of renal functional and histopathologic parameters. Analyses of activated beta1 integrins expression and focal adhesion kinase phosphorylation suggest that its deactivation after IRI was prevented by HUTS-21 treatment. Moreover, HUTS-21 impaired the inflammatory response in vivo, as indicated by inhibition of proinflammatory mediators and the absence of infiltrating cells. Ex vivo adhesion assays using reperfused kidneys revealed that HUTS-21 induced a significant increase of epithelial cell attachment to collagen IV. In conclusion, the data provide evidence that HUTS-21 has a protective effect in renal IRI, preventing tubular epithelial cell detachment by preserving activated beta1 integrins functions.
Collapse
Affiliation(s)
- Ana Molina
- Hospital Ramón y Cajal, Departamento de Patología, Carretera de Colmenar, Km 9.1, 28034 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Basile DP. Rarefaction of peritubular capillaries following ischemic acute renal failure: a potential factor predisposing to progressive nephropathy. Curr Opin Nephrol Hypertens 2004; 13:1-7. [PMID: 15090853 DOI: 10.1097/00041552-200401000-00001] [Citation(s) in RCA: 165] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Long-term renal complications of acute renal failure have generally not been expected in patients that recover from acute renal failure. However, as the incidence of acute renal failure is rising, the incidence of long-term complications is likely to increase. As a corollary to ischemic acute renal failure, ischemic injury in the setting of transplant is a leading cause of delayed graft function. Unlike acute renal failure in native kidneys, delayed graft function is highly predictive of chronic nephropathy and organ failure. It is generally well accepted that acute reversible injuries mediated by ischemia render grafts susceptible toward future demise. The nature of the susceptibility that is conveyed to grafts following ischemic injury is not well understood. RECENT FINDINGS Evidence from animal models suggests that acute injury results in microvascular damage and vessel loss in the kidney, which, as opposed to tubular damage, is largely persistent. In addition, various studies of biopsies of renal transplants suggest that ischemia imposes an early and sustained loss in peritubular capillaries in the transplanted graft. The loss of peritubular capillaries has been associated with nephropathies of diverse etiologies and may represent a single, common pathway towards progressive damage. SUMMARY It is hypothesized that rarefaction of peritubular capillaries represents a critical event, following ischemic injury, that permanently alters renal function and predisposes patients to the development of chronic renal insufficiency. Factors that affect vascular reactivity or the structural dynamics of the kidney vascular system following injury may represent future treatment modalities following renal injury.
Collapse
Affiliation(s)
- David P Basile
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
180
|
Vergara L, Bao X, Cooper M, Bello-Reuss E, Reuss L. Gap-junctional hemichannels are activated by ATP depletion in human renal proximal tubule cells. J Membr Biol 2004; 196:173-84. [PMID: 14724743 DOI: 10.1007/s00232-003-0636-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2003] [Accepted: 10/09/2003] [Indexed: 11/21/2022]
Abstract
We present evidence suggesting that gap-junctional hemichannels (GJH) may be involved in acute ischemic injury of human renal proximal tubule cells (hPT cells). Two GJH, from neighboring cells, join to form an intercellular gap junction channel (GJC). Undocked GJH are permeable to hydrophilic molecules up to 1 kDa, and their opening can significantly alter cell homeostasis. Both GJC and GJH formed by connexin 43 (Cx43) are activated by dephosphorylation. Hence, we tested whether GJH activation during ATP depletion contributes to cell damage in renal ischemia. We found that hPT cells in primary culture express Cx43 (RT-PCR and Western-blot analysis) at the plasma membrane region (immunofluorescence). Divalent-cation removal or pharmacological ATP depletion increased cell loading with the hydrophilic dye 5/6 carboxy-fluorescein (CF, 376 Da) but not with fluorescein-labeled dextran (>1500 Da). Endocytosis and activation of P2X channels were experimentally ruled out. Several GJC blockers inhibited the loading elicited by PKC inhibition. Double labeling (CF and propidium iodide) showed that both Ca(2+) removal and ATP depletion increase the percentage of necrotic cells. Gadolinium reduced both the loading and the degree of necrosis during divalent-cation removal or ATP depletion. In conclusion, GJH activation may play an important role in the damage of human renal proximal tubule cells during ATP depletion. These studies are the first to provide evidence supporting a role of GJH in causing injury in epithelial cells in general and in renal-tubule cells in particular.
