201
|
Cai QY, Lee H, Kim EJ, Moon H, Chang K, Rho J, Hong KS. Magnetic resonance imaging of superparamagnetic iron oxide-labeled macrophage infiltrates in acute-phase renal ischemia-reperfusion mouse model. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 8:365-73. [PMID: 21723242 DOI: 10.1016/j.nano.2011.06.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 05/29/2011] [Accepted: 06/21/2011] [Indexed: 11/25/2022]
Abstract
Macrophages play a key role in the initial pathogenesis of kidney ischemia-reperfusion (I-R) injury, but the mechanism of their spatial and temporal recruitment from circulation remains uncertain. This study aimed to evaluate the feasibility of magnetic resonance imaging (MRI) for detecting intravenously administered superparamagnetic iron oxide (SPIO)-labeled macrophages in an experimental renal I-R mouse model. Unilateral kidney I-R mice were imaged with a 4.7-T MRI scanner before and after administration of SPIO-labeled macrophages (RAW 264.7). On MR images, adoptive transfer of SPIO-labeled macrophages in the acute phase (1-2 days after I-R) caused a band-shaped signal-loss zone resulting from macrophage infiltrations, in the outer medullary region of injured kidneys. MRI detection of macrophages homing to an injured kidney may facilitate early detection and investigation of the pathogenesis of acute kidney injury and be a strategy for determining the treatment of acute renal failure. From the Clinical Editor: This study evaluated the feasibility of magnetic resonance imaging for detecting superparamagnetic iron oxide (SPIO)-labeled macrophages in a renal ischemia-reperfusion mouse model. Similar strategies in humans may facilitate early detection and stratification of acute kidney injury.
Collapse
Affiliation(s)
- Quan-Yu Cai
- Division of MR Research, Korea Basic Science Institute, Cheongwon, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
202
|
White LE, Hassoun HT. Inflammatory Mechanisms of Organ Crosstalk during Ischemic Acute Kidney Injury. Int J Nephrol 2011; 2012:505197. [PMID: 21826270 PMCID: PMC3118535 DOI: 10.4061/2012/505197] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Accepted: 03/10/2011] [Indexed: 01/22/2023] Open
Abstract
Acute kidney injury (AKI) is a common complication during inpatient hospitalization, and clinical outcomes remain poor despite advancements in renal replacement therapy. AKI in the setting of multiple organ failure (MOF) remains a formidable challenge to clinicians and incurs an unacceptably high mortality rate. Kidney ischemia-reperfusion injury (IRI) incites a proinflammatory cascade and releases cellular and soluble mediators with systemic implications for remote organ injury. Evidence from preclinical models cites mechanisms of organ crosstalk during ischemic AKI including the expression of cellular adhesion molecules, lymphocyte trafficking, release of proinflammatory cytokines and chemokines, and modification of the host innate and adaptive immune response systems. In this paper, the influence of kidney IRI on systemic inflammation and distant organ injury will be examined. Recent experimental data and evolving concepts of organ crosstalk during ischemic AKI will also be discussed in detail.
Collapse
Affiliation(s)
- Laura E White
- Department of Surgery, The Methodist Hospital and Research Institute, Houston, TX 77030, USA
| | | |
Collapse
|
203
|
Early interleukin 6 production by leukocytes during ischemic acute kidney injury is regulated by TLR4. Kidney Int 2011; 80:504-15. [PMID: 21633411 DOI: 10.1038/ki.2011.140] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Although leukocytes infiltrate the kidney during ischemic acute kidney injury (AKI) and release interleukin 6 (IL6), their mechanism of activation is unknown. Here, we tested whether Toll-like receptor 4 (TLR4) on leukocytes mediated this activation by interacting with high-mobility group protein B1 (HMGB1) released by renal cells as a consequence of ischemic kidney injury. We constructed radiation-induced bone marrow chimeras using C3H/HeJ and C57BL/10ScNJ strains of TLR4 (-/-) mice and their respective TLR4 (+/+) wild-type counterparts and studied them at 4 h after an ischemic insult. Leukocytes adopted from TLR4 (+/+) mice infiltrated the kidneys of TLR4 (-/-) mice, and TLR4 (-/-) leukocytes infiltrated the kidneys of TLR4 (+/+) mice but caused little functional renal impairment in each case. Maximal ischemic AKI required both radiosensitive leukocytes and radioresistant renal parenchymal and endothelial cells from TLR4 (+/+) mice. Only TLR4 (+/+) leukocytes produced IL6 in vivo and in response to HMGB1 in vitro. Thus, following infiltration of the injured kidney, leukocytes produce IL6 when their TLR4 receptors interact with HMGB1 released by injured renal cells. This underscores the importance of TLR4 in the pathogenesis of ischemic AKI.
Collapse
|
204
|
Hu H, Batteux F, Chéreau C, Kavian N, Marut W, Gobeaux C, Borderie D, Dinh-Xuan AT, Weill B, Nicco C. Clopidogrel protects from cell apoptosis and oxidative damage in a mouse model of renal ischaemia-reperfusion injury. J Pathol 2011; 225:265-75. [DOI: 10.1002/path.2916] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2010] [Revised: 03/31/2011] [Accepted: 04/04/2011] [Indexed: 12/31/2022]
|
205
|
The role of innate immunity in donor organ procurement. Semin Immunopathol 2011; 33:169-84. [DOI: 10.1007/s00281-011-0254-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Accepted: 01/13/2011] [Indexed: 12/18/2022]
|
206
|
Esposito E, Mondello S, Di Paola R, Mazzon E, Italiano D, Paterniti I, Mondello P, Aloisi C, Cuzzocrea S. Glutamine contributes to ameliorate inflammation after renal ischemia/reperfusion injury in rats. Naunyn Schmiedebergs Arch Pharmacol 2011; 383:493-508. [PMID: 21394482 DOI: 10.1007/s00210-011-0610-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Accepted: 02/01/2011] [Indexed: 01/05/2023]
Abstract
The aim of this study was to investigate the effects of glutamine in an in vivo rat model of renal ischemia/reperfusion (I/R) injury. Male Wistar rats underwent bilateral renal pedicle clamping for 45 min followed by reperfusion for 6 h. Glutamine (1.5 mg/kg) was administered intraperitoneally (i.p.) 15 min prior to reperfusion. Plasma concentrations of urea, creatinine, γ-glutamyl transferase (γ-GT), and aspartate aminotransferase (AST) were measured for the assessment of renal function and reperfusion injury. Markers of oxidative stress, expression of the pro-inflammatory mediators inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), AT-1 expression, and changes in the oxidative stress-sensitive nuclear factor kappa B (NF-κB) signaling pathway were measured to investigate whether glutamine can reduce the renal dysfunction. Kidney myeloperoxidase (MPO) activity and malondialdehyde (MDA) levels were measured for assessment of polymorphonuclear (PMN) cell infiltration and lipid peroxidation, respectively. Renal sections were used for histologic grading of renal injury and for immunohistochemical localization of nitrotyrosine and poly(ADP-ribose) synthetase (PARS). In vivo, glutamine significantly reduced the increase in urea, creatinine, γ-GT, AST, produced by renal ischemia/reperfusion (I/R), suggesting an improvement in both renal function and injury. Glutamine significantly reduced iNOS and NF-κB, kidney MPO activity and MDA levels, indicating a reduction in PMN infiltration and lipid peroxidation, respectively. Glutamine reduced the histological evidence of renal damage associated with I/R and caused a substantial reduction in the staining for nitrotyrosine and PARS, suggesting reduced nitrosative and oxidative stress. Moreover, glutamine attenuated the reduction of COX-2 expression and prevented the increased AT-1 expression after I/R. Our results suggest that glutamine reduces the renal dysfunction and injury associated with I/R of the kidney.
Collapse
Affiliation(s)
- Emanuela Esposito
- Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Centro Neurolesi Bonino-Pulejo, Messina, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
207
|
Rong S, Park JK, Kirsch T, Yagita H, Akiba H, Boenisch O, Haller H, Najafian N, Habicht A. The TIM-1:TIM-4 pathway enhances renal ischemia-reperfusion injury. J Am Soc Nephrol 2011; 22:484-95. [PMID: 21355054 DOI: 10.1681/asn.2010030321] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
CD4+ T cells contribute to the pathogenesis of ischemia-reperfusion injury, which is the primary cause of delayed graft failure after kidney transplantation. The TIM-1:TIM-4 pathway participates in the activation/differentiation of CD4+ T cells, suggesting that it may modulate ischemia-reperfusion injury. Here, we studied the role of TIM-1 in a murine uninephrectomized renal ischemia-reperfusion injury model. Blocking the TIM-1:TIM-4 pathway with an antagonistic monoclonal antibody protected renal function and diminished reperfusion injury resulting from 30 minutes of ischemia. Histologic examination showed significantly less evidence of renal damage as evidenced by diminished tubular necrosis, preservation of the brush border, fewer cast formations, and less tubular dilation. Blocking TIM-1 also reduced the number of apoptotic cells and diminished local inflammation within ischemic kidneys, the latter shown by decreased recruitment of macrophages, neutrophils, and CD4+ T cells and by reduced local production of proinflammatory cytokines. Furthermore, TIM-1 blockade significantly improved survival after ischemia-reperfusion injury. Taken together, these data suggest that the TIM-1:TIM-4 pathway enhances injury after renal ischemia-reperfusion injury and may be a therapeutic target.
