1
|
Prionas A, Hamaoui K, Vanezis K, Reebye V, Habib N, Papalois V. The Effect of Interleukin-10 Immunotherapy on Renal Ischemia-Reperfusion Injury: A Systematic Review and Meta-Analysis of Preclinical Studies. Int J Mol Sci 2024; 25:6231. [PMID: 38892418 PMCID: PMC11172821 DOI: 10.3390/ijms25116231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Renal ischemia-reperfusion is a common cause of acute kidney injury leading to significant morbidity and mortality. There are no effective treatments available in clinical practice. This meta-analysis aims to assess the effect of IL-10 immunotherapy on renal ischemia-reperfusion injury. Medline, Embase, Cochrane-library, Google Scholar and clinicaltrials.gov were searched up to 31 March 2023. Preclinical and clinical interventional studies investigating IL-10 immunotherapy for renal ischemia-reperfusion were eligible for inclusion. The primary endpoint was renal function (serum creatinine) following ischemia-reperfusion. The secondary endpoints included mitochondrial integrity, cellular proliferation, regulated cell death (TUNEL assay), expression of inflammatory cytokines (TNF-α, IL-6 and IL-1β), M1/M2 macrophage polarization, tissue integrity (tubular injury score), long-term kidney fibrosis (fibrotic area %) and adverse events (pulmonary toxicity, cardiotoxicity hepatotoxicity). The search returned 861 records. From these, 16 full texts were screened and subsequently, seven animal studies, corresponding to a population of 268 mice/rats, were included. Compared to the control treatment, IL-10 immunotherapy reduced serum creatinine more effectively within 24 h of administration (95% CI: -9.177, -5.601, I2 = 22.42%). IL-10 immunotherapy promoted mitochondrial integrity and cellular proliferation and reduced regulated cell death (95% CI: -11.000, -4.184, I2 = 74.94%). It decreased the expression of TNF-α, IL-6 and IL-1β, led to M2 polarization of the local macrophages, reduced tubular injury score (95% CI: -8.917, -5.755, I2 = 22.71%), and long-term kidney fibrosis (95% CI: -6.963, -3.438, I2 = 0%). No adverse outcomes were captured. In Conclusion, IL-10 immunotherapy safely improves outcomes in animal models of renal ischemia-reperfusion; the translational potential of IL-10 immunotherapy needs to be further investigated in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Vassilios Papalois
- Department of Surgery and Cancer, Imperial College London, London W12 0HS, UK; (A.P.); (K.H.); (K.V.); (V.R.); (N.H.)
| |
Collapse
|
2
|
Watts BA, Tamayo E, Sherwood ER, Good DW. Monophosphoryl lipid A pretreatment suppresses sepsis- and LPS-induced proinflammatory cytokine production in the medullary thick ascending limb. Am J Physiol Renal Physiol 2020; 319:F8-F18. [PMID: 32421349 DOI: 10.1152/ajprenal.00178.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Sepsis is the leading cause of acute kidney injury in critically ill patients. Tumor necrosis factor-α (TNF-α) has been implicated in the pathogenesis of septic kidney injury; however, the sites and mechanisms of renal TNF-α production during sepsis remain to be defined. In the present study, we showed that TNF-α expression is increased in medullary thick ascending limbs (MTALs) of mice with sepsis induced by cecal ligation and puncture. Treatment with lipopolysaccharide (LPS) for 3 h in vitro also increased MTAL TNF-α production. Sepsis and LPS increased MTAL TNF-α expression through activation of the myeloid differentiation factor 88 (MyD88)-IL-1 receptor-associated kinase 1-ERK signaling pathway. Pretreatment with monophosphoryl lipid A (MPLA), a nontoxic immunomodulator that protects against bacterial infection, eliminated the sepsis- and LPS-induced increases in MTAL TNF-α production. The suppressive effect of MPLA on TNF-α was mediated through activation of a phosphatidylinositol 3-kinase-dependent pathway that inhibits MyD88-dependent ERK activation. This likely involves MPLA-phosphatidylinositol 3-kinase-mediated induction of Tollip, which negatively regulates the MyD88-ERK pathway by inhibiting activation of IL-1 receptor-associated kinase 1. These regulatory mechanisms are similar to those previously shown to mediate the effect of MPLA to prevent sepsis-induced inhibition of MTAL [Formula: see text] absorption. These results identify the MTAL as a site of local TNF-α production in the kidney during sepsis and identify molecular mechanisms that can be targeted to attenuate renal TNF-α expression. The ability of MPLA pretreatment to suppress MyD88-dependent ERK signaling in the MTAL during sepsis has the dual beneficial effects of protecting tubule transport functions and attenuating harmful proinflammatory responses.
Collapse
Affiliation(s)
- Bruns A Watts
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Esther Tamayo
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Edward R Sherwood
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David W Good
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.,Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
3
|
Huart J, Krzesinski JM, Jouret F. Genetic susceptibility to delayed graft function following kidney transplantation: a systematic review of the literature. Clin Kidney J 2018; 11:586-596. [PMID: 30090630 PMCID: PMC6070034 DOI: 10.1093/ckj/sfy020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/12/2018] [Indexed: 12/22/2022] Open
Abstract
Delayed graft function (DGF) is defined as the need for dialysis within 7 days following kidney transplantation (KTx). DGF is associated with increased costs, higher risk for acute rejection and decreased long-term graft function. Renal ischaemia/reperfusion (I/R) injury plays a major role in DGF occurrence. Single nucleotide polymorphisms (SNPs) in certain genes may aggravate kidney susceptibility to I/R injury, thereby worsening post-transplant outcomes. The present article proposes an extensive review of the literature about the putative impact of donor or recipient SNPs on DGF occurrence in kidney transplant recipients (KTRs). Among 30 relevant PubMed reports, 16 articles identified an association between 18 SNPs and DGF. These polymorphisms concern 14 different well-known genes and one not-yet-identified gene located on chromosome 18. They have been categorized into five groups according to the role of the corresponding proteins in I/R cascade: (i) oxidative stress, (ii) telomere shortening, (iii) chemokines, (iv) T-cell homeostasis and (v) metabolism of anti-inflammatory molecules. The remaining 14 studies failed to demonstrate any association between the studied SNPs and the occurrence of DGF. A better understanding of the genetic susceptibility to renal I/R injury may help prevent DGF and improve clinical outcomes in KTRs.
Collapse
Affiliation(s)
- Justine Huart
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital (ULg CHU), Liège, Belgium.,Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Jean-Marie Krzesinski
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital (ULg CHU), Liège, Belgium.,Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - François Jouret
- Division of Nephrology, Department of Internal Medicine, University of Liège Hospital (ULg CHU), Liège, Belgium.,Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège, Liège, Belgium
| |
Collapse
|
4
|
Senturk Ciftci H, Demir E, Savran Karadeniz M, Tefik T, Yazici H, Nane I, Savran Oguz F, Aydin F, Turkmen A. Serum and Urinary Levels of Tumor Necrosis Factor-Alpha in Renal Transplant Patients. EXP CLIN TRANSPLANT 2017; 16:671-675. [PMID: 29251577 DOI: 10.6002/ect.2017.0166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Allograft rejection is an important cause of early and long-term graft loss in kidney transplant recipients. Tumor necrosis factor-alpha promotes T-cell activation, the key reaction leading to allograft rejection. Here, we investigated whether serum and urinary tumor necrosis factor-alpha levels can predict allograft rejection. MATERIALS AND METHODS This study included 65 living related-donor renal transplant recipients with mean follow-up of 26 ± 9 months. Serum and urinary tumor necrosis factor-alpha levels were measured at pretransplant and at posttransplant time points (days 1 and 7 and months 3 and 6); serum creatinine levels were also monitored during posttransplant follow-up. Standard enzyme-linked immunoabsorbent assay was used to detect tumor necrosis factor-alpha levels. Clinical variables were monitored. RESULTS Nine of 65 patients (13.8%) had biopsy-proven rejection during follow-up. Preoperative serum and urinary tumor necrosis factor-alpha levels were not significantly different when we compared patients with and without rejection. Serum tumor necrosis factor-alpha levels (in pg/mL) were significantly higher in the allograft rejection versus nonrejection group at day 7 (11.5 ± 4.7 vs 15.4 ± 5.8; P = .029) and month 1 (11.1 ± 4.8 vs 17.8 ± 10.9; P =.003). Urinary tumor necrosis factor-alpha levels (in pg/mL) were also elevated in the allograft rejection versus the nonrejection group at days 1 (10.2 ± 2.5 vs 14.1 ± 6.8; P = .002) and 7 (9.8 ± 2.2 vs 14.5 ± 2.7; P < .001) and at months 1 (8.0 ± 1.7 vs 11.8 ± 2.4; P < .001), 3 (7.7 ± 1.6 vs 9.6 ± 1.7; P = .002), and 6 (7.4 ± 1.6 vs 8.9 ± 0.9; P = .005). CONCLUSIONS Our preliminary findings suggest that tumor necrosis factor-alpha has a role in diagnosing renal transplant rejection. Serum and urinary tumor necrosis factor-alpha levels may be a possible predictor for allograft rejection.