Collapse
Affiliation(s)
- L Vergara
- Department of Physiology and Biophysics, University of Texas Medical Branch, Galveston TX 77555-0641, USA
| | | | | | | | | |
Collapse
|
181
|
Abstract
Oxidative stress, which has been implicated in the pathogenesis of ischemic renal injury, degrades heme proteins, such as cytochrome P450, and causes the elevation in the level of cellular free heme, which can catalyze the formation of reactive oxygen species. Heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, is induced not only by its substrate, heme, but also by oxidative stress. In various models of oxidative tissue injuries, the induction of HO-1 confers protection on tissues from further damages by removing the prooxidant heme, or by virtue of the antioxidative, antiinflammatory, and/or antiapoptotic actions of one or more of the three products, i.e., carbon monoxide, biliverdin IXalpha, and iron by HO reaction. In contrast, the abrogation of HO-1 induction, or chemical inhibition of HO activity, abolishes its beneficial effect on the protection of tissues from oxidative damages. In this article, we review the protective role of HO-1 in renal ischemic injury, and its potential therapeutic applications. In addition, we summarize recent findings in the regulatory mechanism of ho-1 gene expression.
Collapse
Affiliation(s)
- Toru Takahashi
- Department of Anesthesiology and Resuscitology, Okayama University Medical School, Okayama, 700-8558, Japan.
| | | | | | | |
Collapse
|
182
|
Yoshino J, Monkawa T, Tsuji M, Hayashi M, Saruta T. Leukemia inhibitory factor is involved in tubular regeneration after experimental acute renal failure. J Am Soc Nephrol 2004; 14:3090-101. [PMID: 14638908 DOI: 10.1097/01.asn.0000101180.96787.02] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Leukemia inhibitory factor (LIF) is known to play a crucial role in the conversion of mesenchyme into epithelium during nephrogenesis. This study was carried out to test the hypothesis that LIF and LIF receptor (LIFR) are involved in the renal epithelial regeneration after acute renal failure. First, the authors investigated the spatiotemporal expression of LIF and LIFR in fetal and adult rat kidney. In developing kidney, LIF was expressed in the ureteric buds and LIFR was located in nephrogenic mesenchyme and the ureteric buds; in adult kidney, LIF and LIFR expression was confined to the collecting ducts. Next, the authors examined the expression of LIF and LIFR during the recovery phase after ischemia-reperfusion injury. Real-time PCR analysis revealed that LIF mRNA expression was significantly increased from day 1 to day 7 after reperfusion and that LIFR mRNA was upregulated from day 4 to day 14. Histologic analysis demonstrated that the increased expression of LIF mRNA and protein was most marked in the outer medulla, especially in the S3 segment of the proximal tubules. To elucidate the mitogenic role of LIF in the regeneration process, cultured rat renal epithelial (NRK 52E) cells were subjected to ATP depletion (an in vitro model of acute renal failure), and LIF expression was found to be enhanced during recovery after ATP depletion. Blockade of endogenous LIF with a neutralizing antibody significantly reduced the cell number and DNA synthesis during the recovery period. These results suggest that LIF participates in the regeneration process after tubular injury.
Collapse
Affiliation(s)
- Jun Yoshino
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | |
Collapse
|
183
|
Abstract
Inflammation plays a major role in the pathophysiology of acute renal failure resulting from ischemia. In this review, we discuss the contribution of endothelial and epithelial cells and leukocytes to this inflammatory response. The roles of cytokines/chemokines in the injury and recovery phase are reviewed. The ability of the mouse kidney to be protected by prior exposure to ischemia or urinary tract obstruction is discussed as a potential model to emulate as we search for pharmacologic agents that will serve to protect the kidney against injury. Understanding the inflammatory response prevalent in ischemic kidney injury will facilitate identification of molecular targets for therapeutic intervention.
Collapse
Affiliation(s)
- Joseph V Bonventre
- Medical Services, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, and Harvard-Massachusetts Institute of Technology, Division of Health Sciences and Technology, Charlestown, USA.
| | | |
Collapse
|
184
|
Ito T, Williams JD, Fraser D, Phillips AO. Hyaluronan attenuates transforming growth factor-beta1-mediated signaling in renal proximal tubular epithelial cells. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:1979-88. [PMID: 15161634 PMCID: PMC1615759 DOI: 10.1016/s0002-9440(10)63758-3] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Increased expression of hyaluronan (HA) has been associated with both acute renal injury and progressive renal disease, although the functional significance of this remains unclear. There is overwhelming evidence that transforming growth factor (TGF)-beta1 is critical to the development of progressive renal disease. Recent studies suggest an interaction between HA and TGF-beta signaling in cancer cell biology. The aim of this study was to examine the potential role of HA as a modulator of TGF-beta1 function in renal proximal tubular epithelial cells (PTC). Under resting conditions, co-localization of the principal receptor for HA, CD44, and both the TGF-beta type I and type II receptors was demonstrated by immunoprecipitation and western analysis and further confirmed by immunocytochemistry and confocal microscopy. Stimulation of PTC with TGF-beta1 led to increased synthesis of both type III and type IV collagen assessed by Western analysis. Addition of HA did not alter collagen synthesis, but abrogated TGF-beta1-mediated increase in type III and type IV collagen. This effect was blocked by the addition of a blocking antibody to CD44 and also by inhibition of MAP kinase kinase (MEK) activity. Furthermore HA decreased TGF-beta1 activation of a luciferase-SMAD responsive construct, and decreased translocation of SMAD4 into the cell nucleus. We have previously demonstrated an anti-migratory effect of TGF-beta1 in a scratch wounding model. As with HA antagonism of TGF-beta1 extracellular matrix generation, HA reduced the anti-migratory effect of TGF-beta1 in a CD44-dependent manner. In contrast to the effect of TGF-beta1 on collagen synthesis, which is SMAD-dependent, the anti-migratory effect of TGF-beta1 in this model is known to be dependent of activation of RhoA. In the presence of HA, TGF-beta1-mediated activation of RhoA was also abrogated in a CD44-dependent manner. The results suggest that co-localization of CD44 and TGF-beta receptors facilitate modulation of both SMAD and non-SMAD-dependent TGF-beta1-mediated events by HA. Our results therefore suggest that alteration of HA synthesis may represent an endogenous mechanism to limit renal injury.