Collapse
Affiliation(s)
- Song Rong
- Transplant Center, University Hospital Munich, Marchioninistrasse 15, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Collino M, Benetti E, Miglio G, Castiglia S, Rosa AC, Aragno M, Thiemermann C, Fantozzi R. Peroxisome proliferator-activated receptor β/δ agonism protects the kidney against ischemia/reperfusion injury in diabetic rats. Free Radic Biol Med 2011; 50:345-53. [PMID: 21047550 DOI: 10.1016/j.freeradbiomed.2010.10.710] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 10/06/2010] [Accepted: 10/27/2010] [Indexed: 11/26/2022]
Abstract
Diabetes is an important risk factor for ischemic acute kidney injury, whose pharmacological treatment remains an unmet medical need. The peroxisome proliferator-activated receptor (PPAR) β/δ is highly expressed in the kidney, although its role has not yet been elucidated. Here, we used an in vivo model of renal ischemia/reperfusion (I/R) in streptozotocin-induced diabetic rats (i) to evaluate whether diabetes increases kidney susceptibility to I/R injury and (ii) to investigate the effects of PPARβ/δ activation. The degree of renal injury (1h ischemia/6h reperfusion) was significantly increased in diabetic rats compared with nondiabetic littermates. PPARβ/δ expression was increased after I/R, with the highest levels in diabetic rats. Administration of the selective PPARβ/δ agonist GW0742 attenuated the renal dysfunction, leukocyte infiltration, and formation of interleukin-6 and tumor necrosis factor-α. These effects were accompanied by an increased expression of the suppressor of cytokine signaling (SOCS)-3, which plays a critical role in the cytokine-activated signaling pathway. The beneficial effects of GW0742 were attenuated by the selective PPARβ/δ antagonist GSK0660. Thus, we report herein that PPARβ/δ activation protects the diabetic kidney against I/R injury by a mechanism that may involve changes in renal expression of SOCS-3 resulting in a reduced local inflammatory response.
Collapse
Affiliation(s)
- Massimo Collino
- Department of Anatomy, Pharmacology, and Forensic Medicine, University of Turin, Turin, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
209
|
Barrera-Chimal J, Pérez-Villalva R, Cortés-González C, Ojeda-Cervantes M, Gamba G, Morales-Buenrostro LE, Bobadilla NA. Hsp72 is an early and sensitive biomarker to detect acute kidney injury. EMBO Mol Med 2010; 3:5-20. [PMID: 21204265 PMCID: PMC3401998 DOI: 10.1002/emmm.201000105] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 11/01/2010] [Accepted: 11/08/2010] [Indexed: 12/22/2022] Open
Abstract
This study was designed to assess whether heat shock protein Hsp72 is an early and sensitive biomarker of acute kidney injury (AKI) as well as to monitor a renoprotective strategy. Seventy-two Wistar rats were divided into six groups: sham-operated and rats subjected to 10, 20, 30, 45 and 60 min of bilateral ischemia (I) and 24 h of reperfusion (R). Different times of reperfusion (3, 6, 9, 12, 18, 24, 48, 72, 96 and 120 h) were also evaluated in 30 other rats subjected to 30 min of ischemia. Hsp72 messenger RNA (mRNA) and protein levels were determined in both kidney and urine. Hsp72-specificity as a biomarker to assess the success of a renoprotective intervention was evaluated in rats treated with different doses of spironolactone before I/R. Renal Hsp72 mRNA and protein, as well as urinary Hsp72 levels, gradually increased relative to the extent of renal injury induced by different periods of ischemia quantified by histomorphometry as a benchmark of kidney damage. Urinary Hsp72 increased significantly after 3 h and continued rising until 18 h, followed by restoration after 120 h of reperfusion in accord with histopathological findings. Spironolactone renoprotection was associated with normalization of urinary Hsp72 levels. Accordingly, urinary Hsp72 was significantly increased in patients with clinical AKI before serum creatinine elevation. Our results show that urinary Hsp72 is a useful biomarker for early detection and stratification of AKI. In addition, urinary Hsp72 levels are sensitive enough to monitor therapeutic interventions and the degree of tubular recovery following an I/R insult.
Collapse
Affiliation(s)
- Jonatan Barrera-Chimal
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | | | | | | | | |
Collapse
|
210
|
Crikis S, Lu B, Murray-Segal LM, Selan C, Robson SC, d’Apice AJF, Nandurkar HH, Cowan PJ, Dwyer KM. Transgenic overexpression of CD39 protects against renal ischemia-reperfusion and transplant vascular injury. Am J Transplant 2010; 10:2586-95. [PMID: 20840479 PMCID: PMC5472986 DOI: 10.1111/j.1600-6143.2010.03257.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The vascular ectonucleotidases CD39[ENTPD1 (ectonucleoside triphosphate diphosphohydrolase-1), EC 3.6.1.5] and CD73[EC 3.1.3.5] generate adenosine from extracellular nucleotides. CD39 activity is critical in determining the response to ischemia-reperfusion injury (IRI), and CD39 null mice exhibit heightened sensitivity to renal IRI. Adenosine has multiple mechanisms of action in the vasculature including direct endothelial protection, antiinflammatory and antithrombotic effects and is protective in several models of IRI. Mice transgenic for human CD39 (hCD39) have increased capacity to generate adenosine. We therefore hypothesized that hCD39 transgenic mice would be protected from renal IRI. The overexpression of hCD39 conferred protection in a model of warm renal IRI, with reduced histological injury, less apoptosis and preserved serum creatinine and urea levels. Benefit was abrogated by pretreatment with an adenosine A2A receptor antagonist. Adoptive transfer experiments showed that expression of hCD39 on either the vasculature or circulating cells mitigated IRI. Furthermore, hCD39 transgenic kidneys transplanted into syngeneic recipients after prolonged cold storage performed significantly better and exhibited less histological injury than wild-type control grafts. Thus, systemic or local strategies to promote adenosine generation and signaling may have beneficial effects on warm and cold renal IRI, with implications for therapeutic application in clinical renal transplantation.
Collapse
Affiliation(s)
- S. Crikis
- Immunology Research Centre, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia,Department of Medicine, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
| | - B. Lu
- Immunology Research Centre, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
| | - L. M. Murray-Segal
- Immunology Research Centre, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
| | - C. Selan
- Immunology Research Centre, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
| | - S. C. Robson
- Liver Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - A. J. F. d’Apice
- Immunology Research Centre, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia,Department of Medicine, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia,Corresponding author: Anthony JF d’Apice,
| | - H. H. Nandurkar
- Immunology Research Centre, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia,Department of Medicine, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
| | - P. J. Cowan
- Immunology Research Centre, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia,Department of Medicine, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
| | - K. M. Dwyer
- Immunology Research Centre, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia,Department of Medicine, The University of Melbourne, St. Vincent’s Hospital, Fitzroy, Victoria, 3065, Australia
| |
Collapse
|
211
|
Facio FN, Sena AA, Araújo LP, Mendes GE, Castro I, Luz MAM, Yu L, Oliani SM, Burdmann EA. Annexin 1 mimetic peptide protects against renal ischemia/reperfusion injury in rats. J Mol Med (Berl) 2010; 89:51-63. [PMID: 20953576 DOI: 10.1007/s00109-010-0684-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 09/01/2010] [Accepted: 09/18/2010] [Indexed: 10/18/2022]
Abstract
Inflammation is currently recognized as a key mechanism in the pathogenesis of renal ischemia-reperfusion (I/R) injury. The importance of infiltrating neutrophil, lymphocytes, and macrophage in this kind of injury has been assessed with conflicting results. Annexin 1 is a protein with potent neutrophil anti-migratory activity. In order to evaluate the effects of annexin A1 on renal I/R injury, uninephrectomized rats received annexin A1 mimetic peptide Ac2-26 (100 μg) or vehicle before 30 min of renal artery clamping and were compared to sham surgery animals. Annexin A1 mimetic peptide granted a remarkable protection against I/R injury, preventing glomerular filtration rate and urinary osmolality decreases and acute tubular necrosis development. Annexin A1 infusion aborted neutrophil extravasation and attenuated macrophage infiltration but did not prevent tissue lymphocyte traffic. I/R increased annexin A1 expression (assessed by transmission electron microscopy) in renal epithelial cells, which was attenuated by exogenous annexin A1 infusion. Additionally, annexin A1 reduced I/R injury in isolated proximal tubules suspension. Annexin A1 protein afforded striking functional and structural protection against renal I/R. These results point to an important role of annexin A1 in the epithelial cells defense against I/R injury and indicate that neutrophils are key mediators for the development of tissue injury after renal I/R. If these results were confirmed in clinical studies, annexin A1 might emerge as an important tool to protect against I/R injury in renal transplantation and in vascular surgery.