Collapse
Affiliation(s)
- Hayriye Senturk Ciftci
- From the Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Zuo H, Li Y, Cui Y, An Y. Cardioprotective effect of Malva sylvestris L. in myocardial ischemic/reprefused rats. Biomed Pharmacother 2017; 95:679-684. [PMID: 28886527 DOI: 10.1016/j.biopha.2017.08.111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 08/24/2017] [Accepted: 08/24/2017] [Indexed: 02/04/2023] Open
Abstract
PURPOSE The present investigation evaluated the cardioprotective effect of Malva sylvestris L. (MS) on myocardial ischemic/reperfusion (MI/R) in rats. METHODS All animals were divided into four groups: the sham operated group, ischemia/reperfusion group (MI/R), and the MS (250 and 500mg/kg) treated groups, who received MS 250 and 500mg/kg intragastrically for 15 consecutive days, respectively. At the end of the protocol, concentrations of aspartate transaminase (AST), creatine kinase-MB fraction (CK-MB) and lactate dehydrogenase (LDH) were estimated in serum and the concentrations of other parameters, such as C-reactive protein, macrophage inflammatory protein 1 alpha (MIP-1α), and nitric oxide (NO) were also estimated in the blood. Tissue homogenate concentrations of inflammatory cytokines, such as tumour necrosis factor-α (TNF-α), interlukin-1β (IL-1β), IL-10 and IL-6 as well as oxidative stress parameters, such as lipid peroxidation, catalase, and superoxide dismutase were estimated in MI/R rats. RESULT Significant decreases (p<0.01) in AST, LDH, and CK-MB levels were observed in the MS-treated group compared with those in the MI/R group. C-reactive protein and MIP-1α levels decreased in the MS-treated group compared with those in the MI/R group. Plasma NO level was significantly enhanced in the MS-treated group than in the MI/R group. Moreover, treatment with MS significantly reduced TNF-α, IL-1β, and IL-6 levels and increased IL-10 levels in the MS group compared with the MI/R group. Treatment with MS also attenuated the altered oxidative stress parameters in MI/R rats. CONCLUSION The present results indicate the cardioprotective effects of MS of reducing oxidative stress and the inflammatory response in MI/R rats.
Collapse
Affiliation(s)
- Hanheng Zuo
- Cardiovascular Division, The Medical College of Qingdao University, Qingdao, Shandong 266021, China
| | - Yinping Li
- Cardiac Care Unit, The Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, China
| | - Yinghua Cui
- Cardiac Care Unit, The Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, China
| | - Yi An
- Cardiovascular Division, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 222071, China.
| |
Collapse
|
6
|
Zhang Y, Rong S, Feng Y, Zhao L, Hong J, Wang R, Yuan W. Simvastatin attenuates renal ischemia/reperfusion injury from oxidative stress via targeting Nrf2/HO-1 pathway. Exp Ther Med 2017; 14:4460-4466. [PMID: 29067120 DOI: 10.3892/etm.2017.5023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 06/15/2017] [Indexed: 12/26/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury of the kidneys is commonly encountered in the clinic. The present study assessed the efficacy of simvastatin in preventing I/R-induced renal injury in a rat model and investigated the corresponding molecular mechanisms. Rats were divided into 3 groups, including a sham, I/R and I/R + simvastatin group. The results revealed that in the I/R group, the levels of blood urea nitrogen, serum creatinine and lactate dehydrogenase were significantly higher than those in the sham group, which was significantly inhibited by simvastatin pre-treatment. I/R significantly decreased superoxide dismutase activity compared with that in the sham group, which was largely rescued by simvastatin. Furthermore, I/R significantly increased the malondialdehyde content compared with that in the sham group, which was reduced by simvastatin. Hematoxylin-eosin staining revealed no obvious morphological abnormalities in the sham group, while I/R led to notable tubular cell swelling, vacuolization, cast formation and tubular necrosis, which was rescued by simvastatin. A terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling assay demonstrated that I/R significantly increased the number of apoptotic cells compared with that in the sham group, which was significantly inhibited by simvastatin. Western blot analysis demonstrated that simvastatin upregulated I/R-induced increases of nuclear factor erythroid-2-related factor 2 (Nrf2) and anti-oxidant enzyme heme oxygenase-1 (HO-1). Reverse-transcription quantitative PCR indicated that changes in the mRNA levels of Nrf2 and HO-1 were consistent with the western blot results. It was concluded that simvastatin treatment led to upregulation of HO-1 protein levels through activating the Nrf2 signaling pathway to ultimately protect the kidneys from I/R-associated oxidative damage.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Emergency Intensive Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 200082, P.R. China
| | - Shu Rong
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai 200082, P.R. China
| | - Yi Feng
- Department of Emergency Intensive Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 200082, P.R. China
| | - Liqun Zhao
- Department of Emergency Intensive Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 200082, P.R. China
| | - Jiang Hong
- Department of Emergency Intensive Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 200082, P.R. China
| | - Ruilan Wang
- Department of Emergency Intensive Medicine, Shanghai General Hospital of Nanjing Medical University, Shanghai 200082, P.R. China
| | - Weijie Yuan
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai 200082, P.R. China
| |
Collapse
|
7
|
Li QP, Wei RB, Yang X, Zheng XY, Su TY, Huang MJ, Yin Z, Chen XM. Protective Effects and Mechanisms of Shenhua Tablet () on Toll-Like Receptors in Rat Model of Renal Ischemia-Reperfusion Injury. Chin J Integr Med 2017; 25:37-44. [PMID: 28466227 DOI: 10.1007/s11655-017-2756-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVES To investigate the protective effects and potential mechanisms of Shenhua Tablet (, SHT) on the toll-like receptors (TLRs)-mediated signaling pathways in a rat model of kidney ischemia-reperfusion injury (IRI). METHODS Sixty male Wistar rats were randomly divided into 5 groups: sham surgery, model control, astragaloside (150 mg•kg-1•d-1), low- and high-dose SHT (1.5 and 3.0 g•kg-1•d-1, repectively) groups. One week after drug treatment, rats underwent surgery to establish the IRI models. At 24 h and 72 h after the modeling, serum creatinine (Scr) and blood urea nitrogen (BUN) were analyzed; pathological damage were scored after periodic acid-Schiffstaining. TLR2, TLR4 and myeloid differentiation factor 88 (MyD88) protein and mRNA expressions were detected by inmmunohistochemistry, Western blot and qPCR. Tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) protein expressions were detected by enzyme linked immunosorbent assay. RESULTS Compared with the sham group, the model group exhibited severe change in renal function (Scr: 189.42±21.50, P<0.05), pathological damage (damage score: 4.50±0.55, P<0.05), and the expression levels of TLR2, TLR4, MyD88, TNF-α, IL-6 were significantly higher than other groups. Meanwhile, the levels of TLRs in model group showed upward tendency from 24 to 72 h, unparalleled with pathological and functional changes. The aforementioned parameters were alleviated to a certain extent, and, in addition to TLRs, presented the obvious downward trending from the 24 to 72 h after the intervention in the SHT and astragaloside groups relative to the model (P<0.05); in particular, the most significant mitigation of these changes was observed in the SHT-H group (P<0.05). CONCLUSION TLRs may be an important spot to treat and research in acute kidney injury. SHT could effectively mitigate renal injuries and promote recovery of IRI injuries through suppression of degeneration induced by up-regulation of TLR2 and TLR4 expression levels in the MyD88-dependent signaling pathway and exhibit some dose dependence.
Collapse
Affiliation(s)
- Qing-Ping Li
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Ri-Bao Wei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China.
| | - Xi Yang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Xiao-Yong Zheng
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Ting-Yu Su
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Meng-Jie Huang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Zhong Yin
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| | - Xiang-Mei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, 100853, China
| |
Collapse
|
8
|
Hou L, Chen G, Feng B, Zhang XS, Zheng XF, Xiang Y, Zhao GY, Min WP. Small interfering RNA targeting TNF-α gene significantly attenuates renal ischemia-reperfusion injury in mice. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2016; 36:634-638. [PMID: 27752902 DOI: 10.1007/s11596-016-1638-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 08/22/2016] [Indexed: 01/05/2023]
Abstract
Tumor necrosis factor-alpha (TNF-α) has been found to be centrally involved in the development of ischemia-reperfusion injury (IRI)-induced inflammation and apoptosis. Knockdown of TNF-α gene using small interfering RNA (siRNA) may protect renal IRI. Renal IRI was induced in mice by clamping the left renal pedicle for 25 or 35 min. TNF-α siRNA was administered intravenously to silence the expression of TNF-α. The therapeutic effects of siRNA were evaluated in terms of renal function, histological examination, and overall survival following lethal IRI. A single systemic injection of TNF-α siRNA resulted in significant knockdown of TNF-α expression in ischemia-reperfusion injured kidney. In comparison with control mice, levels of BUN and serum creatinine were significantly reduced in mice treated with siRNA. Pathological examination demonstrated that tissue damage caused by IRI was markedly reduced as a result of TNF-α siRNA treatment. Furthermore, survival experiments showed that nearly 90% of control mice died from lethal IRI, whereas more than 50% of siRNApretreated mice survived until the end of the eight-day observation period. We have demonstrated for the first time that silencing TNF-α by specific siRNA can significantly reduce renal IRI and protect mice against lethal kidney ischemia, highlighting the potential for siRNA-based clinical therapy.