Collapse
Affiliation(s)
- Takafumi Ito
- Institute of Nephrology, University of Wales College of Medicine, Cardiff, Wales
| | | | | | | |
Collapse
|
185
|
Shimizu MHM, Araujo M, Borges SMM, de Tolosa EMC, Seguro AC. Influence of age and vitamin E on post-ischemic acute renal failure. Exp Gerontol 2004; 39:825-30. [PMID: 15130677 DOI: 10.1016/j.exger.2004.02.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2003] [Revised: 02/18/2004] [Accepted: 02/20/2004] [Indexed: 11/18/2022]
Abstract
The aging process causes progressive deterioration in kidney structure and function. Aberrant generation of reactive oxygen species has been implicated in both age-related and ischemia-related tissue injury. Vitamin E (VE), one of the most powerful and effective exogenous antioxidants, prevents lipid peroxidation and protects against the effects of oxidative stress. The objective of this study was to determine the influence of age and VE on post-ischemic acute renal failure (ARF). Young adult, middle-aged and aged male Wistar rats were maintained on three different 30-day diets: Normal, VE absent and VE supplemented. On day 30, urinary protein and serum cholesterol and VE were measured. On day 31, rats were subjected to 60' clamping of the left renal artery plus right nephrectomy. Inulin clearance (InCl) was performed 48 h after renal ischemia. Malondialdehyde (MDA) was measured in the cortex of normal and 48-h post-ischemic kidneys. Urinary protein and serum cholesterol were higher in aged rats than in other rats. With aging, InCl decreased progressively. Vitamin E deficiency aggravated ARF. In middle-aged and aged rats, VE supplementation protected against ARF. In the absence of VE, MDA increased with age. In conclusion, our data suggest that ARF becomes more severe with age and that ischemia/reperfusion injury is exacerbated when antioxidant-scavenging ability of the kidney is impaired by VE deficiency. Supplementation with VE is essential for protecting aging kidneys against ischemic ARF.
Collapse
Affiliation(s)
- Maria Heloisa Massola Shimizu
- Laboratorio Pesquisa Basica LIM/12, Faculdade de Medicina USP, Av. Dr. Arnaldo, 455 3o. andar, sala 3310, CEP: 01246-000 Sao Paulo, SP, Brazil
| | | | | | | | | |
Collapse
|
186
|
Zahedi K, Wang Z, Barone S, Tehrani K, Yokota N, Petrovic S, Rabb H, Soleimani M. Identification of stathmin as a novel marker of cell proliferation in the recovery phase of acute ischemic renal failure. Am J Physiol Cell Physiol 2004; 286:C1203-11. [PMID: 15075220 DOI: 10.1152/ajpcell.00432.2003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ischemic renal injury can be classified into the initiation and extension phase followed by the recovery phase. The recovery phase is characterized by increased dedifferentiated and mitotic cells in the damaged tubules. Suppression subtractive hybridization was performed by using RNA from normal and ischemic kidneys to identify the genes involved in the physiological response to ischemia-reperfusion injury (IRI). The expression of stathmin mRNA increased by fourfold at 24 h of reperfusion. The stathmin mRNA did not increase in sodium-depleted animals or in animals with active, persistent injury secondary to cis-platinum. Immunofluorescent labeling demonstrated that the expression of stathmin increased dramatically at 48 h of reperfusion. Labeling with antibodies to stathmin and proliferating cell nuclear antigen (PCNA) indicates that the expression of stathmin was induced before the upregulation of PCNA and that all PCNA-positive cells expressed stathmin. Double immunofluorescent labeling demonstrated the colocalization of stathmin with vimentin, a marker of dedifferentiated cells. Stathmin expression was also significantly enhanced in acute tubular necrosis in humans. On the basis of its induction profile in IRI, the data indicating its enhanced expression in proliferating cells and regenerating organs, we propose that stathmin is a marker of dedifferentiated, mitotically active epithelial cells that may contribute to tubular regeneration and could prove useful in distinguishing the injury phase from recovery phase in IRI.