Collapse
Affiliation(s)
- Fernando N Facio
- Division of Nephrology, São José do Rio Preto Medical School, Av. Brigadeiro Faria Lima 5416, São José do Rio Preto, São Paulo, 15090-000, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Chen J, John R, Richardson JA, Shelton JM, Zhou XJ, Wang Y, Wu QQ, Hartono JR, Winterberg PD, Lu CY. Toll-like receptor 4 regulates early endothelial activation during ischemic acute kidney injury. Kidney Int 2010; 79:288-99. [PMID: 20927041 DOI: 10.1038/ki.2010.381] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Ischemic acute kidney injury (AKI) triggers an inflammatory response which exacerbates injury that requires increased expression of endothelial adhesion molecules. To study this further, we used in situ hybridization, immunohistology, and isolated endothelial cells, and found increased Toll-like receptor 4 (TLR4) expression on endothelial cells of the vasa rectae of the inner stripe of the outer medulla of the kidney 4 h after reperfusion. This increase was probably due to reactive oxygen species, known to be generated early during ischemic AKI, because the addition of hydrogen peroxide increased TLR4 expression in MS1 microvascular endothelial cells in vitro. Endothelial TLR4 may regulate adhesion molecule (CD54 and CD62E) expression as they were increased on endothelia of wild-type but not TLR4 knockout mice in vivo. Further, the addition of high-mobility group protein B1, a TLR4 ligand released by injured cells, increased adhesion molecule expression on endothelia isolated from wild-type but not TLR4 knockout mice. TLR4 was localized to proximal tubules in the cortex and outer medulla after 24 h of reperfusion. Thus, at least two different cell types express TLR4, each of which contributes to renal injury by temporally different mechanisms during ischemic AKI.
Collapse
Affiliation(s)
- Jianlin Chen
- Department of Internal Medicine Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8856, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
213
|
Guo H. Acute Allograft Renal Failure With Marked Hyperuricemia Developing During Mizoribine Administration: A Case Report With Review of the Literature. Transplant Proc 2010; 42:2804-7. [DOI: 10.1016/j.transproceed.2010.05.148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 01/28/2010] [Accepted: 05/12/2010] [Indexed: 11/28/2022]
|
214
|
Garwood S. Cardiac surgery-associated acute renal injury: new paradigms and innovative therapies. J Cardiothorac Vasc Anesth 2010; 24:990-1001. [PMID: 20702119 DOI: 10.1053/j.jvca.2010.05.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Indexed: 01/02/2023]
Affiliation(s)
- Susan Garwood
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06520-8051, USA.
| |
Collapse
|
215
|
Tadagavadi RK, Wang W, Ramesh G. Netrin-1 Regulates Th1/Th2/Th17 Cytokine Production and Inflammation through UNC5B Receptor and Protects Kidney against Ischemia–Reperfusion Injury. THE JOURNAL OF IMMUNOLOGY 2010; 185:3750-8. [DOI: 10.4049/jimmunol.1000435] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
216
|
Zhang ZX, Shek K, Wang S, Huang X, Lau A, Yin Z, Sun H, Liu W, Garcia B, Rittling S, Jevnikar AM. Osteopontin Expressed in Tubular Epithelial Cells Regulates NK Cell-Mediated Kidney Ischemia Reperfusion Injury. THE JOURNAL OF IMMUNOLOGY 2010; 185:967-973. [DOI: 10.4049/jimmunol.0903245] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Abstract
Renal ischemia reperfusion injury (IRI) occurs after reduced renal blood flow and is a major cause of acute injury in both native and transplanted kidneys. Studies have shown diverse cell types in both the innate and the adaptive immune systems participate in kidney IRI as dendritic cells, macrophages, neutrophils, B cells, CD4+ NK+ cells, and CD4+ T cells all contribute to this form of injury. Recently, we have found that NK cells induce apoptosis in tubular epithelial cells (TECs) and also contribute to renal IRI. However, the mechanism of NK cell migration and activation during kidney IRI remains unknown. In this study, we have identified that kidney TECs express a high level of osteopontin (OPN) in vitro and in vivo. C57BL/6 OPN-deficient mice have reduced NK cell infiltration with less tissue damage compared with wild-type C57BL/6 mice after ischemia. OPN can directly activate NK cells to mediate TEC apoptotic death and can also regulate chemotaxis of NK cells to TECs. Taken together, our study’s results indicate that OPN expression by TECs is an important factor in initial inflammatory responses that involves NK cells activity in kidney IRI. Inhibiting OPN expression at an early stage of IRI may be protective and preserve kidney function after transplantation.
Collapse
Affiliation(s)
- Zhu-Xu Zhang
- *The Multi-Organ Transplant Program, London Health Sciences Centre
- †Department of Medicine,
- ‡Department of Pathology, and
- §Lawson Health Research Institute, London, Ontario, Canada; and
| | - Kelvin Shek
- ¶Department of Microbiology and Immunology, University of Western Ontario
| | - Shuang Wang
- *The Multi-Organ Transplant Program, London Health Sciences Centre
- §Lawson Health Research Institute, London, Ontario, Canada; and
| | - Xuyan Huang
- *The Multi-Organ Transplant Program, London Health Sciences Centre
- §Lawson Health Research Institute, London, Ontario, Canada; and
| | - Arthur Lau
- *The Multi-Organ Transplant Program, London Health Sciences Centre
- ‡Department of Pathology, and
| | - Ziqin Yin
- *The Multi-Organ Transplant Program, London Health Sciences Centre
- §Lawson Health Research Institute, London, Ontario, Canada; and
| | - Hongtao Sun
- *The Multi-Organ Transplant Program, London Health Sciences Centre
- §Lawson Health Research Institute, London, Ontario, Canada; and
| | - Weihua Liu
- *The Multi-Organ Transplant Program, London Health Sciences Centre
- §Lawson Health Research Institute, London, Ontario, Canada; and
| | - Bertha Garcia
- *The Multi-Organ Transplant Program, London Health Sciences Centre
- †Department of Medicine,
- ‡Department of Pathology, and
- §Lawson Health Research Institute, London, Ontario, Canada; and
| | - Susan Rittling
- ‖Department of Cytokine Biology, The Forsyth Institute, Boston, MA 02115
| | - Anthony M. Jevnikar
- *The Multi-Organ Transplant Program, London Health Sciences Centre
- †Department of Medicine,
- ‡Department of Pathology, and
- §Lawson Health Research Institute, London, Ontario, Canada; and
- ¶Department of Microbiology and Immunology, University of Western Ontario
| |
Collapse
|
217
|
Oliveira GMD, Masuda MO, Rocha NN, Schor N, Hooper CS, Araújo-Jorge TCD, Henriques-Pons A. Absence of Fas-L aggravates renal injury in acute Trypanosoma cruzi infection. Mem Inst Oswaldo Cruz 2010; 104:1063-71. [PMID: 20140366 DOI: 10.1590/s0074-02762009000800002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Accepted: 10/13/2009] [Indexed: 01/28/2023] Open
Abstract
Trypanosoma cruzi infection induces diverse alterations in immunocompetent cells and organs, myocarditis and congestive heart failure. However, the physiological network of disturbances imposed by the infection has not been addressed thoroughly. Regarding myocarditis induced by the infection, we observed in our previous work that Fas-L-/- mice (gld/gld) have very mild inflammatory infiltration when compared to BALB/c mice. However, all mice from both lineages die in the early acute phase. Therefore, in this work we studied the physiological connection relating arterial pressure, renal function/damage and cardiac insufficiency as causes of death. Our results show that a broader set of dysfunctions that could be classified as a cardio/anaemic/renal syndrome is more likely responsible for cardiac failure and death in both lineages. However, gld/gld mice had very early glomerular deposition of IgM and a more intense renal inflammatory response with reduced renal filtration, which is probably responsible for the premature death in the absence of significant myocarditis in gld/gld.
Collapse
|
218
|
Lung inflammation is induced by renal ischemia and reperfusion injury as part of the systemic inflammatory syndrome. Inflamm Res 2010; 59:861-9. [PMID: 20396927 DOI: 10.1007/s00011-010-0198-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2009] [Revised: 03/27/2010] [Accepted: 03/31/2010] [Indexed: 10/19/2022] Open
Abstract
INTRODUCTION Ischemia and reperfusion injury (IRI) are mainly caused by leukocyte activation, endothelial dysfunction and production of reactive oxygen species. Moreover, IRI can lead to a systemic response affecting distant organs, such as the lungs. AIM The objective was to study the pulmonary inflammatory systemic response after renal IRI. METHODS Male C57Bl/6 mice were subjected to 45 min of bilateral renal ischemia, followed by 4, 6, 12, 24 and 48 h of reperfusion. Blood was collected to measure serum creatinine and cytokine concentrations. Bronchoalveolar lavage fluid (BALF) was collected to determine the number of cells and PGE(2) concentration. Expressions of iNOS and COX-2 in lung were determined by Western blot. Gene analyses were quantified by real time PCR. RESULTS Serum creatinine increased in the IRI group compared to sham mainly at 24 h after IRI (2.57 +/- 0.16 vs. 0.43 +/- 0.07, p < 0.01). The total number of cells in BAL fluid was higher in the IRI group in comparison with sham, 12 h (100 x 10(4) +/- 15.63 vs. 18.1 x 10(4) +/- 10.5, p < 0.05) 24 h (124 x 10(4) +/- 8.94 vs. 23.2 x 10(4) +/- 3.5, p < 0.05) and 48 h (79 x 10(4) +/- 15.72 vs. 22.2 x 10(4) +/- 4.2, p < 0.05), mainly by mononuclear cells and neutrophils. Pulmonary COX-2 and iNOS were up-regulated in the IRI group. TNF-alpha, IL-1beta, MCP-1, KC and IL-6 mRNA expression were up-regulated in kidney and lungs 24 h after renal IRI. ICAM-1 mRNA was up-regulated in lungs 24 h after renal IRI. Serum TNF-alpha, IL-1beta and MCP-1 and BALF PGE(2) concentrations were increased 24 h after renal IRI. CONCLUSION Renal IRI induces an increase of cellular infiltration, up-regulation of COX-2, iNOS and ICAM-1, enhanced chemokine expression and a Th1 cytokine profile in lung demonstrating that the inflammatory response is indeed systemic, possibly leading to an amplification of renal injury.