Collapse
Affiliation(s)
- Ling Hou
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, 430030, China.
- Key Laboratory of Organ Transplantation, Ministry of Public Health, Wuhan, 430030, China.
| | - Biao Feng
- Departments of Surgery, Microbiology and Immunology, and Pathology, University of Western Ontario, London, Canada
| | - Xu-Sheng Zhang
- Departments of Surgery, Microbiology and Immunology, and Pathology, University of Western Ontario, London, Canada
| | - Xiu-Fen Zheng
- Departments of Surgery, Microbiology and Immunology, and Pathology, University of Western Ontario, London, Canada
| | - Ying Xiang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Public Health, Wuhan, 430030, China
| | - Guang-Yuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei-Ping Min
- Departments of Surgery, Microbiology and Immunology, and Pathology, University of Western Ontario, London, Canada
| |
Collapse
|
9
|
Iida S, Tsuda H, Tanaka T, Kish DD, Abe T, Su CA, Abe R, Tanabe K, Valujskikh A, Baldwin WM, Fairchild RL. IL-1 Receptor Signaling on Graft Parenchymal Cells Regulates Memory and De Novo Donor-Reactive CD8 T Cell Responses to Cardiac Allografts. THE JOURNAL OF IMMUNOLOGY 2016; 196:2827-37. [PMID: 26856697 DOI: 10.4049/jimmunol.1500876] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 01/04/2016] [Indexed: 01/03/2023]
Abstract
Reperfusion of organ allografts induces a potent inflammatory response that directs rapid memory T cell, neutrophil, and macrophage graft infiltration and their activation to express functions mediating graft tissue injury. The role of cardiac allograft IL-1 receptor (IL-1R) signaling in this early inflammation and the downstream primary alloimmune response was investigated. When compared with complete MHC-mismatched wild-type cardiac allografts, IL-1R(-/-) allografts had marked decreases in endogenous memory CD8 T cell and neutrophil infiltration and expression of proinflammatory mediators at early times after transplant, whereas endogenous memory CD4 T cell and macrophage infiltration was not decreased. IL-1R(-/-) allograft recipients also had marked decreases in de novo donor-reactive CD8, but not CD4, T cell development to IFN-γ-producing cells. CD8 T cell-mediated rejection of IL-1R(-/-) cardiac allografts took 3 wk longer than wild-type allografts. Cardiac allografts from reciprocal bone marrow reconstituted IL-1R(-/-)/wild-type chimeric donors indicated that IL-1R signaling on graft nonhematopoietic-derived, but not bone marrow-derived, cells is required for the potent donor-reactive memory and primary CD8 T cell alloimmune responses observed in response to wild-type allografts. These studies implicate IL-1R-mediated signals by allograft parenchymal cells in generating the stimuli-provoking development and elicitation of optimal alloimmune responses to the grafts.
Collapse
Affiliation(s)
- Shoichi Iida
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Division of Immunobiology, Research Institute for Biological Science, Science University of Tokyo, Chiba 278-8510, Japan; Department of Urology, Tokyo Women's Medical University, Tokyo 162-0054, Japan
| | - Hidetoshi Tsuda
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Urology, Osaka University School of Medicine, Osaka 565-0871, Japan; and
| | - Toshiaki Tanaka
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195
| | - Danielle D Kish
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195
| | - Toyofumi Abe
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Urology, Osaka University School of Medicine, Osaka 565-0871, Japan; and
| | - Charles A Su
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Ryo Abe
- Division of Immunobiology, Research Institute for Biological Science, Science University of Tokyo, Chiba 278-8510, Japan; Department of Urology, Tokyo Women's Medical University, Tokyo 162-0054, Japan
| | - Kazunari Tanabe
- Division of Immunobiology, Research Institute for Biological Science, Science University of Tokyo, Chiba 278-8510, Japan; Department of Urology, Tokyo Women's Medical University, Tokyo 162-0054, Japan
| | - Anna Valujskikh
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195
| | - William M Baldwin
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Robert L Fairchild
- Department of Immunology, Cleveland Clinic, Cleveland, OH 44195; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| |
Collapse
|
10
|
Gönül Y, Genç A, Ahsen A, Bal A, Hazman Ö, Toktaş M, Ulu MS, Özdinç Ş, Songur A. The effects of IL-18BP on mRNA expression of inflammatory cytokines and apoptotic genes in renal injury induced by infrarenal aortic occlusion. J Surg Res 2015; 202:33-42. [PMID: 27083945 DOI: 10.1016/j.jss.2015.12.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 12/08/2015] [Accepted: 12/16/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Renal injury is an important complication of infrarenal aortic occlusion (IAO), which is mainly encountered during the postoperative period. Aortic clamping procedure may lead to turbulent blood flow and eventually vasoconstriction at renal arterial level of the abdominal aorta. IL-18BP has well-known antioxidant and anti-inflammatory properties. In this study, we aimed to determine whether IL-18BP has anti-inflammatory, antiapoptotic, antioxidant, and protective effects on acute kidney damage induced by IAO rat model. MATERIALS AND METHODS A total of 30 adult male Wistar-Albino rats were equally and randomly separated to three groups as follows: SHAM laparotomy, ischemia-reperfusion (IR), and IR + IL-18BP. We applied 30-min IAO and 2-h reperfusion. Inflammatory cytokine levels (TNF-α, IL-1β, IL-18, IL-6, and IFN-γ) and oxidative stress parameters (TAS, TOS, and OSI) were measured. In addition to this, urea and creatinine levels, histopathology of kidney, mRNA expression levels of inflammatory cytokines, and apoptotic genes were investigated. RESULTS Urea and creatinine, tissue and serum levels of TNF-α, IL-6, IL-18, IFN-γ, and TOS, and oxidative stress index (OSI) were found significantly lower in IR + IL-18BP group, when compared to the IR group. Moreover, mRNA expression levels of inflammatory cytokines and apoptotic genes were prominently depressed in IR + IL-18BP pre-treatment group in histopathologic examination, there was a significant difference between the IR and other three groups (P < 0.001). These improvements were demonstrated with a total score of histopathologic damage. In our previous studies, we have demonstrated that IL-18BP has antioxidant, inflammatory, and protective effects on liver and spinal cord IR injury. Data established from the present study suggest that IL-18BP may exert anti-inflammatory, antiapoptotic, antioxidant, and protective effects on IAO-induced acute kidney injury in rats, and this would be the first study to be conducted in this field. CONCLUSIONS Data established from the present study suggest that IL-18BP may exert anti-inflammatory, antiapoptotic, antioxidant, and protective effects on IAO-induced acute kidney injury in rats.
Collapse
Affiliation(s)
- Yücel Gönül
- Department of Anatomy, Faculty of Medicine, Afyon Kocatepe University, Afyonkarahisar.
| | - Abdurrahman Genç
- Department of Physiology, Faculty of Medicine, Afyon Kocatepe University, Afyonkarahisar
| | - Ahmet Ahsen
- Department of Nephrology, Faculty of Medicine, Afyon Kocatepe University, Afyonkarahisar
| | - Ahmet Bal
- Department of General Surgery, Faculty of Medicine, Afyon Kocatepe University, Afyonkarahisar
| | - Ömer Hazman
- Department of chemistry, Biochemistry Division, Faculty of Science and Arts, Afyon Kocatepe University, Afyonkarahisar
| | - Muhsin Toktaş
- Department of Anatomy, Faculty of Medicine, Turgut Özal University, Ankara, Turkey
| | - M Sena Ulu
- Department of Nephrology, Faculty of Medicine, Afyon Kocatepe University, Afyonkarahisar
| | - Şerife Özdinç
- Department of Emergency Medicine, Faculty of Medicine, Afyon Kocatepe University, Afyonkarahisar
| | - Ahmet Songur
- Department of Anatomy, Faculty of Medicine, Afyon Kocatepe University, Afyonkarahisar
| |
Collapse
|
11
|
Ishida R, Kami D, Kusaba T, Kirita Y, Kishida T, Mazda O, Adachi T, Gojo S. Kidney-specific Sonoporation-mediated Gene Transfer. Mol Ther 2015; 24:125-34. [PMID: 26419704 PMCID: PMC4754547 DOI: 10.1038/mt.2015.171] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 09/07/2015] [Indexed: 12/17/2022] Open
Abstract
Sonoporation can deliver agents to target local organs by systemic administration, while decreasing the associated risk of adverse effects. Sonoporation has been used for a variety of materials and in a variety of organs. Herein, we demonstrated that local sonoporation to the kidney can offer highly efficient transfer of oligonucleotides, which were systemically administrated to the tubular epithelium with high specificity. Ultrasonic wave irradiation to the kidney collapsed the microbubbles and transiently affected the glomerular filtration barrier and increased glomerular permeability. Oligonucleotides were passed through the barrier all at once and were absorbed throughout the tubular epithelium. Tumor necrosis factor alpha (TNFα), which plays a central role in renal ischemia-reperfusion injury, was targeted using small interfering RNA (siRNA) with renal sonoporation in a murine model. The reduction of TNFα expression after single gene transfer significantly inhibited the expression of kidney injury markers, suggesting that systemic administration of siRNA under temporary and local sonoporation could be applicable in the clinical setting of ischemic acute kidney injury.