Collapse
Affiliation(s)
- Kamyar Zahedi
- Division of Nephrology and Hypertension, Children's Hospital Research Foundation, 3333 Burnet Ave., Cincinnati, OH 45229-3039, USA.
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Doi K, Suzuki Y, Nakao A, Fujita T, Noiri E. Radical scavenger edaravone developed for clinical use ameliorates ischemia/reperfusion injury in rat kidney. Kidney Int 2004; 65:1714-23. [PMID: 15086910 DOI: 10.1111/j.1523-1755.2004.00567.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one) is a potent scavenger of free radicals and has the antioxidant ability to inhibit lipid peroxidation. Its protective effect on brain ischemia has been shown. This study aimed to elucidate its possible therapeutic effects on renal oxidative stress in a rat ischemia/reperfusion model. METHODS Ischemic acute renal failure was induced by 45-minute clamp of the left renal artery followed by administration of edaravone. Renal function and pathologic findings were examined. The generation of free radicals was observed by the fluorogenic probe, dichlorodihydrofluorescein (CM-H(2)DCFDA), and lipid peroxidation was observed by immunochemical staining and Western analysis of 4-hydroxy-2-hexenal (HHE). The ability of edaravone to reduce in vitro oxidative stress was investigated using cultured human renal tubular cells (HKC-8) and the cell viability assay with Alamar blue. RESULTS Edaravone attenuated renal function and pathologic findings significantly. The protective effect of edaravone showed the dose-response was at the same level as that of dimethylthiourea. In addition, edaravone significantly reduced the generation of free radicals in the tubular cells indicated by CM-H(2)DCFDA. Immunochemical staining and Western analysis of HHE showed significant suppression of lipid peroxidation in edaravone-treated kidney. In HKC-8 cells loaded with CM-H(2)DCFDA, a marked elevation of fluorescence was observed after exposure to hydrogen peroxide (0.5 mmol/L, for 1 hour), which was reduced by edaravone. The cell viability assay with Alamar blue also showed the protective effect of edaravone. CONCLUSION Edaravone ameliorates renal ischemia/reperfusion injury by scavenging free radicals produced in renal tubular cells and inhibiting lipid peroxidation.
Collapse
Affiliation(s)
- Kent Doi
- Department of Nephrology and Endocrinology, University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
188
|
Kiew LV, Munavvar AS, Law CH, Azizan AN, Nazarina AR, Sidik K, Johns EJ. Effect of antisense oligodeoxynucleotides for ICAM-1 on renal ischaemia-reperfusion injury in the anaesthetised rat. J Physiol 2004; 557:981-9. [PMID: 15047774 PMCID: PMC1665139 DOI: 10.1113/jphysiol.2004.061788] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
An antisense oligodeoxynucleotide (As-ODN) to the 3' untranslated region of the mRNA sequence expressing the intracellular adhesion molecule-1 (ICAM-1) was employed to determine ICAM-1's role in renal ischaemia-reperfusion injury in the rat. Wistar-Kyoto rats receiving i.v. either lipofectin-As-ODN (As-ODN group), lipofectin-reverse ODN (Rv-ODN group) or lipofectin (ischaemia control group) 8 h prior to study were anaesthetized and subjected to 30 min of renal artery occlusion. Renal haemodynamic and excretory parameters were monitored before and after renal ischaemia. On termination of the study renal tissue was subjected to histological and Western blot analysis. Renal blood flow decreased in the 3 h post-ischaemia period in the ischaemia control and Rv-ODN groups, but was maintained in the As-ODN group. Glomerular filtration rate was depressed initially but gradually increased to 10% above basal levels in the ischaemia control and Rv-ODN groups, but was below basal levels (20%) in the As-ODN group. There was a three- to fourfold increase in sodium and water excretion following ischaemia in the ischaemia control and reverse-ODN groups but not in the As-ODN treated group. The As-ODN ameliorated the histological evidence of ischaemic damage and reduced ICAM-1 protein levels to a greater extent in the medulla than cortex. These observations suggested that in the post-ischaemic period afferent and efferent arteriolar tone was increased with a loss of reabsorptive capacity which was in part due to ICAM-1. The possibility arises that the action of ICAM-1 at vascular and tubular sites in the deeper regions of the kidney contributes to the ischaemia-reperfusion injury.