Collapse
|
219
|
Depletion of kidney CD11c+ F4/80+ cells impairs the recovery process in ischaemia/reperfusion-induced acute kidney injury. Nephrol Dial Transplant 2010; 25:2908-21. [DOI: 10.1093/ndt/gfq183] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
220
|
Ekor M, Emerole GO, Farombi EO. Phenolic extract of soybean (Glycine max) attenuates cisplatin-induced nephrotoxicity in rats. Food Chem Toxicol 2010; 48:1005-12. [PMID: 20109512 DOI: 10.1016/j.fct.2009.12.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 11/20/2009] [Accepted: 12/24/2009] [Indexed: 01/13/2023]
Abstract
The present study investigated the modulatory role of phenolic extract of soybean (PESB) in a rat model of nephrotoxic acute renal failure induced by cisplatin. Cisplatin (2 mg/kg/day) was administered to the rats for 5 days and the animals were pretreated with PESB (250-1000 mg/kg). Blood urea nitrogen reduced by 49.8% and 59.0%, serum creatinine by 34.7% and 62.1% and urinary N-acetyl-beta-D-glucosaminidase also decreased by 37.7% and 49.2% following treatment with 250- and 500-mg/kg doses of the extract respectively in the cisplatin-treated rats. The extract also significantly increased renal myeloperoxidase activity by 26.8% and 40.6% at these doses. PESB also decreased renal xanthine oxidase activity and serum nitrate/nitrite in the cisplatin-treated rats. In addition, PESB significantly attenuated the marked renal oxidative damage that accompanied cisplatin treatment. The extract improved liver histology and significantly increased the activities of the antioxidant enzymes measured [superoxide dismutase, catalase, glutathione-S-transferase], prevented glutathione depletion and decreased malondialdehyde level following cisplatin treatment. Furthermore, cisplatin-induced decrease in the activities of glucose-6-phosphatase and 5'-nucleotidase in these rats was attenuated only at 250 mg/kg dose of the extract. We concluded therefore that PESB via antioxidant and possibly anti-inflammatory actions offered protective benefit against cisplatin-mediated acute toxic injury to the kidney.
Collapse
Affiliation(s)
- Martins Ekor
- Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | | |
Collapse
|
221
|
Zhou W, Guan Q, Kwan CCH, Chen H, Gleave ME, Nguan CYC, Du C. Loss of clusterin expression worsens renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 2010; 298:F568-78. [DOI: 10.1152/ajprenal.00399.2009] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Prevention of ischemia-reperfusion injury (IRI) is a challenge in clinical care of the patients with kidney transplants or acute kidney injury, and understanding of the intrinsic mechanisms of resistance to injury in the kidney will lead to a novel therapy. Clusterin, a secreted glycoprotein, is an antiapoptotic protein in cancer cells. Our study is to investigate the role of clusterin in renal IRI. Renal IRI in mice was induced by clamping renal vein and artery for 45 or 50 min at 32°C. Apoptosis of renal tubular epithelial cells (TECs) was determined by FACS analysis. Clusterin expression was examined by Western blot or immunohistochemistry. Here, we showed that clusterin protein was induced in TECs following IRI, and more tubules expressed clusterin in the kidneys following ischemia at higher temperatures. In human proximal TEC HKC-8 cultures, clusterin was upregulated by removal of serum and growth factors in medium and was downregulated by TNF-α-IFN-γ mixture. The levels of clusterin were positively correlated with cell survival in these conditions. Knockdown or knockout of clusterin expression enhanced the sensitivity of TECs to apoptosis. In experimental models of renal IRI, deficiency in clusterin expression worsened the injury, as indicated by a significant increase in renal tissue damage with higher levels of serum creatinine and blood urea nitrogen and by a poorer recovery from the injury in clusterin-deficient mice compared with wild-type mice. Our data indicate that the reduction of inducible expression of clusterin results in an increase in TEC apoptosis in the cultures and renders mice susceptibility to IRI, implying a protective role of clusterin in kidney injury.
Collapse
Affiliation(s)
- Wenjun Zhou
- Department of Urologic Sciences, University of British Columbia, Vancouver
- Department of Surgery, University of Montreal, Montreal, Quebec, Canada
| | - Qiunong Guan
- Department of Urologic Sciences, University of British Columbia, Vancouver
| | - Chris C. H. Kwan
- Department of Urologic Sciences, University of British Columbia, Vancouver
| | - Huifang Chen
- Department of Surgery, University of Montreal, Montreal, Quebec, Canada
| | - Martin E. Gleave
- Department of Urologic Sciences, University of British Columbia, Vancouver
| | - Christopher Y. C. Nguan
- Department of Urologic Sciences, University of British Columbia, Vancouver
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia; and
| | - Caigan Du
- Department of Urologic Sciences, University of British Columbia, Vancouver
- Immunity and Infection Research Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia; and
| |
Collapse
|
222
|
Mediators of inflammation in acute kidney injury. Mediators Inflamm 2010; 2009:137072. [PMID: 20182538 PMCID: PMC2825552 DOI: 10.1155/2009/137072] [Citation(s) in RCA: 355] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 11/18/2009] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) remains to be an independent risk factor for mortality and morbidity. Inflammation is now believed to play a major role in the pathopathophysiology of AKI. It is hypothesized that in ischemia, sepsis and nephrotoxic models that the initial insult results in morphological and/or functional changes in vascular endothelial cells and/or in tubular epithelium. Then, leukocytes including neutrophils, macrophages, natural killer cells, and lymphocytes infiltrate into the injured kidneys. The injury induces the generation of inflammatory mediators like cytokines and chemokines by tubular and endothelial cells which contribute to the recruiting of leukocytes into the kidneys. Thus, inflammation has an important role in the initiation and extension phases of AKI. This review will focus on the mediators of inflammation contributing to the pathogenesis of AKI.
Collapse
|
223
|
Mitchell JR, Verweij M, Brand K, van de Ven M, Goemaere N, van den Engel S, Chu T, Forrer F, Müller C, de Jong M, van IJcken W, IJzermans JNM, Hoeijmakers JHJ, de Bruin RWF. Short-term dietary restriction and fasting precondition against ischemia reperfusion injury in mice. Aging Cell 2010; 9:40-53. [PMID: 19878145 DOI: 10.1111/j.1474-9726.2009.00532.x] [Citation(s) in RCA: 199] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Dietary restriction (DR) extends lifespan and increases resistance to multiple forms of stress, including ischemia reperfusion injury to the brain and heart in rodents. While maximal effects on lifespan require long-term restriction, the kinetics of onset of benefits against acute stress is not known. Here, we show that 2-4 weeks of 30% DR improved survival and kidney function following renal ischemia reperfusion injury in mice. Brief periods of water-only fasting were similarly effective at protecting against ischemic damage. Significant protection occurred within 1 day, persisted for several days beyond the fasting period and extended to another organ, the liver. Protection by both short-term DR and fasting correlated with improved insulin sensitivity, increased expression of markers of antioxidant defense and reduced expression of markers of inflammation and insulin/insulin-like growth factor-1 signaling. Unbiased transcriptional profiling of kidneys from mice subject to short-term DR or fasting revealed a significant enrichment of signature genes of long-term DR. These data demonstrate that brief periods of reduced food intake, including short-term daily restriction and fasting, can increase resistance to ischemia reperfusion injury in rodents and suggest a rapid onset of benefits of DR in mammals.
Collapse
Affiliation(s)
- James R Mitchell
- Department of Genetics, Erasmus Medical Center, Cancer Genomics Center, Dr. Molewaterplein 50, 3015 GE, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Abstract
The aging kidney undergoes several important anatomic and physiologic changes that increase the risk of acute kidney injury (formerly acute renal failure) in the elderly. This article reviews these changes and discusses the diagnoses frequently encountered in the elderly patient with acute kidney injury. The incidence, staging, evaluation, management, and prognosis of acute kidney injury are also examined with special focus given to older adults.