Collapse
Affiliation(s)
- Ryo Ishida
- Division of Nephrology, Department of Internal Medicine, Graduate School of Medical Science Kyoto Prefectural University of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medical Science Kyoto Prefectural University of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuro Kusaba
- Division of Nephrology, Department of Internal Medicine, Graduate School of Medical Science Kyoto Prefectural University of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuhei Kirita
- Division of Nephrology, Department of Internal Medicine, Graduate School of Medical Science Kyoto Prefectural University of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tsunao Kishida
- Department of Immunology, Graduate School of Medical Science Kyoto Prefectural University of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science Kyoto Prefectural University of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takaomi Adachi
- Division of Nephrology, Department of Internal Medicine, Graduate School of Medical Science Kyoto Prefectural University of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medical Science Kyoto Prefectural University of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
12
|
Chaturvedi S, Robinson LA. Slit2-Robo signaling in inflammation and kidney injury. Pediatr Nephrol 2015; 30:561-6. [PMID: 24777535 DOI: 10.1007/s00467-014-2825-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/03/2014] [Accepted: 04/07/2014] [Indexed: 12/31/2022]
Abstract
Acute kidney injury is an increasingly common global health problem and is associated with severe morbidity and mortality. In addition to facing high mortality rates, the survivors of acute kidney injury are at increased risk of developing chronic kidney disease and end-stage renal disease. Renal ischemia-reperfusion injury (IRI) is the most common cause of acute kidney injury, and results from impaired delivery of oxygen and nutrients to the kidney. Massive leukocyte influx into the post-ischemic kidney is one of the hallmarks of IRI. The recruited leukocytes exacerbate tissue damage and, if uncontrolled, initiate the progressive changes that lead to renal fibrosis and chronic kidney disease. Early on, recruitment and activation of platelets promotes microthrombosis in the injured kidney, further exacerbating kidney damage. The diversity, complexity, and multiplicity of pathways involved in leukocyte recruitment and platelet activation make it extremely challenging to control these processes, and past efforts have met with limited success in human trials. A generalized strategy to inhibit infiltration of inflammatory leukocytes and platelets, thereby reducing inflammation and injury, may prove to be more beneficial. In this review, we summarize recent findings demonstrating that the neuronal guidance cues, Slit and Roundabout (Robo), prevent the migration of multiple leukocyte subsets towards diverse inflammatory chemoattractants, and have potent anti-platelet functions in vitro and in vivo. These properties uniquely position Slit2 as a novel therapeutic that could be used to prevent acute kidney injury associated with IRI.
Collapse
Affiliation(s)
- Swasti Chaturvedi
- Division of Nephrology, Department of Paediatrics, Christian Medical College, Vellore, Tamil Nadu, India
| | | |
Collapse
|
13
|
Visnagri A, Kandhare AD, Bodhankar SL. Renoprotective effect of berberine via intonation on apoptosis and mitochondrial-dependent pathway in renal ischemia reperfusion-induced mutilation. Ren Fail 2015; 37:482-93. [DOI: 10.3109/0886022x.2014.996843] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
14
|
Heitmeier M, McCracken R, Micanovic R, Khan S, El-Achkar TM. The role of tumor necrosis factor alpha in regulating the expression of Tamm-Horsfall Protein (uromodulin) in thick ascending limbs during kidney injury. Am J Nephrol 2014; 40:458-67. [PMID: 25503683 DOI: 10.1159/000369836] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 11/10/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND Tamm-Horsfall Protein (THP) is a glycoprotein expressed exclusively by cells of the thick ascending loop (TAL) of Henle. THP has a protective role in acute kidney injury (AKI), and its expression is downregulated in the early stages of injury. Tumor necrosis factor alpha (TNFα) is a cytokine endogenously expressed by the TAL and is also induced by AKI. Therefore, we hypothesized that TNFα is a key regulator of THP expression. METHODS We used a mouse model of AKI (ischemia-reperfusion injury, IRI) and a cell culture system of a TAL cell line (MKTAL). RESULTS We show that TNFα is upregulated by TAL cells early after AKI in vivo. The expression of THP and its transcription factor Hepatocyte nuclear factor 1β (HNF1β) were concomitantly decreased at the peak of injury. Furthermore, recombinant TNFα inhibits significantly, and in a dose-dependent manner, the expression of THP, but not HNF1β in MKTAL cells. Interestingly, neither TNFα neutralization nor genetic deletion of TNFα increased THP or HNF levels after injury in vivo. CONCLUSION Our data suggest that TNFα can inhibit the expression of THP in TAL cells via an HNF1β-independent mechanism, but the downregulation of THP expression in the early AKI does not depend on TNFα. We propose that TNFα regulates THP expression in a homeostatic setting, but the impact of TNFα on THP during kidney injury is superseded by other factors that could inhibit HNF1β-mediated expression of THP.
Collapse
|
15
|
Hoyer DP, Gallinat A, Swoboda S, Wohlschläger J, Rauen U, Paul A, Minor T. Subnormothermic machine perfusion for preservation of porcine kidneys in a donation after circulatory death model. Transpl Int 2014; 27:1097-106. [PMID: 24963744 DOI: 10.1111/tri.12389] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 05/12/2014] [Accepted: 06/23/2014] [Indexed: 01/26/2023]
Abstract
Machine perfusion for preservation led to compelling success for the outcome of renal transplantation. Further refinements of methods to decrease preservation injury remain an issue of high interest. This study investigates functional and morphological aspects of kidneys preserved by subnormothermic (20 °C) machine perfusion (SNTM) compared with oxygenated hypothermic machine perfusion (HMPox) and cold storage (CS) in a donation after circulatory death (DCD) model. After 30 min of warm ischaemia, porcine kidneys were randomly assigned to preservation for 7 h by CS, HMPox or SNTM. Afterwards, kidneys were reperfused for 2 h with autologous blood in vitro for assessment of function and integrity. Application of SNTM for preservation led to significantly higher blood flow and urine output compared with both other groups. SNTM led to a twofold increased creatinine clearance compared with HMPox and 10-fold increased creatinine clearance compared with CS. Structural integrity was best preserved by SNTM. In conclusion, this is the first study on SNTM for kidneys from DCD donors. SNTM seems to be a promising preservation method with the potential to improve functional parameters of kidneys during reperfusion.
Collapse
Affiliation(s)
- Dieter P Hoyer
- Department of General, Visceral and Transplantation Surgery, University Hospital Essen, Essen, Germany
| | | | | | | | | | | | | |
Collapse
|
16
|
Serine Protease Inhibitor-6 Inhibits Granzyme B–Mediated Injury of Renal Tubular Cells and Promotes Renal Allograft Survival. Transplantation 2014; 98:402-10. [DOI: 10.1097/tp.0000000000000237] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
17
|
Secher N, Soendergaard P, Ravlo K, Soelling C, Granfeldt A, Wogensen L, Keller AK, Moeldrup U, Ostraat EO, Joergensen TM, Jespersen B, Toennesen E. No effect of remote ischaemic conditioning on inflammation in a porcine kidney transplantation model. Transpl Immunol 2014; 31:98-104. [DOI: 10.1016/j.trim.2014.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/26/2014] [Accepted: 05/27/2014] [Indexed: 02/04/2023]
|
18
|
The effect of mTOR-inhibition on NF-κB activity in kidney ischemia-reperfusion injury in mice. Transplant Proc 2014; 45:1708-14. [PMID: 23769029 DOI: 10.1016/j.transproceed.2013.02.110] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 02/07/2013] [Accepted: 02/27/2013] [Indexed: 02/05/2023]
Abstract
Kidney ischemia-reperfusion injury (IRI) is associated with a robust inflammatory response, which is regulated by nuclear factor-kappaB (NF-κB), mainly its heterodimeric form p65/p50. Considering immunomodulatory properties of mammalian target of rapamycin (mTOR) inhibitors, the effect of everolimus on NF-κB activation in kidney IRI was determined in this study. IRI was induced in C57/BL6 mice by clamping both renal pedicles for 45 minutes. Application of everolimus (0.25 mg/kg bw subcutaneously daily) was started one day before IRI induction. Both everolimus-treated and nontreated mice were sacrificed at several times starting at 30 minutes and finishing on day 7 after IRI induction. The NF-κB activity, proinflammatory cytokines IL-1β, TNF-α, and anti-inflammatory cytokine IL-10 production were determined in kidneys. Compared with nontreated animals, everolimus-treated animals showed significantly increased TNF-α (2741.6 ± 201.72 pg/mg; 1925 ± 185.81 pg/mg, P < .05) and IL-1β (11.47 ± 1.2 pg/mg; 4.3 ± 0.13 pg/mg, P < .01) production on day 2 after IRI induction accompanied by significantly greater NF-κB/DNA binding activity and p65 nuclear expression (P < .01). Two hours after IRI induction, everolimus-treated animals showed significantly increased IL-1β mRNA expression (P < .05) followed by increased IL-1β protein concentrations when compared with nontreated animals measured 6 hours after IRI induction (11.71 ± 1.5 pg/mg; 7.5 ± 1.11 pg/mg, P < .01). Both experimental groups showed increased NF-κB/DNA binding activity at 7 days after IRI induction. Significantly increased nuclear p65 expression was measured in nontreated animals (P < .01), whereas everolimus-treated hosts showed significantly increased nuclear RelB expression (P < .01). These data suggested that everolimus potentiated innate immunity in the early phase of IRI, stimulating the production of NF-κB-driven proinflammatory cytokines such as TNF-α and IL-1β. The NF-κB activity was potentiated under m-TOR inhibition during kidney IRI, implicating a possible beneficial role of alternative NF-κB activation during the repair phase.