Collapse
Affiliation(s)
- Lik Voon Kiew
- Department of Pharmaceutical Sciences, University of Science Malaysia, Penang, Malaysia
| | | | | | | | | | | | | |
Collapse
|
189
|
Shao Y, Connors BA, Evan AP, Willis LR, Lifshitz DA, Lingeman JE. Morphological changes induced in the pig kidney by extracorporeal shock wave lithotripsy: nephron injury. ACTA ACUST UNITED AC 2004; 275:979-89. [PMID: 14533172 DOI: 10.1002/ar.a.10115] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
While shock wave lithotripsy (SWL) is known to cause significant damage to the kidney, little is known about the initial injury to cells along the nephron. In this study, one kidney in each of six juvenile pigs (6-7 weeks old) was treated with 1,000 shock waves (at 24 kV) directed at a lower pole calyx with an unmodified HM-3 lithotripter. Three pigs were utilized as sham-controls. Kidneys were fixed by vascular perfusion immediately after SWL or sham-SWL. Three of the treated kidneys were used to quantitate lesion size. Cortical and medullary samples for light (LM) and transmission electron microscopy (TEM) were taken from the focal zone for the shock waves (F2), the contralateral kidney, and the kidneys of sham-SWL pigs. Because preservation of the tissue occurred within minutes of SWL, the initial injury caused by the shock waves could be separated from secondary changes. No tissue damage was observed in contralateral sham-SWL kidneys, but treated kidneys showed signs of injury, with a lesion of 0.2% +/- 0.1% of renal volume. Intraparenchymal hemorrhage and injury to tubules was found at F2 in both the cortex and medulla of SWL-treated kidneys. Tubular injury was always associated with intraparenchymal bleeding, and the range of tissue injury included total destruction of tubules, focal cellular fragmentation, necrosis, cell vacuolization, and membrane blebbing. The initial injury caused by SWL was cellular fragmentation and necrosis. Cellular vacuolization, membrane blebbing, and disorganization of apical brush borders appear to be secondary changes related to hypoxia.
Collapse
Affiliation(s)
- Youzhi Shao
- Department of Histology, Jinzhou Medical College, Jinzhou, Liaoning Province, People's Republic of China
| | | | | | | | | | | |
Collapse
|
190
|
van Rossum GSAT, Drummen GPC, Verkleij AJ, Post JA, Boonstra J. Activation of cytosolic phospholipase A2 in Her14 fibroblasts by hydrogen peroxide: a p42/44MAPK-dependent and phosphorylation-independent mechanism. Biochim Biophys Acta Mol Cell Biol Lipids 2004; 1636:183-95. [PMID: 15164766 DOI: 10.1016/j.bbalip.2003.12.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2003] [Accepted: 12/03/2003] [Indexed: 11/24/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in the pathogenesis of diseases as well as various normal cellular processes. It has been suggested that ROS function as mediators of signal transduction, given that they can mimic growth factor-induced signaling. The ROS H2O2 has been reported to activate phospholipase A2 (PLA2) and, therefore, we investigated if and through which pathway ROS activate cytosolic PLA2 (cPLA2) in Her14 fibroblasts. cPLA2 was activated concentration-dependently by H2O2 in a transient manner. In addition, the lipophilic cumene hydroperoxide was shown to induce cPLA2 activity in the same manner. H2O2-induced cPLA2 activity in Her14 cells was partially phosphorylation-dependent, which was mediated through the Raf-MEK-p42/44(MAPK) pathway and occurred partially through a phosphorylation-independent mechanism. ROS can lead to changes in the (micro) viscosity of membranes due to the presence oxidized lipids, thereby increasing the substrate availability for cPLA2. In support of this, treatment of Her14 cells with H2O2 induced lipid peroxidation time-dependently as determined from degradation of lipid arachidonate and linoleate and the formation of aldehydic degradation products. Furthermore, H2O2 induced translocation of cPLA2 to the membrane fraction in a calcium-independent fashion, with a concomitant increase in cPLA2 activity. Collectively, the results suggest that oxidative stress-induced cPLA2 activity is partially phosphorylation-dependent and is further increased due to increased substrate availability by the action of ROS on membranes.
Collapse
Affiliation(s)
- Gerda S A T van Rossum
- Department of Molecular Cell Biology, Institute and Graduate School of Biomembranes, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
191
|
Ishibe S, Joly D, Zhu X, Cantley LG. Phosphorylation-dependent paxillin-ERK association mediates hepatocyte growth factor-stimulated epithelial morphogenesis. Mol Cell 2004; 12:1275-85. [PMID: 14636584 DOI: 10.1016/s1097-2765(03)00406-4] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Activation of the hepatocyte growth factor (HGF) receptor c-met results in the regulation of cell-matrix interactions, including the MAPK-dependent stimulation of epithelial cell morphogenesis. In the present study we demonstrate that HGF stimulates the localization of ERK to sites of cell-matrix interactions and that this is mediated by the tyrosine phosphorylation-dependent association of inactive ERK and the focal adhesion complex protein paxillin. In addition, paxillin was found to associate with the upstream MAP kinases Raf and MEK, resulting in a complex that can mediate localized ERK activation. Mutation of the ERK binding site in paxillin prevented HGF-stimulated ERK-paxillin association and eliminated HGF-induced cell spreading and branching process formation. These experiments reveal that paxillin-dependent ERK activation at sites of cell-matrix interaction is critical for HGF-stimulated epithelial morphogenesis.