Collapse
|
225
|
Kfouri F, de Castro I, Testagrossa L, Delle H, da Silva AMG, Bastos APA, Vieira JM, Yu L. Role of p21 and oxidative stress on renal tubular resistance after acute ischaemic injury. Nephrol Dial Transplant 2010; 25:1795-803. [PMID: 20054021 DOI: 10.1093/ndt/gfp719] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Subsequent ischaemic episodes may induce renal resistance. P21 is a cell cycle inhibitor that may be induced by oxygen-free radicals and may have a protective effect in ischaemic acute kidney injury (AKI). This study aimed at evaluating the role of oxidative stress and p21 on tubular resistance in a model of acquired resistance after renal ischaemia and in isolated renal tubules. METHODS Wistar rats were divided into: Group 1--sham; Group 2--sham operated and after 2 days submitted to 45-min ischaemia; and Group 3--45-min ischaemia followed after 2 days by a second 45-min ischaemia. Plasma urea was evaluated on Days 0, 2 and 4. Serum creatinine, creatinine clearance and oxidants (thiobarbituric acid-reactive substances) were determined 48 h after the second procedure (Day 4). Histology, immunohistochemistry for lymphocytes (CD3), macrophages (ED1), proliferation (PCNA) and apoptosis (TUNEL) were also evaluated. Rat proximal tubules (PTs) were isolated by collagenase digestion and Percoll gradient from control rats and rats previously subjected to 35 min of ischaemia. PTs were submitted to 15-min hypoxia followed by 45-min reoxygenation. Cell injury was assessed by lactate dehydrogenase release and hydroperoxide production (xylenol orange). RESULTS Ischaemia induced AKI in Group 2 and 3 rats. Subsequent ischaemia did not aggravate renal injury, demonstrating renal resistance (Group 3). Renal function recovery was similar in Group 2 and 3. Plasma and urine oxidants were similar among in Group 2 and 3. Histology disclosed acute tubular necrosis in Group 2 and 3. Lymphocyte infiltrates were similar among all groups whereas macrophages infiltrate was greater in Group 3. Cell proliferation was greater in Group 2 compared with Group 3. Apoptosis was similar in groups 2 and 3. The p21 expression was increased only in Group 3 whereas it was similar in groups 1 and 2. PTs from the ischaemia group were sensitive to hypoxia but resistant to reoxygenation injury which was followed by lower hydroperoxide production compared to control PT. CONCLUSION Renal resistance induced by ischaemia was associated with cell mechanism mediators involving oxidative stress and increased p21 expression.
Collapse
Affiliation(s)
- Flavia Kfouri
- Division of Nephrology, University of São Paulo School of Medicine, Sao Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
226
|
Abstract
Acute kidney injury (AKI) is a common clinical syndrome in hospitalized patients associated with high morbidity and mortality rates. Despite several years of improvement in the medical care of the severely ill, there has been little improvement in outcome. Furthermore, effective means of preventing and treating AKI have remained elusive. Although dialysis has been the mainstay of treating AKI for > 40 years, several questions regarding its application remain unsettled, including method (continuous vs intermittent), timing, and dose. The purpose of this review is to summarize recent advances in the epidemiology and treatment of AKI in hospitalized patients.
Collapse
Affiliation(s)
- Kevin W Finkel
- Division of Renal Diseases and Hypertension, Section of Critical Care Nephrology, University of Texas Medical School at Houston, Houston, TX 77030, USA.
| | | |
Collapse
|
227
|
Silverstein DM. Inflammation after renal transplantation: Role in the development of graft dysfunction and potential therapies. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/17471060802302339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
228
|
Jung YJ, Kim DH, Lee AS, Lee S, Kang KP, Lee SY, Jang KY, Sung MJ, Park SK, Kim W. Peritubular capillary preservation with COMP-angiopoietin-1 decreases ischemia-reperfusion-induced acute kidney injury. Am J Physiol Renal Physiol 2009; 297:F952-60. [DOI: 10.1152/ajprenal.00064.2009] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ischemia followed by reperfusion induces microvascular endothelial cell injury, leading to the loss of functions such as regulation of vascular tone, tissue perfusion, permeability, and inflammation in kidney. Improvement of this endothelial dysfunction could be a good approach to treating ischemia-reperfusion-induced renal injury. Cartilage oligomeric matrix protein-angiopoietin-1 (COMP-Ang1) is a variant of native angiogenic factor angiopoietin-1 engineered to have higher activity. We evaluated the protective effect of COMP-Ang1 in an ischemia-reperfusion renal injury model. COMP-Ang1 preserved renal peritubular capillaries after ischemia-reperfusion injury without recruiting pericytes. Pretreatment with COMP-Ang1 attenuated the increase of blood urea nitrogen and serum creatinine levels after ischemia-reperfusion. In addition, the morphological examination indicated less tubular injury in mice pretreated with COMP-Ang1 than in those treated with the vehicle. COMP-Ang1 treatment reduced the increase in the number of Gr-1-positive neutrophils or ER-HR3-positive macrophages infiltrating kidneys, increased phosphorylation of Akt, and preserved renal tissue perfusion flow and microvascular permeability. Furthermore, COMP-Ang1 decreased renal interstitial fibrosis 30 days after the ischemia-reperfusion injury. In conclusion, COMP-Ang1 can be a possible endothelial cell-targeted therapy for preventing ischemia-reperfusion-induced acute kidney injury.
Collapse
Affiliation(s)
- Yu Jin Jung
- Renal Regeneration Laboratory and Department of Internal Medicine,
| | - Duk Hoon Kim
- Renal Regeneration Laboratory and Department of Internal Medicine,
| | - Ae Sin Lee
- Renal Regeneration Laboratory and Department of Internal Medicine,
| | - Sik Lee
- Renal Regeneration Laboratory and Department of Internal Medicine,
| | - Kyung Pyo Kang
- Renal Regeneration Laboratory and Department of Internal Medicine,
| | | | - Kyu Yun Jang
- Department of Pathology, Research Institute of Clinical Medicine, Chonbuk National University Medical School, Jeonju; and
| | - Mi Jeong Sung
- Food Function Research Center, Korea Food Research Institute, Songnam, South Korea
| | - Sung Kwang Park
- Renal Regeneration Laboratory and Department of Internal Medicine,
| | - Won Kim
- Renal Regeneration Laboratory and Department of Internal Medicine,
| |
Collapse
|
229
|
Fukuzawa N, Schenk AD, Petro M, Nonomura K, Baldwin WM, Fairchild RL. High renal ischemia temperature increases neutrophil chemoattractant production and tissue injury during reperfusion without an identifiable role for CD4 T cells in the injury. Transpl Immunol 2009; 22:62-71. [PMID: 19664707 DOI: 10.1016/j.trim.2009.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 07/17/2009] [Accepted: 07/28/2009] [Indexed: 11/18/2022]
Abstract
Various leukocyte populations, including neutrophils and CD4 T cells, have been implicated as mediators of acute renal ischemic injury. The influence of ischemic temperature on molecular and cellular mechanisms mediating this injury was tested in a mouse model. Wild-type C57BL/6, B6.CD4(-/-), B6.CD8(-/-), and B6.RAG-1(-/-) mice subjected to bilateral renal pedicle occlusion for 30 min at a higher (37 degrees C) but not a lower (32 degrees C) ischemic maintenance temperature had clear evidence of renal dysfunction and histopathology. Ischemia imposed at the higher temperature also increased CXCL1/KC and CXCL2/MIP-2 levels and neutrophils, but not T cells or macrophages, infiltrating into the ischemic kidneys. Depletion of neutrophils but not T cells attenuated the acute ischemic injury. These results indicate the influence of ischemic temperature and time on the production of neutrophil chemoattractants and subsequent neutrophil infiltration to mediate acute ischemic injury but fail to identify a role for adaptive immune components in this injury.
Collapse
Affiliation(s)
- Nobuyuki Fukuzawa
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | | | | | |
Collapse
|
230
|
Satpute SR, Park JM, Jang HR, Agreda P, Liu M, Gandolfo MT, Racusen L, Rabb H. The role for T cell repertoire/antigen-specific interactions in experimental kidney ischemia reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2009; 183:984-92. [PMID: 19561110 DOI: 10.4049/jimmunol.0801928] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
T cells have been implicated in the early pathogenesis of ischemia reperfusion injury (IRI) of kidney, liver, lung, and brain. It is not known whether Ag-TCR engagement followed by Ag-specific T cell activation participates in IRI. T cell-deficient nu/nu mice are moderately resistant to renal IRI, which can be reversed upon reconstitution with syngeneic T cells. In this study, we found that nu/nu mice reconstituted with DO11.10 T cells, limited in their TCR repertoire, have significantly less kidney dysfunction and tubular injury after renal IRI compared with that in nu/nu mice reconstituted with wild-type T cells having a diverse TCR repertoire. CD4(+) T cells infiltrating ischemic kidneys of nu/nu mice reconstituted with DO11.10 T cells exhibited lower IFN-gamma production than that of wild-type controls. Frequency of regulatory T cells in kidneys of these mice was similar in both DO11.10 T cells and wild-type T cell recipient groups. DO11.10 mice immunized with OVA-CFA had significantly worse kidney function at 24 h after ischemia than those immunized with CFA alone. Thus, without T cell activation, diverse TCR repertoire was important for renal IRI in naive mice. However, once T cells were activated in an Ag-specific manner through TCR in DO11.10 mice, a restricted TCR repertoire no longer limited the extent of kidney injury. Thus, both TCR repertoire-dependent and -independent factors mediate T cell functions in kidney IRI.