Collapse
|
19
|
Renal atrophy after ischemia–reperfusion injury depends on massive tubular apoptosis induced by TNFα in the later phase. Med Mol Morphol 2014; 47:213-23. [DOI: 10.1007/s00795-013-0067-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 12/11/2013] [Indexed: 10/25/2022]
|
20
|
Adachi T, Sugiyama N, Gondai T, Yagita H, Yokoyama T. Blockade of Death Ligand TRAIL Inhibits Renal Ischemia Reperfusion Injury. Acta Histochem Cytochem 2013; 46:161-70. [PMID: 24610963 PMCID: PMC3929614 DOI: 10.1267/ahc.13022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 10/04/2013] [Indexed: 12/23/2022] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is a leading cause of acute kidney injury (AKI). Many investigators have reported that cell death via apoptosis significantly contributed to the pathophysiology of renal IRI. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the tumor necrosis factor superfamily, and induces apoptosis and inflammation. However, the role of TRAIL in renal IRI is unclear. Here, we investigated whether TRAIL contributes to renal IRI and whether TRAIL blockade could attenuate renal IRI. AKI was induced by unilateral clamping of the renal pedicle for 60 min in male FVB/N mice. We found that the expression of TRAIL and its receptors were highly upregulated in renal tubular cells in renal IRI. Neutralizing anti-TRAIL antibody or its control IgG was given 24 hr before ischemia and a half-dose booster injection was administered into the peritoneal cavity immediately after reperfusion. We found that TRAIL blockade inhibited tubular apoptosis and reduced the accumulation of neutrophils and macrophages. Furthermore, TRAIL blockade attenuated renal fibrosis and atrophy after IRI. In conclusion, our study suggests that TRAIL is a critical pathogenic factor in renal IRI, and that TRAIL could be a new therapeutic target for the prevention of renal IRI.
Collapse
Affiliation(s)
- Takaomi Adachi
- Department of Anatomy and Developmental Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Noriyuki Sugiyama
- Department of Anatomy and Developmental Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
- Department of Anatomy and Cell Biology, Division of Life Science, Osaka Medical College
| | - Tatsuro Gondai
- Department of Anatomy and Developmental Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine
| | - Takahiko Yokoyama
- Department of Anatomy and Developmental Biology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| |
Collapse
|
21
|
Yoshida T, Kumagai H, Kohsaka T, Ikegaya N. Relaxin protects against renal ischemia-reperfusion injury. Am J Physiol Renal Physiol 2013; 305:F1169-76. [PMID: 23946288 DOI: 10.1152/ajprenal.00654.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Relaxin, a pregnancy hormone, has antiapoptotic and anti-inflammatory properties. The aim of this study was to determine the effects of relaxin on ischemia-reperfusion (IR)-induced acute kidney injury. Male rats underwent unilateral nephrectomy and contralateral renal IR (45 min of renal pedicle clamping). Rats were divided into three groups: 1) sham group, 2) IR group, and 3) IR-RLX group (rats treated with relaxin before ischemia). In this group, relaxin was infused at 500 ng/h via subcutaneous osmotic minipump for 24 h beginning 2 h before renal ischemia. At 24 h after reperfusion, renal function was assessed and kidneys were removed for analysis. There was no significant difference in blood pressure among the three groups. IR increased plasma levels of creatinine and urea nitrogen, and relaxin provided protection against the increases in these two parameters. Relaxin significantly decreased plasma TNF-α levels and renal TNF receptor 1 mRNA expression, compared with the IR group. Semiquantitative assessment of the histological lesions showed marked structural damage in IR rats compared with the IR-RLX rats. RLX significantly reduced apoptotic cell counts compared with the IR group. Overexpression of caspase-3 observed in the IR kidneys was reduced in the IR-RLX group. The results demonstrated that relaxin provided protection against IR-induced renal injury by reducing apoptosis and inflammation.
Collapse
Affiliation(s)
- Takuya Yoshida
- Dept. of Medicine, Yaizu Municipal General Hospital, 1000, Dohbara, Yaizu, Shizuoka, 425-8505, Japan.
| | | | | | | |
Collapse
|
22
|
Rostami Z, Einollahi B, Ghadiani MH. Does living donor hyperoxia have an impact on kidney graft function after transplantation? Nephrourol Mon 2013; 5:835-9. [PMID: 24282796 PMCID: PMC3830912 DOI: 10.5812/numonthly.11870] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 05/06/2013] [Indexed: 11/16/2022] Open
Abstract
Background Improvement in the outcome of organ transplantation is related to advances in patient selection criteria, organ preservation, operative techniques, perioperative care and efficacy of immunosuppressive agents. Objectives We aimed to evaluate the effects of higher levels of arterial PaO2 in donors on DGF (delayed graft function). Patients and Methods Forty patients over 18 years old with stage 4-5 chronic kidney disease (CKD) who received a kidney from living donors were enrolled. They were randomly grouped in to the case (n = 17) and control (n = 23) groups and were followed for 2 weeks after transplantation. Donors were exposed to 60% oxygen for at least 2 hours with a face-mask (venture mask) for 2 consecutive days before transplantation until arterial oxygen pressure increased in arterial blood gas to 200 mmHg. Neutrophil gelatinase associated lipocalin (NGAL), Interleuk-18 (IL-18), tumor necrosis factor- α (TNF-α) and transforming growth factor–β (TGF-β) could be good biomarkers for early diagnosis of kidney injury in renal transplant recipients; we assessed kidney function with these biomarkers. Results Forty living kidney transplantations including 17 cases and 23 controls were performed; female gender was more prevalent in recipients (n = 16, 40%). The mean age of recipients was 36.1 ± 12.4 (18-67) years old. DGF was detected in 2 (5.95%) individuals, from whom one was in the case group and the other one in the control group. In the univariate analysis, there was no significant correlation between age and biomarkers in urine and serum unless for the second serum NGAL (P = 0.02, r = -0.06) and second urine IL 18 (P = 0.03, r = -0.5) which had a negative correlation, and first urine TNF α (P = 0.02, r = 0.7) which had a positive correlation. Conclusions Oxygen therapy in the case group had no significant impact on protection from DGF.
Collapse
Affiliation(s)
- Zohreh Rostami
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
- Corresponding author: Zohreh Rostami, Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran. Tel: +98-9121544897, Fax: +98-2181262073, E-mail:
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, IR Iran
| | | |
Collapse
|
23
|
Kohei N, Tanabe T, Horita S, Omoto K, Ishida H, Yamaguchi Y, Tanabe K. Sequential analysis of donor-specific antibodies and pathological findings in acute antibody-mediated rejection in a rat renal transplantation model. Kidney Int 2013; 84:722-32. [PMID: 23615506 DOI: 10.1038/ki.2013.117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 01/25/2013] [Accepted: 01/31/2013] [Indexed: 11/09/2022]
Abstract
Alloantibodies contribute significantly to renal transplant rejection by activation of complement and various cytokines with a variety of effector cells, and are a major cause of allograft loss. Although there is clinical evidence of antibody- and complement-mediated injury in renal transplantation, the mechanism of antibody-mediated rejection remains largely unknown. In order to understand the sequential production of antibodies and complement components, we presensitized recipient rats by skin transplantation. Anti-donor-specific IgG levels reached a maximum 2 weeks following presensitization after which the rats underwent renal transplantation from the same donor strain. We then evaluated sequential pathological findings based on the Banff classification and several factors related to graft rejection. In this presensitized model, peritubular capillaries were already dilated and stained for C4d. Neutrophil and mononuclear cell infiltration in these capillaries was detected beginning 2 h after transplantation. Donor-specific antibody IgG levels decreased rapidly and anti-IgG antibody stained glomerular and peritubular capillaries in the grafts beginning 2 h after transplantation. Additionally, several cytokines and complement components showed marked changes in the presensitized group. Thus, in the donor-specific presensitized recipient, alloantibodies and complement were activated immediately after transplant. C4d deposition in peritubular capillaries appears to be a key factor for the diagnosis of antibody-associated rejection.