Collapse
Affiliation(s)
- Shuta Ishibe
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
192
|
Ren JY, Chang CQ, Fung PCW, Shen JG, Chan FHY. Free radical EPR spectroscopy analysis using blind source separation. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2004; 166:82-91. [PMID: 14675823 DOI: 10.1016/j.jmr.2003.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
In this paper, we propose a novel approach for electron paramagnetic resonance (EPR) mixture spectra analysis based on blind source separation (BSS) technique. EPR spectrum of a free radical is often superimposed by overlapping spectra of other species. It is important and challenging to accurately identify and quantify the 'pure' spectra from such mixtures. In this study, an automated BSS method implementing independent component analysis is used to extract the components from mixed EPR spectra that contain overlapping components of different paramagnetic centers. To apply this method, there is no requirement to know the component spectra or the number of components in advance. The method is applied to analyze free radical EPR spectra which are collected from standard chemical system, cultured cell suspense, and ex vivo rat kidneys by spin trapping EPR technique. Results show that the BSS method proposed here is capable of identifying the component EPR spectra from mixtures with unknown compositions. The BSS technique can offer powerful aids in resolving spectral overlapping problems in general EPR spectroscopy analysis.
Collapse
Affiliation(s)
- J Y Ren
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong
| | | | | | | | | |
Collapse
|
193
|
Kobayashi H, Jo SK, Kawamoto S, Yasuda H, Hu X, Knopp MV, Brechbiel MW, Choyke PL, Star RA. Polyamine dendrimer-based MRI contrast agents for functional kidney imaging to diagnose acute renal failure. J Magn Reson Imaging 2004; 20:512-8. [PMID: 15332261 DOI: 10.1002/jmri.20147] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
PURPOSE To choose an efficacious renal functional MRI contrast agent to image early renal tubular damage. We synthesized and compared smaller polyamine dendrimer-based MRI contrast agents (<60 kD) that, unlike Gd-[DTPA], transiently accumulate in renal tubules and can be used to visualize renal structural and functional damage. MATERIALS AND METHODS Six dendrimer-based MRI contrast agents smaller than 60 kD were studied by high resolution dynamic micro-MRI and compared to Gd-[DTPA]-dimeglumine and Gadomer-17. The best agent, DAB-G2, was further tested in a mouse ischemia/reperfusion model to validate its efficacy. RESULTS Despite unequal renal clearance rates, all polyamine dendrimer agents visualized the renal functional anatomy of the mice better than Gd-[DTPA]-dimeglumine and Gadomer-17. DAB-G2 was excreted most rapidly, yet was able to visualize mild renal tubular injury very early after injury. CONCLUSION DAB-G2 was found to be the best candidate for functional kidney imaging and enabled early diagnosis of acute renal injury.
Collapse
Affiliation(s)
- Hisataka Kobayashi
- Metabolism Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Ichimura T, Hung CC, Yang SA, Stevens JL, Bonventre JV. Kidney injury molecule-1: a tissue and urinary biomarker for nephrotoxicant-induced renal injury. Am J Physiol Renal Physiol 2003; 286:F552-63. [PMID: 14600030 DOI: 10.1152/ajprenal.00285.2002] [Citation(s) in RCA: 457] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Nephrotoxicity is a common side effect of therapeutic interventions, environmental insults, and exposure to toxicants in the workplace. Although biomarkers for nephrotoxicity are available, they often lack sensitivity and are not specific as indicators of epithelial cell injury. Kidney injury molecule-1 (Kim-1) is a type 1 membrane protein with extracellular immunoglobulin and mucin domains. The mRNA and protein for Kim-1 are expressed at very low levels in normal rodent kidney, but expression increases dramatically after injury in proximal tubule epithelial cells in postischemic rodent kidney and in humans during ischemic acute renal failure. To evaluate the utility of Kim-1 as a biomarker for other types of renal injury, we analyzed tissue and urinary expression in response to three different types of nephrotoxicants in the rat: S-(1,1,2,2-tetrafluoroethyl)-l-cysteine (TFEC), folic acid, and cisplatin. Marked increases in Kim-1 expression were confirmed by immunoblotting in all three models. The protein was shown to be localized to the proximal tubule epithelial cell by immunofluorescence. Furthermore, Kim-1 protein was detected in urine of toxicant-treated rats. The temporal pattern of expression in response to TFEC is similar to the Kim-1 expression pattern in the postischemic kidney. In folic acid-treated kidneys, Kim-1 is clearly localized to the apical brush border of the well-differentiated proximal tubular epithelial cells. After folic acid treatment, expression of Kim-1 is present in the urine despite no significant increase in serum creatinine. Cisplatin treatment results in early detection of urinary Kim-1 protein and diffuse Kim-1 expression in S3 cells of the proximal tubule. Kim-1 can be detected in the tissue and urine on days 1 and 2 after cisplatin administration, occurring before an increase in serum creatinine. The upregulation of expression of Kim-1 and its presence in the urine in response to exposure to various types of nephrotoxicants suggest that this protein may serve as a general biomarker for tubular injury and repair processes.