Collapse
|
231
|
Wang Y, John R, Chen J, Richardson JA, Shelton JM, Bennett M, Zhou XJ, Nagami GT, Zhang Y, Wu QQ, Lu CY. IRF-1 promotes inflammation early after ischemic acute kidney injury. J Am Soc Nephrol 2009; 20:1544-55. [PMID: 19443641 DOI: 10.1681/asn.2008080843] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Acute renal ischemia elicits an inflammatory response that may exacerbate acute kidney injury, but the regulation of the initial signals that recruit leukocytes is not well understood. Here, we found that IFN regulatory factor 1 (IRF-1) was a critical, early proinflammatory signal released during ischemic injury in vitro and in vivo. Within 15 min of reperfusion, proximal tubular cells of the S3 segment produced IRF-1, which is a transcription factor that activates proinflammatory genes. Transgenic knockout of IRF-1 ameliorated the impairment of renal function, morphologic injury, and inflammation after acute ischemia. Bone marrow chimera experiments determined that maximal ischemic injury required IRF-1 expression by both leukocytes and radioresistant renal cells, the latter identified as S3 proximal tubule cells in the outer medulla by in situ hybridization and immunohistochemistry. In vitro, reactive oxygen species, generated during ischemia/reperfusion injury, stimulated expression of IRF-1 in an S3 proximal tubular cell line. Taken together, these data suggest that IRF-1 gene activation by reactive oxygen species is an early signal that promotes inflammation after ischemic renal injury.
Collapse
Affiliation(s)
- Yanxia Wang
- Department of Internal Medicine-Nephrology, University of Texas Southwestern Medical School, 5323 Harry Hines Boulevard, Dallas, TX 75390-8856, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Naesens M, Li L, Ying L, Sansanwal P, Sigdel TK, Hsieh SC, Kambham N, Lerut E, Salvatierra O, Butte AJ, Sarwal MM. Expression of complement components differs between kidney allografts from living and deceased donors. J Am Soc Nephrol 2009; 20:1839-51. [PMID: 19443638 DOI: 10.1681/asn.2008111145] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A disparity remains between graft survival of renal allografts from deceased donors and from living donors. A better understanding of the molecular mechanisms that underlie this disparity may allow the development of targeted therapies to enhance graft survival. Here, we used microarrays to examine whole genome expression profiles using tissue from 53 human renal allograft protocol biopsies obtained both at implantation and after transplantation. The gene expression profiles of living-donor kidneys and pristine deceased-donor kidneys (normal histology, young age) were significantly different before reperfusion at implantation. Deceased-donor kidneys exhibited a significant increase in renal expression of complement genes; posttransplantation biopsies from well-functioning, nonrejecting kidneys, regardless of donor source, also demonstrated a significant increase in complement expression. Peritransplantation phenomena, such as donor death and possibly cold ischemia time, contributed to differences in complement pathway gene expression. In addition, complement gene expression at the time of implantation was associated with both early and late graft function. These data suggest that complement-modulating therapy may improve graft outcomes in renal transplantation.
Collapse
Affiliation(s)
- Maarten Naesens
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Figura M, Chilton L, Liacini A, Viskovic MM, Phan V, Knight D, Millar TM, Patel K, Kubes P, Giles WR, Tibbles LA. Blockade of K(ATP) channels reduces endothelial hyperpolarization and leukocyte recruitment upon reperfusion after hypoxia. Am J Transplant 2009; 9:687-96. [PMID: 19292831 DOI: 10.1111/j.1600-6143.2009.02553.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Ischemia/reperfusion injury in renal transplantation leads to slow or initial nonfunction, and predisposes to acute and chronic rejection. In fact, severe ischemia reperfusion injury can significantly reduce graft survival, even with modern immunosuppressive agents. One of the mechanisms by which ischemia/reperfusion causes injury is activation of endothelial cells resulting in inflammation. Although several therapies can be used to prevent leukocyte recruitment to ischemic vessels (e.g. antiadhesion molecule antibodies), there have been no clinical treatments reported that can prevent initial immediate neutrophil recruitment upon reperfusion. Using intravital microscopy, we describe abrogation of immediate neutrophil recruitment to ischemic microvessels by the K(ATP) antagonist glibenclamide (Glyburide). Further, we show that glibenclamide can reduce leukocyte recruitment in vitro under physiologic flow conditions. ATP-regulated potassium channels (K(ATP)) are important in the control of cell membrane polarization. Here we describe profound hyperpolarization of endothelial cells during hypoxia, and the reduction of this hyperpolarization using glibenclamide. These findings suggest that control of endothelial membrane potential during ischemia may be an important therapeutic tool in avoiding ischemia/reperfusion injury, and therefore, enhancing transplant long-term function.
Collapse
Affiliation(s)
- M Figura
- Institute of Infection, Immunity and Inflammation, Faculty of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Susa D, Mitchell JR, Verweij M, van de Ven M, Roest H, van den Engel S, Bajema I, Mangundap K, Ijzermans JNM, Hoeijmakers JHJ, de Bruin RWF. Congenital DNA repair deficiency results in protection against renal ischemia reperfusion injury in mice. Aging Cell 2009; 8:192-200. [PMID: 19338497 DOI: 10.1111/j.1474-9726.2009.00463.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cockayne syndrome and other segmental progerias with inborn defects in DNA repair mechanisms are thought to be due in part to hypersensitivity to endogenous oxidative DNA damage. The accelerated aging-like symptoms of this disorder include dysmyelination within the central nervous system, progressive sensineuronal hearing loss and retinal degeneration. We tested the effects of congenital nucleotide excision DNA repair deficiency on acute oxidative stress sensitivity in vivo. Surprisingly, we found mouse models of Cockayne syndrome less susceptible than wild type animals to surgically induced renal ischemia reperfusion injury, a multifactorial injury mediated in part by oxidative damage. Renal failure-related mortality was significantly reduced in Csb(-/-) mice, kidney function was improved and proliferation was significantly higher in the regenerative phase following ischemic injury. Protection from ischemic damage correlated with improved baseline glucose tolerance and insulin sensitivity and a reduced inflammatory response following injury. Protection was further associated with genetic ablation of a different Cockayne syndrome-associated gene, Csa. Our data provide the first functional in vivo evidence that congenital DNA repair deficiency can induce protection from acute stress in at least one organ. This suggests that while specific types of unrepaired endogenous DNA damage may lead to detrimental effects in certain tissues, they may at the same time elicit beneficial adaptive changes in others and thus contribute to the tissue specificity of disease symptoms.
Collapse
Affiliation(s)
- Denis Susa
- Department of Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Abstract
Acute kidney injury (AKI) is a very common condition encountered in a hospital setting. AKI is an independent risk factor for in-hospital mortality. In this review, we discuss in detail about the tubular, inflammatory and vascular molecular targets of AKI which may result in therapies to improve mortality and biomarkers for earlier diagnosis of AKI.
Collapse
|
236
|
He Z, Dursun B, Oh DJ, Lu L, Faubel S, Edelstein CL. Macrophages are not the source of injurious interleukin-18 in ischemic acute kidney injury in mice. Am J Physiol Renal Physiol 2009; 296:F535-42. [PMID: 19129255 PMCID: PMC2660194 DOI: 10.1152/ajprenal.90634.2008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2008] [Accepted: 01/05/2009] [Indexed: 01/09/2023] Open
Abstract
We previously reported in ischemic acute kidney injury (AKI) in mice that caspase-1-mediated production of interleukin-18 (IL-18) is pathogenic and that macrophage depletion by liposome-encapsulated clodronate (LEC) is protective. Therefore, our aim was to determine whether macrophages are a source of IL-18 in ischemic AKI in mice. On immunofluorescence staining of the outer stripe of outer medulla, the number of macrophages double stained for CD11b and IL-18 was significantly increased in AKI and significantly decreased by LEC. Adoptive transfer of RAW 264.7 cells, a mouse macrophage line that constitutively expresses IL-18 mRNA, reversed the functional protection against AKI in both LEC-treated wild-type and caspase-1 -/- mice. To test whether IL-18 in macrophages is necessary to cause AKI, we adoptively transferred macrophages in which IL-18 was inhibited. Peritoneal macrophages isolated from wild-type mice, IL-18 binding protein transgenic (IL-18 BP Tg) mice, and IL-18 -/- mice were used. IL-18 BP Tg mice overexpress human IL-18 BP and exhibit decreased biological activity of IL-18. Adoptive transfer of peritoneal macrophages from wild-type as well as IL-18 BP Tg and IL-18 -/- mice reversed the functional protection against AKI in LEC-treated mice. In summary, adoptive transfer of RAW cells, that constitutively express IL-18, reverses the functional protection in macrophage-depleted wild-type and caspase-1 -/- mice with AKI. However, adoptive transfer of peritoneal macrophages in which IL-18 function was inhibited also reverses the functional protection in macrophage-depleted mice. In conclusion, IL-18 from adoptive transfer of macrophages is not sufficient to cause ischemic AKI.
Collapse
Affiliation(s)
- Zhibin He
- Division of Renal Diseases, University of Colorado Denver and the Health Sciences Center, Aurora, Colorado 80262, USA
| | | | | | | | | | | |
Collapse
|
237
|
Abstract
Ischemia reperfusion injury (IRI) is a common and important clinical problem in many different organ systems, including kidney, brain, heart, liver, lung, and intestine. IRI occurs during all deceased donor organ transplants. IRI is a highly complex cascade of events that includes interactions between vascular endothelium, interstitial compartments, circulating cells, and numerous biochemical entities. It is well established that the innate immune system, such as complement, neutrophils, cytokines, chemokines, and macrophages participate in IRI. Recent data demonstrates an important role for lymphocytes, particularly T cells but also B cells in IRI. Lymphocytes not only participate in augmenting injury responses after IRI, but could also be playing a protective role depending on the cell type and stage of injury. Furthermore, lymphocytes appear to be participating in the healing response from IRI. These new data open the possibility for lymphocyte targeted therapeutics to improve the short and long term outcomes from IRI.