Collapse
Affiliation(s)
- Naoki Kohei
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
24
|
Gao G, Zhang B, Ramesh G, Betterly D, Tadagavadi RK, Wang W, Reeves WB. TNF-α mediates increased susceptibility to ischemic AKI in diabetes. Am J Physiol Renal Physiol 2013; 304:F515-21. [PMID: 23283990 DOI: 10.1152/ajprenal.00533.2012] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Diabetes is a risk factor for the development of acute kidney injury (AKI) in humans and rodents. However, the mechanistic basis for this observation is unknown. The present studies evaluated the role of inflammation and TNF-α in ischemic AKI in a model of type 2 diabetes mellitus (DM). Diabetic (db/db) and nondiabetic (db/+) littermates were subjected to 20 min of bilateral renal ischemia. The nondiabetic mice developed only mild and transient renal dysfunction. In contrast, the equivalent ischemic insult provoked severe and sustained renal dysfunction in the db/db mice. The expression of TNF-α and Toll-like receptor 4 (TLR4) mRNA was measured in the kidneys of diabetic mice before and after renal ischemia; db/db mice exhibited greater increases in TNF-α and TLR4 mRNA expression following ischemia than did db/+. In addition, urinary excretion of TNF-α after ischemia was higher in db/db mice than in db/+ mice. To determine the possible role of TNF-α in mediating the enhanced susceptibility of diabetic mice to ischemic injury, db/db mice were injected with either a neutralizing anti-mouse TNF-α antibody or nonimmune globulin and then subjected to 20 min of bilateral renal ischemia. Treatment of the db/db mice with the TNF-α antibody provided significant protection against the ischemic injury. These data support the view that diabetes increases the susceptibility to ischemia-induced renal dysfunction. This increased susceptibility derives from a heightened inflammatory response involving TNF-α and perhaps TLR4 signaling.
Collapse
Affiliation(s)
- Guofeng Gao
- Div. of Nephrology, Rm. C5830, Penn State College of Medicine, 500 Univ. Dr., Hershey, PA 17033, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
Heemann U, Viklicky O. The role of belataceptin transplantation: results and implications of clinical trials in the context of other new biological immunosuppressant agents. Clin Transplant 2012. [DOI: 10.1111/ctr.12044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Uwe Heemann
- Department of Nephrology; Klinikum Rechts der Isar der; Technischen Universität München; München; Germany
| | - Ondrej Viklicky
- Department of Nephrology, Transplant Center; Institute for Clinical and Experimental Medicine; Prague; Czech Republic
| |
Collapse
|
26
|
Miyazaki H, Morishita J, Ueki M, Nishina K, Shiozawa S, Maekawa N. The effects of a selective inhibitor of c-Fos/activator protein-1 on endotoxin-induced acute kidney injury in mice. BMC Nephrol 2012; 13:153. [PMID: 23173923 PMCID: PMC3557146 DOI: 10.1186/1471-2369-13-153] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 11/18/2012] [Indexed: 12/17/2022] Open
Abstract
Background Sepsis has been identified as the most common cause of acute kidney injury (AKI) in intensive care units. Lipopolysaccharide (LPS) induces the production of several proinflammatory cytokines including tumor necrosis factor (TNF)-alpha, a major pathogenetic factor in septic AKI. c-Fos/activator protein (AP)-1 controls the expression of these cytokines by binding directly to AP-1 motifs in the cytokine promoter regions. T-5224 is a new drug developed by computer-aided drug design that selectively inhibits c-Fos/AP-1 binding to DNA. In this study, we tested whether T-5224 has a potential inhibitory effect against LPS-induced AKI, by suppressing the TNF-alpha inflammatory response and other downstream effectors. Methods To test this hypothesis, male C57BL/6 mice at 7 weeks old were divided into three groups (control, LPS and T-5224 groups). Mice in the control group received saline intraperitoneally and polyvinylpyrrolidone solution orally. Mice in the LPS group were injected intraperitoneally with a 6 mg/kg dose of LPS and were given polyvinylpyrrolidone solution immediately after LPS injection. In the T-5224 group, mice were administered T-5224 orally at a dose of 300 mg/kg immediately after LPS injection. Serum concentrations of TNF-alpha, interleukin (IL)-1beta, IL-6 and IL-10 were measured by ELISA. Moreover, the expression of intercellular adhesion molecule (ICAM)-1 mRNA in kidney was examined by quantitative real-time RT-PCR. Finally, we evaluated renal histological changes. Results LPS injection induced high serum levels of TNF-alpha, IL-1beta and IL-6. However, the administration of T-5224 inhibited the LPS-induced increase in these cytokine levels. The serum levels of IL-10 in the LPS group and T-5224 group were markedly elevated compared with the control group. T-5224 also inhibited LPS-induced ICAM-1 mRNA expression. Furthermore histological studies supported an anti-inflammatory role of T-5224. Conclusions In endotoxin-induced AKI, T-5224 inhibited the production of TNF-alpha and other downstream effectors. In contrast, T-5224 did not inhibit IL-10, an anti-inflammatory cytokine. These data support that the use of T-5224 is a promising new treatment for septic kidney injury.
Collapse
Affiliation(s)
- Hiroyuki Miyazaki
- Division of Anesthesiology and Perioperative Medicine, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Cardiac surgery-associated acute kidney injury (AKI) is a major health problem that is extremely common and has a significant effect on cardiac surgical outcomes. AKI occurs in nearly 30 % of patients undergoing cardiac surgery, with about 1-2 % of these ultimately requiring dialysis. The development of AKI predicts a significant increase in morbidity and mortality independent of other risk factors. The pathogenetic mechanisms associated with cardiac surgery-associated AKI include several biochemical pathways, of which the most important are hemodynamic, inflammatory and nephrotoxic factors. Risk factors for AKI have been identified in several models, and these facilitate physicians to prognosticate and develop a strategy for tackling patients predisposed to developing renal dysfunction. Effective therapy of the condition is still suboptimal, and hence the accent has always been on risk factor modification. Thus, strategies for reducing preoperative anemia, perioperative blood transfusions and surgical re-explorations may be effective in attenuating the incidence and severity of this complication.
Collapse
Affiliation(s)
- Satyen Parida
- Department of Anesthesiology and Critical Care, Jawaharlal Institute of Postgraduate Medical Education & Research, Dhanvantari Nagar, Pondicherry, India.
| | | |
Collapse
|
28
|
Zhang ZX, Min WP, Jevnikar AM. Use of RNA interference to minimize ischemia reperfusion injury. Transplant Rev (Orlando) 2012; 26:140-55. [DOI: 10.1016/j.trre.2011.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 03/22/2011] [Indexed: 12/21/2022]
|
29
|
Rodrigues CE, Sanches TR, Volpini RA, Shimizu MHM, Kuriki PS, Camara NOS, Seguro AC, Andrade L. Effects of continuous erythropoietin receptor activator in sepsis-induced acute kidney injury and multi-organ dysfunction. PLoS One 2012; 7:e29893. [PMID: 22235348 PMCID: PMC3250486 DOI: 10.1371/journal.pone.0029893] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 12/06/2011] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Despite advances in supportive care, sepsis-related mortality remains high, especially in patients with acute kidney injury (AKI). Erythropoietin can protect organs against ischemia and sepsis. This effect has been linked to activation of intracellular survival pathways, although the mechanism remains unclear. Continuous erythropoietin receptor activator (CERA) is an erythropoietin with a unique pharmacologic profile and long half-life. We hypothesized that pretreatment with CERA would be renoprotective in the cecal ligation and puncture (CLP) model of sepsis-induced AKI. METHODS RATS WERE RANDOMIZED INTO THREE GROUPS: control; CLP; and CLP+CERA (5 µg/kg body weight, i.p. administered 24 h before CLP). At 24 hours after CLP, we measured creatinine clearance, biochemical variables, and hemodynamic parameters. In kidney tissue, we performed immunoblotting--to quantify expression of the Na-K-2Cl cotransporter (NKCC2), aquaporin 2 (AQP2), Toll-like receptor 4 (TLR4), erythropoietin receptor (EpoR), and nuclear factor kappa B (NF-κB)--and immunohistochemical staining for CD68 (macrophage infiltration). Plasma interleukin (IL)-2, IL-1β, IL-6, IL-10, interferon gamma, and tumor necrosis factor alpha were measured by multiplex detection. RESULTS Pretreatment with CERA preserved creatinine clearance and tubular function, as well as the expression of NKCC2 and AQP2. In addition, CERA maintained plasma lactate at normal levels, as well as preserving plasma levels of transaminases and lactate dehydrogenase. Renal expression of TLR4 and NF-κB was lower in CLP+CERA rats than in CLP rats (p<0.05 and p<0.01, respectively), as were CD68-positive cell counts (p<0.01), whereas renal EpoR expression was higher (p<0.05). Plasma levels of all measured cytokines were lower in CLP+CERA rats than in CLP rats. CONCLUSION CERA protects against sepsis-induced AKI. This protective effect is, in part, attributable to suppression of the inflammatory response.