Collapse
Affiliation(s)
- Takaharu Ichimura
- Medical Services, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
195
|
Vergara L, Bao X, Bello-Reuss E, Reuss L. Do connexin 43 gap-junctional hemichannels activate and cause cell damage during ATP depletion of renal-tubule cells? ACTA PHYSIOLOGICA SCANDINAVICA 2003; 179:33-8. [PMID: 12940936 DOI: 10.1046/j.1365-201x.2003.01198.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
UNLABELLED We review our evidence in favour of the hypothesis that gap-junctional hemichannels (GJH) are activated by depletion of adenosine triphosphate (ATP) in human renal proximal tubule cells in primary culture (hPT cells). Undocked GJH permit fluxes of ions and hydrophilic molecules up to 1 kDa, and thus their opening can cause alterations of cell composition conducive to cell damage. We show that hPT cells express connexin 43 (Cx43) (at the mRNA and protein levels) and that the protein is expressed on the plasma membrane. Moderate levels of pharmacological depletion of ATP increased plasma-membrane permeability, as shown by loading with the hydrophilic dye 5/6 carboxyfluorescein (CF, 376 Da) and other low-molecular weight dyes, but not with fluorescein-labelled dextran (>1500 Da). Roles for endocytosis and activation of purinergic-receptor channels were experimentally ruled out. Moderate ATP depletion also caused necrosis, assessed by cell permeabilization to propidium iodide. Prolonged exposure to gadolinium reduced both the dye loading and the necrosis induced by ATP depletion, i.e. it protected the cells. Cx43 overexpressed in insect cells, purified to homogeneity and reconstituted in proteoliposomes formed hemichannels that are activated by dephosphorylation of Ser368, a residue in a protein-kinase-C consensus phosphorylation sequence near the end of the C-terminal domain. CONCLUSIONS (1) ATP depletion of hPT cells induces a Gd3+-sensitive permeability of the plasma membrane to hydrophilic dyes with a cut-off size consistent with Cx43 GJH. (2) ATP depletion also increases the percentage of necrotic cells, an effect also reduced by Gd3+. (3) The experiments with purified Cx43 reconstituted in liposomes suggest that dephosphorylation of Ser368 is sufficient to activate GJH, although other mechanisms may be involved in some cells.
Collapse
Affiliation(s)
- L Vergara
- Department of Physiology and Biophysics, University of Texas Medical Branch, Galveston 77555-0641, USA
| | | | | | | |
Collapse
|
196
|
Bonventre JV, Weinberg JM. Recent advances in the pathophysiology of ischemic acute renal failure. J Am Soc Nephrol 2003; 14:2199-210. [PMID: 12874476 DOI: 10.1097/01.asn.0000079785.13922.f6] [Citation(s) in RCA: 546] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Joseph V Bonventre
- Renal Division, Brigham and Women's Hospital, and Department of Medicine, Harvard Medical School, and the Harvard--MIT, Division of Health Sciences and Technology, Charlestown, Massachusetts, USA.
| | | |
Collapse
|
197
|
Wald FA, Figueroa Y, Oriolo AS, Salas PJI. Membrane repolarization is delayed in proximal tubules after ischemia-reperfusion: possible role of microtubule-organizing centers. Am J Physiol Renal Physiol 2003; 285:F230-40. [PMID: 12709392 DOI: 10.1152/ajprenal.00024.2003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We have previously shown that microtubule-organizing centers (MTOCs) attach to the apical network of intermediate filaments (IFs) in epithelial cells in culture and in epithelia in vivo. Because that attachment is important for the architecture of microtubules (MTs) in epithelia, we analyzed whether chemical anoxia in LLC-PK1 and CACO-2 cells or unilateral ischemia-reperfusion in rat kidney (performed under fluorane anesthesia) had an effect on the binding and distribution of MTOCs. In culture, we found that chemical anoxia induces MTOC detachment from IFs by morphological and biochemical criteria. In reperfused rat proximal tubules, noncentrosomal MTOCs were fully detached from the cytoskeleton and scattered throughout the cytoplasm at 3 days after reperfusion, when brush borders were mostly reassembled. At that time, MTs were also fully reassembled but, as expected, lacked their normal apicobasal orientation. Two apical membrane markers expressed in S2 and S3 segments were depolarized at the same stage. At 8 days after reperfusion, membrane polarity, MTOCs, and MTs were back to normal. Na+-K+-ATPase was also found redistributed, not to the apical domain but rather to an intracellular compartment, as described by others (Alejandro VS, Nelson W, Huie P, Sibley RK, Dafoe D, Kuo P, Scandling JD Jr., and Myers BD. Kidney Int 48: 1308-1315, 1995). The prolonged depolarization of the apical membrane may have implications in the pathophysiology of acute renal failure.