Collapse
Affiliation(s)
- Douglas Linfert
- Nephrology Division, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
238
|
Sharfuddin AA, Sandoval RM, Berg DT, McDougal GE, Campos SB, Phillips CL, Jones BE, Gupta A, Grinnell BW, Molitoris BA. Soluble thrombomodulin protects ischemic kidneys. J Am Soc Nephrol 2009; 20:524-34. [PMID: 19176699 DOI: 10.1681/asn.2008060593] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Altered coagulation and inflammation contribute to the pathogenesis of ischemic renal injury. Thrombomodulin is a necessary factor in the anticoagulant protein C pathway and has inherent anti-inflammatory properties. We studied the effect of soluble thrombomodulin (sTM) in a hypoperfusion model of ischemic kidney injury. To markedly reduce infrarenal aortic blood flow and femoral arterial pressures, we clamped the suprarenal aorta of rats, occluding them 90%, for 60 min. Reversible acute kidney injury (AKI) occurred at 24 h in rats subjected to hypoperfusion. Histologic analysis at 24 h revealed acute tubular necrosis (ATN), and intravital two-photon microscopy showed flow abnormalities in the microvasculature and defects of endothelial permeability. Pretreatment with rat sTM markedly reduced both I-R-induced renal dysfunction and tubular histologic injury scores. sTM also significantly improved microvascular erythrocyte flow rates, reduced microvascular endothelial leukocyte rolling and attachment, and minimized endothelial permeability to infused fluorescence dextrans, assessed by intravital quantitative multiphoton microscopy. Furthermore, sTM administered 2 h after reperfusion protected against ischemia-induced renal dysfunction at 24 h and improved survival. By using an sTM variant, we also determined that the protective effects of sTM were independent of its ability to generate activated protein C. These data suggest that sTM may have therapeutic potential for ischemic AKI.
Collapse
Affiliation(s)
- Asif A Sharfuddin
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Furuichi K, Gao JL, Horuk R, Wada T, Kaneko S, Murphy PM. Chemokine receptor CCR1 regulates inflammatory cell infiltration after renal ischemia-reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2009; 181:8670-6. [PMID: 19050287 DOI: 10.4049/jimmunol.181.12.8670] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neutrophils and macrophages rapidly infiltrate the kidney after renal ischemia-reperfusion injury, however specific molecular recruitment mechanisms have not been fully delineated for these cell types. Here we provide genetic and pharmacologic evidence supporting a positive role for the chemokine receptor CCR1 in macrophage and neutrophil infiltration in a 7 day mouse model of renal ischemia-reperfusion injury. By day 7, injured kidneys from mice lacking CCR1 contained 35% fewer neutrophils and 45% fewer macrophages than injured kidneys from wild-type control mice. Pretreatment of wild-type mice with the specific CCR1 antagonist BX471 also suppressed neutrophil and macrophage infiltration in the model. Injured kidneys from mice lacking CCR1 also had reduced content of the CCR1 ligands CCL3 (MIP-1alpha) and CCL5 (RANTES) compared with injured kidneys from wild-type controls, suggesting a leukocyte source for these inflammatory chemokines and existence of a CCR1-dependent positive feedback loop for leukocyte infiltration in the model. Local leukocyte proliferation and apoptosis were detected after injury, but were not dependent on CCR1. Also, the extent of necrotic and fibrotic damage and decline in renal function in injured kidneys was similar in wild-type and CCR1-deficient mice. Thus, CCR1 appears to regulate trafficking of macrophages and neutrophils to kidney in a mouse model of renal ischemia-reperfusion injury, however this activity does not appear to affect tissue injury.
Collapse
Affiliation(s)
- Kengo Furuichi
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
240
|
Compartmentalization of neutrophils in the kidney and lung following acute ischemic kidney injury. Kidney Int 2009; 75:689-98. [PMID: 19129795 DOI: 10.1038/ki.2008.648] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
During renal ischemia-reperfusion, local and distant tissue injury is caused by an influx of neutrophils into the affected tissues. Here we measured the kinetics of margination and transmigration of neutrophils in vivo in the kidney and lungs following renal ischemia-reperfusion. After bilateral renal injury, kidney neutrophil content increased threefold at 24 h. The neutrophils were found primarily in the interstitium and to a lesser degree marginated to the vascular endothelium. These interstitial neutrophils had significantly lower levels of intracellular IFN-gamma, IL-4, IL-6, and IL-10 a tendency for decreased amounts of IL-4 and TNF-alpha compared to the marginated neutrophils. Localization of the neutrophils to the kidney interstitium was confirmed by high resolution microscopy and these sites of transmigration were directly associated with areas of increased vascular permeability. Activation of the adenosine 2A receptor significantly decreased both kidney neutrophil transmigration by about half and vascular permeability by about a third. After unilateral renal ischemia-reperfusion, the unclipped kidney and lungs did not accumulate interstitial neutrophils or have increased vascular permeability despite a marked increase of neutrophil margination in the lungs. Our findings suggest there is a sequential recruitment and transmigration of neutrophils from the vasculature into the kidney interstitium at the site of tissue injury following renal ischemia-reperfusion.
Collapse
|
241
|
|
242
|
Grenier N, Hauger O, Eker O, Combe C, Couillaud F, Moonen C. Molecular magnetic resonance imaging of the genitourinary tract: recent results and future directions. Magn Reson Imaging Clin N Am 2008; 16:627-41, viii. [PMID: 18926427 DOI: 10.1016/j.mric.2008.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This article focuses on preclinical and early clinical applications of renal cell MR imaging, on new developments in MR control of intrarenal gene therapy, and on several potential applications of molecular imaging techniques, mainly targeting cell receptors and enzyme activity, which could find exciting applications within the genitourinary tract.
Collapse
Affiliation(s)
- Nicolas Grenier
- UMR-CNRS 5231 Imagerie Moléculaire et Fonctionnelle, Université Victor Segalen-Bordeaux 2, Bordeaux-Cedex, France.
| | | | | | | | | | | |
Collapse
|
243
|
Zhang ZX, Wang S, Huang X, Min WP, Sun H, Liu W, Garcia B, Jevnikar AM. NK cells induce apoptosis in tubular epithelial cells and contribute to renal ischemia-reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2008; 181:7489-98. [PMID: 19017938 DOI: 10.4049/jimmunol.181.11.7489] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Renal ischemia-reperfusion injury (IRI) can result in acute renal failure with mortality rates of 50% in severe cases. NK cells are important participants in early-stage innate immune responses. However, their role in renal tubular epithelial cell (TEC) injury in IRI is currently unknown. Our data indicate that NK cells can kill syngeneic TEC in vitro. Apoptotic death of TEC in vitro is associated with TEC expression of the NK cell ligand Rae-1, as well as NKG2D on NK cells. In vivo following IRI, there was increased expression of Rae-1 on TEC. FACS analyses of kidney cell preparations indicated a quantitative increase in NKG2D-bearing NK cells within the kidney following IRI. NK cell depletion in wild-type C57BL/6 mice was protective, while adoptive transfer of NK cells worsened injury in NK, T, and B cell-null Rag2(-/-)gamma(c)(-/-) mice with IRI. NK cell-mediated kidney injury was perforin (PFN)-dependent as PFN(-/-) NK cells had minimal capacity to kill TEC in vitro compared with NK cells from wild-type, FasL-deficient (gld), or IFN-gamma(-/-) mice. Taken together, these results demonstrate for the first time that NK cells can directly kill TEC and that NK cells contribute substantially to kidney IRI. NK cell killing may represent an important underrecognized mechanism of kidney injury in diverse forms of inflammation, including transplantation.
Collapse
Affiliation(s)
- Zhu-Xu Zhang
- The Multi-Organ Transplant Program, London Health Sciences Centre, London, Ontario, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
244
|
Gupta A, Gerlitz B, Richardson MA, Bull C, Berg DT, Syed S, Galbreath EJ, Swanson BA, Jones BE, Grinnell BW. Distinct functions of activated protein C differentially attenuate acute kidney injury. J Am Soc Nephrol 2008; 20:267-77. [PMID: 19092124 DOI: 10.1681/asn.2008030294] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Administration of activated protein C (APC) protects from renal dysfunction, but the underlying mechanism is unknown. APC exerts both antithrombotic and cytoprotective properties, the latter via modulation of protease-activated receptor-1 (PAR-1) signaling. We generated APC variants to study the relative importance of the two functions of APC in a model of LPS-induced renal microvascular dysfunction. Compared with wild-type APC, the K193E variant exhibited impaired anticoagulant activity but retained the ability to mediate PAR-1-dependent signaling. In contrast, the L8W variant retained anticoagulant activity but lost its ability to modulate PAR-1. By administering wild-type APC or these mutants in a rat model of LPS-induced injury, we found that the PAR-1 agonism, but not the anticoagulant function of APC, reversed LPS-induced systemic hypotension. In contrast, both functions of APC played a role in reversing LPS-induced decreases in renal blood flow and volume, although the effects on PAR-1-dependent signaling were more potent. Regarding potential mechanisms for these findings, APC-mediated PAR-1 agonism suppressed LPS-induced increases in the vasoactive peptide adrenomedullin and infiltration of iNOS-positive leukocytes into renal tissue. However, the anticoagulant function of APC was responsible for suppressing LPS-induced stimulation of the proinflammatory mediators ACE-1, IL-6, and IL-18, perhaps accounting for its ability to modulate renal hemodynamics. Both variants reduced active caspase-3 and abrogated LPS-induced renal dysfunction and pathology. We conclude that although PAR-1 agonism is solely responsible for APC-mediated improvement in systemic hemodynamics, both functions of APC play distinct roles in attenuating the response to injury in the kidney.