Collapse
Affiliation(s)
- Camila E Rodrigues
- Department of Nephrology, University of São Paulo School of Medicine, São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Pech T, Fujishiro J, Finger T, Ohsawa I, Praktiknjo M, von Websky M, Wehner S, Abu-Elmagd K, Kalff JC, Schaefer N. Perioperative infliximab application has marginal effects on ischemia-reperfusion injury in experimental small bowel transplantation in rats. Langenbecks Arch Surg 2011; 397:131-40. [PMID: 21960137 DOI: 10.1007/s00423-011-0853-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 09/14/2011] [Indexed: 12/16/2022]
Abstract
PURPOSE Ischemia-reperfusion injury leads to impaired smooth muscle function and inflammatory reactions after intestinal transplantation. In previous studies, infliximab has been shown to effectively protect allogenic intestinal grafts in the early phase after transplantation with resulting improved contractility. This study was designed to reveal protective effects of infliximab on ischemia-reperfusion injury in isogenic transplantation. METHODS Isogenic, orthotopic small bowel transplantation was performed in Lewis rats (3 h cold ischemia). Five groups were defined: non-transplanted animals with no treatment (group 1), isogenic transplanted animals with vehicle treatment (groups 2/3) or with infliximab treatment (5 mg/kg body weight intravenously, directly after reperfusion; groups 4/5). The treated animals were sacrificed after 3 (group 2/4) or 24 h (group 3/5). Histological and immunohistochemical analysis, TUNEL staining, real-time RT-PCR, and contractility measurements in a standard organ bath were used for determination of ischemia-reperfusion injury. RESULTS All transplanted animals showed reduced smooth muscle function, while no significant advantage of infliximab treatment was observed. Reduced infiltration of neutrophils was noted in the early phase in animals treated with infliximab. The structural integrity of the bowel and infiltration of ED1-positive monocytes and macrophages did not improve with infliximab treatment. At 3 h after reperfusion, mRNA expression of interleukin (IL)-6, TNF-α, IL-10, and iNOS and MCP-1 displayed increased activation in the infliximab group. CONCLUSION The protective effects of infliximab in the early phase after experimental small bowel transplantation seem to be unrelated to ischemia-reperfusion injury. The promising effects in allogenic transplantation indicate the need for further experiments with infliximab as complementary treatment under standard immunosuppressive therapy. Further experiments should focus on additional infliximab treatment in the setting of acute rejection.
Collapse
Affiliation(s)
- T Pech
- Department of Surgery, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Efrati S, Berman S, Hamad RA, Siman-Tov Y, Ilgiyaev E, Maslyakov I, Weissgarten J. Effect of captopril treatment on recuperation from ischemia/reperfusion-induced acute renal injury. Nephrol Dial Transplant 2011; 27:136-45. [PMID: 21680852 DOI: 10.1093/ndt/gfr256] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Ischemia/reperfusion triggers acute kidney injury (AKI), mainly via aggravating hypoxia, oxidative stress, inflammation and renin-angiotensin system (RAS) activation. We investigated the role of angiotensin-converting enzyme (ACE) inhibition on the progression of AKI in a rat model of ischemia/reperfusion. METHODS Ninety-nine Sprague-Dawley rats were subjected to 1 h ischemia/reperfusion and/or left unilateral nephrectomy, with concurrent intraperitoneal implantation of Alzet pump. Via this pump, they were continuously infused with captopril 0.5 mg/kg/day, captopril 2 mg/kg/day or saline. The rats were sacrificed following 24, 48 or 168 h. Blood samples, 24-h urine collections and kidneys were allocated, to evaluate renal function, angiotensin-II, nitric oxide (NO), apoptosis, hypoxia, oxidative stress and inflammation. RESULTS Serum creatinine and cystatin-C significantly increased in ischemic rats, coinciding with histopathologic intrarenal damage, decreased NO, augmented angiotensin-II, interleukin (IL)-6, IL-10, transforming growth factor-beta. At the acute reperfusion stage, captopril prevented excessive angiotensin-II synthesis, ameliorated renal dysfunction, inhibited intrarenal inflammation and improved histopathologic findings. Most of the renoprotective effects of captopril were limited predominantly to acute reperfusion stage. Concurrently, captopril significantly decreased NO availability, exacerbated intrarenal hypoxia and augmented oxidative stress. CONCLUSIONS At the acute stage of renal ischemia/reperfusion-induced AKI, ACE inhibition substantially contributed to the amelioration of acute injury by improving renal function, inhibiting systemic and intrarenal angiotensin-II, attenuating intrarenal inflammation and preserving renal tissue structure. Later on, at the post-reperfusion stage, most of the beneficial effects of captopril administration on the recuperating post-ischemic kidney were no longer evident. Concurrently, ACE inhibition exacerbated intrarenal hypoxia and accelerated oxidative stress, indicating that renal adaptation to some consequences of ischemia does require bioavailability of RAS components.
Collapse
Affiliation(s)
- Shai Efrati
- Research & Development Unit, Assaf Harofeh Medical Center, Zerifin 70300, Tel Aviv University, Israel.
| | | | | | | | | | | | | |
Collapse
|
32
|
Effect of Ligusticum wallichii aqueous extract on oxidative injury and immunity activity in myocardial ischemic reperfusion rats. Int J Mol Sci 2011; 12:1991-2006. [PMID: 21673935 PMCID: PMC3111646 DOI: 10.3390/ijms12031991] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 02/22/2011] [Accepted: 03/09/2011] [Indexed: 11/17/2022] Open
Abstract
We investigated the efficacy of Ligusticum wallichi aqueous extract (LWE) for myocardial protection against ischemia-reperfusion injury. Rats were fed for five weeks with either a control diet (sham and ischemia reperfusion (IR) model control groups) or a diet mixed with 0.2%, 0.4% or 0.6% Ligusticum wallichi extract. At the end of the five week period, hearts were excised and subjected to global ischemia for 30 min followed by reperfusion for 2 h. The hearts were compared for indices of oxidative stress and immunity activities. Administration of Ligusticum wallichi extract significantly decreased serum TNF-α, IL-6, IL-8, NO, MIP-1α, CRP and myocardium MDA levels, and serum CK, LDH and AST activities, and increased myocardium Na+-K+-ATPase, Ca2+-Mg2+-ATPase, NOS, SOD, CAT, GSH-Px and TAOC activities. The results indicate that Ligusticum wallichii extract treatment can enhance myocardial antioxidant status and improve the immunity profile in ischemic-reperfusion rats.
Collapse
|
33
|
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: 104] [Impact Index Per Article: 7.4] [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
|
34
|
TNF superfamily: a growing saga of kidney injury modulators. Mediators Inflamm 2010; 2010. [PMID: 20953353 PMCID: PMC2952810 DOI: 10.1155/2010/182958] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 08/31/2010] [Accepted: 09/06/2010] [Indexed: 12/30/2022] Open
Abstract
Members of the TNF superfamily participate in kidney disease. Tumor necrosis factor (TNF) and Fas ligand regulate renal cell survival and inflammation, and therapeutic targeting improves the outcome of experimental renal injury. TNF-related apoptosis-inducing ligand (TRAIL and its potential decoy receptor osteoprotegerin are the two most upregulated death-related genes in human diabetic nephropathy. TRAIL activates NF-kappaB in tubular cells and promotes apoptosis in tubular cells and podocytes, especially in a high-glucose environment. By contrast, osteoprotegerin plays a protective role against TRAIL-induced apoptosis. Another family member, TNF-like weak inducer of apoptosis (TWEAK induces inflammation and tubular cell death or proliferation, depending on the microenvironment. While TNF only activates canonical NF-kappaB signaling, TWEAK promotes both canonical and noncanonical NF-kappaB activation in tubular cells, regulating different inflammatory responses. TWEAK promotes the secretion of MCP-1 and RANTES through NF-kappaB RelA-containing complexes and upregulates CCl21 and CCL19 expression through NF-kappaB inducing kinase (NIK-) dependent RelB/NF-kappaB2 complexes. In vivo TWEAK promotes postnephrectomy compensatory renal cell proliferation in a noninflammatory milieu. However, in the inflammatory milieu of acute kidney injury, TWEAK promotes tubular cell death and inflammation. Therapeutic targeting of TNF superfamily cytokines, including multipronged approaches targeting several cytokines should be further explored.