Collapse
Affiliation(s)
- Flavia A Wald
- Department of Cell Biology and Anatomy, University of Miami School of Medicine, 1600 NW 10th Ave., Miami, FL 33136, USA
| | | | | | | |
Collapse
|
198
|
Abstract
The role of innate immunity in allograft injury is just beginning to become clear, and complement is probably one of a number of factors that are activated very early in the course of transplantation. Kidney transplantation into complement-inhibited rats reduces subsequent inflammation of the graft, probably as a result of reduction of ischemia reperfusion damage as well as diminution of immune mediated damage. Closer analysis of the role of locally synthesised components in mice has suggested that regional synthesis of complement proteins, in particular by the renal tubule, may play a more important role than circulating components. A marked effect on the antidonor T cell response may be explained by the triggering of complement receptors present on antigen presenting cells or T cells infiltrating the graft, or by a more direct effect of complement on the liaison between proximal tubule cells and T cells. Therapeutic control is likely to require a shift to a more targeted approach, directed at complement components produced in the extravascular tissue compartment.
Collapse
Affiliation(s)
- Steven H Sacks
- Guy's Hospital, King's College, London, Nephrology and Transplantation, London, United Kingdom.
| | | |
Collapse
|
199
|
Erkan E, Sakarcan A, Haklar G, Yalcin S. Thyroxine prevents reoxygenation injury in isolated proximal tubule cells. Pediatr Nephrol 2003; 18:636-43. [PMID: 12734748 DOI: 10.1007/s00467-003-1123-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2001] [Revised: 12/11/2002] [Accepted: 12/12/2002] [Indexed: 10/21/2022]
Abstract
Ischemia is characterized by cessation of blood flow and oxygen delivery to tissues that results in disruption of cellular structure and organelles. However, restoration of blood flow following ischemia causes reperfusion injury, characterized by further damage in the tissues mediated by reactive oxygen species. In the kidney, reactive oxygen molecules have been implicated in ischemic, toxic and immunological glomerular damage. Thyroxine has been shown to be cytoprotective in toxic and ischemic injury. Thyroxine's cytoprotective effect is postulated to be secondary to stimulation of intracellular ATP synthesis. However, the underlying mechanism of that beneficial effect remains to be investigated. In this study we investigated the effect of thyroxine (T4) on free oxygen radical production in an in vitro model of reperfusion injury. Free oxygen radical (FOR) levels were determined by a chemiluminescence method after freshly isolated rabbit proximal tubule cells were subjected to 15 min of anoxia followed by 45 min of reoxygenation. Reoxygenation injury resulted in a significant increase in FOR levels (P<0.0001). FOR levels were significantly lower in the group treated with thyroxine (P=0.01) and cells treated with thyroxine displayed better preservation of cellular structure. We conclude that thyroxine's cytoprotective effect might be via decreased synthesis of FOR, and thyroxine treatment may confer cytoprotection in renal conditions characterized by FOR-mediated injury.
Collapse
Affiliation(s)
- Elif Erkan
- Marmara University School of Medicine, Department of Pediatrics, Istanbul, Turkey.
| | | | | | | |
Collapse
|
200
|
Bonventre JV. Dedifferentiation and proliferation of surviving epithelial cells in acute renal failure. J Am Soc Nephrol 2003; 14 Suppl 1:S55-61. [PMID: 12761240 DOI: 10.1097/01.asn.0000067652.51441.21] [Citation(s) in RCA: 423] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In contrast to the heart or brain, the kidney can completely recover from an ischemic or toxic insult that results in cell death. During recovery from ischemia/reperfusion injury, surviving tubular epithelial cells dedifferentiate and proliferate, eventually replacing the irreversibly injured tubular epithelial cells and restoring tubular integrity. Repair of the kidney parallels kidney organogenesis in the high rate of DNA synthesis and apoptosis and in patterns of gene expression. As has been shown by proliferating cell nuclear antigen and 5-bromo 2'-deoxyuridine labeling studies and, in unpublished studies, by counting mitotic spindles identified by labeling with antitubulin antibody, the proliferative response is rapid and extensive, involving many of the remaining cells of the proximal tubule. This extensive proliferative capacity is interpreted to reflect the intrinsic ability of the surviving epithelial cell to adapt to the loss of adjacent cells by dedifferentiating and proliferating. Adhesion molecules likely play important roles in the regulation of renal epithelial cell migration, proliferation, and differentiation, as do cytokines and chemokines. Better understanding of all of the characteristics resulting in dedifferentiation and proliferation of the proximal tubule epithelial cell and cell-cell and cell-matrix interactions important for this repair function will lead to novel approaches to therapies designed to facilitate the processes of recovery in humans.
Collapse
Affiliation(s)
- Joseph V Bonventre
- Brigham and Women's Hospital, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Massachusetts, USA.
| |
Collapse
|