Collapse
Affiliation(s)
- Akanksha Gupta
- Biotechnology Discovery Research, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285-0444, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Jang HR, Rabb H. The innate immune response in ischemic acute kidney injury. Clin Immunol 2008; 130:41-50. [PMID: 18922742 DOI: 10.1016/j.clim.2008.08.016] [Citation(s) in RCA: 253] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2008] [Accepted: 08/10/2008] [Indexed: 11/30/2022]
Abstract
Kidney ischemia reperfusion injury is a major cause of morbidity in both allograft and native kidneys. Ischemia reperfusion-induced acute kidney injury is characterized by early, alloantigen-independent inflammation. Major components of the innate immune system are activated and participate in the pathogenesis of acute kidney injury, plus prime the allograft kidney for rejection. Soluble members of innate immunity implicated in acute kidney injury include the complement system, cytokines, and chemokines. Toll-like receptors (TLRs) are also important contributors. Effector cells that participate in acute kidney injury include the classic innate immune cells, neutrophils and macrophages. Recent data has unexpectedly identified lymphocytes as participants of early acute kidney injury responses. In this review, we will focus on immune mediators that participate in the pathogenesis of ischemic acute kidney injury.
Collapse
Affiliation(s)
- Hye Ryoun Jang
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, Ross Building, Room 965, 720 Rutland Avenue, Baltimore, MD 21205, USA
| | | |
Collapse
|
246
|
Fernandes Bertocchi AP, Campanhole G, Wang PHM, Gonçalves GM, Damião MJ, Cenedeze MA, Beraldo FC, de Paula Antunes Teixeira V, dos Reis MA, Mazzali M, Pacheco-Silva A, Câmara NOS. A Role for galectin-3 in renal tissue damage triggered by ischemia and reperfusion injury. Transpl Int 2008; 21:999-1007. [DOI: 10.1111/j.1432-2277.2008.00705.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
247
|
Turgut F, Bayrak O, Catal F, Bayrak R, Atmaca AF, Koc A, Akbas A, Akcay A, Unal D. Antioxidant and protective effects of silymarin on ischemia and reperfusion injury in the kidney tissues of rats. Int Urol Nephrol 2008; 40:453-60. [PMID: 18368506 DOI: 10.1007/s11255-008-9365-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2007] [Accepted: 03/04/2008] [Indexed: 11/28/2022]
Abstract
BACKGROUND Renal ischemia/reperfusion (I/R) injury is a major cause of acute renal failure. Silymarin is extracted from Silybum marianum and Cynara cardunculus seeds and fruits. The aim of this study is to investigate whether silymarin administration prevents the damage induced by I/R in rat kidneys. MATERIALS AND METHODS Thirty male Wistar rats were randomly divided into five experimental groups (n = 6, each) as follows; control group, sham-operated group, I/R group, silymarin group, and I/R + silymarin group. In the I/R and I/R + silymarin groups, both renal arteries were occluded using nontraumatic microvascular clamps for 45 min. Then, at the end of 24 h of reperfusion, the animals were killed. Kidney function tests, the serum and tissue antioxidant enzymes and oxidant products were determined. RESULTS Animals that were subjected to I/R exhibited significant increase in serum urea, creatinine, and cystatin C levels compared with the rats treated with silymarin prior to the I/R process (P < 0.001). The serum enzymatic activities of superoxide dismutase and glutathione peroxidase significantly decreased in the I/R group; however, this reduction was significantly improved by the treatment with silymarin (P < 0.001 and P < 0.05, respectively). Renal I/R produced a significant increase in serum and tissue malondialdehyde, nitric oxide, and protein carbonyl as compared with controls. Treatment with silymarin resulted in significant reduction in these markers (P < 0.001). CONCLUSION Based on our findings, silymarin protects the kidneys against I/R injury. This finding may provide a basis for the development of novel therapeutic strategies for protection against the damages caused by I/R.
Collapse
Affiliation(s)
- Faruk Turgut
- Department of Nephrology, Fatih University, School of Medicine, Hosdere cad no: 145, Y. Ayranci, 06540 Ankara, Turkey.
| | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Legrand M, Mik EG, Johannes T, Payen D, Ince C. Renal hypoxia and dysoxia after reperfusion of the ischemic kidney. MOLECULAR MEDICINE (CAMBRIDGE, MASS.) 2008. [PMID: 18488066 DOI: 10.2119/2008-00006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ischemia is the most common cause of acute renal failure. Ischemic-induced renal tissue hypoxia is thought to be a major component in the development of acute renal failure in promoting the initial tubular damage. Renal oxygenation originates from a balance between oxygen supply and consumption. Recent investigations have provided new insights into alterations in oxygenation pathways in the ischemic kidney. These findings have identified a central role of microvascular dysfunction related to an imbalance between vasoconstrictors and vasodilators, endothelial damage and endothelium-leukocyte interactions, leading to decreased renal oxygen supply. Reduced microcirculatory oxygen supply may be associated with altered cellular oxygen consumption (dysoxia), because of mitochondrial dysfunction and activity of alternative oxygen-consuming pathways. Alterations in oxygen utilization and/or supply might therefore contribute to the occurrence of organ dysfunction. This view places oxygen pathways' alterations as a potential central player in the pathogenesis of acute kidney injury. Both in regulation of oxygen supply and consumption, nitric oxide seems to play a pivotal role. Furthermore, recent studies suggest that, following acute ischemic renal injury, persistent tissue hypoxia contributes to the development of chronic renal dysfunction. Adaptative mechanisms to renal hypoxia may be ineffective in more severe cases and lead to the development of chronic renal failure following ischemia-reperfusion. This paper is aimed at reviewing the current insights into oxygen transport pathways, from oxygen supply to oxygen consumption in the kidney and from the adaptation mechanisms to renal hypoxia. Their role in the development of ischemia-induced renal damage and ischemic acute renal failure are discussed.
Collapse
Affiliation(s)
- Matthieu Legrand
- Department of Physiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
249
|
Legrand M, Mik EG, Johannes T, Payen D, Ince C. Renal hypoxia and dysoxia after reperfusion of the ischemic kidney. Mol Med 2008; 14:502-16. [PMID: 18488066 DOI: 10.2119/2008-00006.legrand] [Citation(s) in RCA: 195] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 04/17/2008] [Indexed: 12/18/2022] Open
Abstract
Ischemia is the most common cause of acute renal failure. Ischemic-induced renal tissue hypoxia is thought to be a major component in the development of acute renal failure in promoting the initial tubular damage. Renal oxygenation originates from a balance between oxygen supply and consumption. Recent investigations have provided new insights into alterations in oxygenation pathways in the ischemic kidney. These findings have identified a central role of microvascular dysfunction related to an imbalance between vasoconstrictors and vasodilators, endothelial damage and endothelium-leukocyte interactions, leading to decreased renal oxygen supply. Reduced microcirculatory oxygen supply may be associated with altered cellular oxygen consumption (dysoxia), because of mitochondrial dysfunction and activity of alternative oxygen-consuming pathways. Alterations in oxygen utilization and/or supply might therefore contribute to the occurrence of organ dysfunction. This view places oxygen pathways' alterations as a potential central player in the pathogenesis of acute kidney injury. Both in regulation of oxygen supply and consumption, nitric oxide seems to play a pivotal role. Furthermore, recent studies suggest that, following acute ischemic renal injury, persistent tissue hypoxia contributes to the development of chronic renal dysfunction. Adaptative mechanisms to renal hypoxia may be ineffective in more severe cases and lead to the development of chronic renal failure following ischemia-reperfusion. This paper is aimed at reviewing the current insights into oxygen transport pathways, from oxygen supply to oxygen consumption in the kidney and from the adaptation mechanisms to renal hypoxia. Their role in the development of ischemia-induced renal damage and ischemic acute renal failure are discussed.
Collapse
Affiliation(s)
- Matthieu Legrand
- Department of Physiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
250
|
Sutton TA. Alteration of microvascular permeability in acute kidney injury. Microvasc Res 2008; 77:4-7. [PMID: 18938184 DOI: 10.1016/j.mvr.2008.09.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Accepted: 09/03/2008] [Indexed: 10/21/2022]
Abstract
Functional and structural abnormalities in the renal microvasculature are important processes contributing to the pathophysiology of ischemic acute kidney injury (AKI). Renewed interest in the complex interplay between tubular injury, inflammation and microvascular alterations has emerged in order to gain a better understanding of acute kidney injury syndromes. This review examines alterations of the renal microvasculature as they relate to ischemic and septic AKI with an emphasis on the mechanisms involved in altered microvascular permeability.
Collapse
Affiliation(s)
- Timothy A Sutton
- Division of Nephrology, Department of Medicine, Indiana Center for Biological Microscopy, Indiana University School of Medicine, 950 West Walnut Street R-II, 202 Indianapolis, IN 46202, USA.
| |
Collapse
|