Collapse
|
35
|
Tadagavadi RK, Reeves WB. Endogenous IL-10 attenuates cisplatin nephrotoxicity: role of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2010; 185:4904-11. [PMID: 20844196 DOI: 10.4049/jimmunol.1000383] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sterile inflammation is associated with tissue injury and organ failure. Recent studies indicate that certain endogenous cytokines and immune cells may limit tissue injury by reducing immune-mediated inflammatory responses. Cisplatin is a commonly used anticancer chemotherapeutic agent but causes acute kidney injury and dysfunction. In a recent study, we showed that renal dendritic cells attenuate cisplatin-induced kidney injury by reducing inflammation. In this study, we investigated the effect of endogenous IL-10 and dendritic cell IL-10 in cisplatin-mediated kidney injury. Cisplatin treatment caused increases in renal IL-10R1 expression and STAT3 phosphorylation. In response to cisplatin treatment, IL-10 knockout mice showed more rapid and greater increases in blood urea nitrogen and serum creatinine compared with wild-type mice, indicating that endogenous IL-10 ameliorates kidney injury in cisplatin nephrotoxicity. Renal infiltration of IFN-γ-producing neutrophils was markedly increased in IL-10 knockout mice compared with wild-type mice. However, IFN-γ neutralization had no impact on renal dysfunction, suggesting IFN-γ-independent mechanisms of tissue injury in cisplatin nephrotoxicity. Renal dendritic cells showed high expression of IL-10 in response to cisplatin treatment. We further investigated the effect of dendritic cell-derived IL-10 in cisplatin nephrotoxicity using a conditional cell ablation approach. Mixed bone marrow chimeric mice lacking IL-10 in dendritic cells showed moderately greater renal dysfunction than chimeric mice positive for IL-10 in dendritic cells. These data demonstrate that endogenous IL-10 reduces cisplatin nephrotoxicity and associated inflammation. Moreover, IL-10 produced by dendritic cells themselves accounts for a portion of the protective effect of dendritic cells in cisplatin nephrotoxicity.
Collapse
|
36
|
Expression of transforming growth factor-beta1 limits renal ischemia-reperfusion injury. Transplantation 2010; 89:1320-7. [PMID: 20458271 DOI: 10.1097/tp.0b013e3181d8e9dc] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Renal ischemia-reperfusion injury (IRI) largely contributes to kidney transplant dysfunction and acute kidney injury, but its pathogenesis is not fully understood. In this study, the role of transforming growth factor (TGF)-beta1 in renal IRI is investigated using TGF-beta1 deficient mice. METHOD Human renal tubular epithelial cells (TEC) line (HK-2) was used as an in vitro model, and cell apoptosis was determined by flow cytometric analysis. Renal IRI was induced in mice by clamping renal vein and artery for 45 min at 32 degrees C. RESULTS Here, we showed that in cultures of HK-2 cells, TGF-beta1 expression was up-regulated by tumor necrosis factor (TNF)-alpha. Neutralization of TGF-beta1 activity increased both spontaneous and TNF-alpha-mediated apoptosis, and knockdown of TGF-beta1 expression increased the sensitivity of cell apoptosis to TNF-alpha. In a mouse model of renal IRI, a deficiency in TGF-beta1 expression increased the severity of renal injury, as indicated by more severe renal tubular damage, higher levels of serum creatinine or blood urea nitrogen in TGF-beta1 deficient mice as compared with those in wild-type controls. Further experiments showed that the antiapoptosis of TGF-beta1 correlated with up-regulation of Bcl-2 in kidney cells. CONCLUSION Expression of TGF-beta1 in TECs, potentially induced by proinflammatory TNF-alpha, renders TECs resistance to cell death. In mice, TGF-beta1 deficiency results in more prone to IRI. These data imply that TGF-beta1 may act as a feedback survival factor in the resistance to kidney injury and maintenance of epithelium homeostasis.
Collapse
|
37
|
Grootjans J, Lenaerts K, Derikx JPM, Matthijsen RA, de Bruïne AP, van Bijnen AA, van Dam RM, Dejong CHC, Buurman WA. Human intestinal ischemia-reperfusion-induced inflammation characterized: experiences from a new translational model. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2283-91. [PMID: 20348235 DOI: 10.2353/ajpath.2010.091069] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human intestinal ischemia-reperfusion (IR) is a frequent phenomenon carrying high morbidity and mortality. Although intestinal IR-induced inflammation has been studied extensively in animal models, human intestinal IR induced inflammatory responses remain to be characterized. Using a newly developed human intestinal IR model, we show that human small intestinal ischemia results in massive leakage of intracellular components from ischemically damaged cells, as indicated by increased arteriovenous concentration differences of intestinal fatty acid binding protein and soluble cytokeratin 18. IR-induced intestinal barrier integrity loss resulted in free exposure of the gut basal membrane (collagen IV staining) to intraluminal contents, which was accompanied by increased arteriovenous concentration differences of endotoxin. Western blot for complement activation product C3c and immunohistochemistry for activated C3 revealed complement activation after IR. In addition, intestinal IR resulted in enhanced tissue mRNA expression of IL-6, IL-8, and TNF-alpha, which was accompanied by IL-6 and IL-8 release into the circulation. Expression of intercellular adhesion molecule-1 was markedly increased during reperfusion, facilitating influx of neutrophils into IR-damaged villus tips. In conclusion, this study for the first time shows the sequelae of human intestinal IR-induced inflammation, which is characterized by complement activation, production and release of cytokines into the circulation, endothelial activation, and neutrophil influx into IR-damaged tissue.
Collapse
Affiliation(s)
- Joep Grootjans
- Department of Surgery, NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Jang HR, Gandolfo MT, Ko GJ, Satpute SR, Racusen L, Rabb H. B cells limit repair after ischemic acute kidney injury. J Am Soc Nephrol 2010; 21:654-65. [PMID: 20203156 DOI: 10.1681/asn.2009020182] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
There is no established modality to repair kidney damage resulting from ischemia-reperfusion injury (IRI). Early responses to IRI involve lymphocytes, but the role of B cells in tissue repair after IRI is unknown. Here, we examined B cell trafficking into postischemic mouse kidneys and compared the repair response between control (wild-type) and muMT (B cell-deficient) mice with and without adoptive transfer of B cells. B cells infiltrated postischemic kidneys and subsequently activated and differentiated to plasma cells during the repair phase. Plasma cells expressing CD126 increased and B-1 B cells trafficked into postischemic kidneys with distinct kinetics. An increase in B lymphocyte chemoattractant in the kidney preceded B cell trafficking. Postischemic kidneys of muMT mice expressed higher IL-10 and vascular endothelial growth factor and exhibited more tubular proliferation and less tubular atrophy. Adoptive transfer of B cells into muMT mice reduced tubular proliferation and increased tubular atrophy. Treatment with anti-CD126 antibody increased tubular proliferation and reduced tubular atrophy in the late repair phase. These results demonstrate that B cells may limit the repair process after kidney IRI. Targeting B cells could have therapeutic potential to improve repair after IRI.
Collapse
Affiliation(s)
- Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Acute kidney injury (AKI) continues to contribute significantly to morbidity and mortality in the ICU setting, especially when associated with distant organ dysfunction. There is increasing evidence that AKI directly contributes to organ dysfunction in lung, brain, liver, heart and other organs. This review will examine our current understanding of the deleterious organ crosstalk in the critically ill, which can provide a framework for developing novel therapeutics. RECENT FINDINGS The majority of studies correlating AKI with distant organ dysfunction have demonstrated the pathophysiological importance of proinflammatory and proapoptotic pathways as well as oxidative stress and reactive oxygen species (ROS) production. Leukocyte activation and infiltration, changes in levels of soluble factors such as cytokines and chemokines, and regulation of cell death in extra-renal organs are potentially important mechanisms by which AKI modulates multiorgan dysfunction. SUMMARY There is increasing knowledge of AKI and deleterious interorgan crosstalk that arises, at least in part, due to the imbalance of immune, inflammatory, and soluble mediator metabolism that attends severe insults to the kidney. Further studies can build on these new mechanistic observations to develop strategies to improve outcomes in the critically ill patient.
Collapse
Affiliation(s)
- Xiang Li
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
40
|
Ishii D, Schenk AD, Baba S, Fairchild RL. Role of TNFalpha in early chemokine production and leukocyte infiltration into heart allografts. Am J Transplant 2010; 10:59-68. [PMID: 19958333 PMCID: PMC2806937 DOI: 10.1111/j.1600-6143.2009.02921.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The acute phase cytokines IL-1beta, IL-6 and TNFalpha are produced early during inflammatory processes, including ischemia-reperfusion. The appearance and role of these cytokines in the early inflammation following reperfusion of grafts remain poorly defined. This study investigated the role of TNFalpha in the induction of early leukocyte infiltration into vascularized heart allografts. TNFalpha and IL-6 mRNA levels reached an initial peak 3 h posttransplant and a second peak at 9-12 h with equivalent levels in iso- and allografts. A single dose of anti-TNFalpha mAb given at reperfusion decreased neutrophil and macrophage chemoattractant levels and early neutrophil, macrophage and memory CD8 T-cell infiltration into allografts. Anti-TNFalpha mAb also extended graft survival from 8.6+/-0.6 days to 14.1+/-0.8 days. When assessed on day 7 posttransplant, the number of donor-reactive CD8 T cells producing IFN-gamma in the spleen was reduced almost 70% in recipients treated with anti-TNFalpha mAb. Whereas anti-CD154 mAb prolonged survival to day 21, administration of anti-TNFalpha and anti-CD154 mAb delayed rejection to day 32 and resulted in long-term (>80 days) survival of 40% of the heart allografts. These data implicate TNFalpha as an important mediator of early inflammatory events in allografts that undermine graft survival.
Collapse
Affiliation(s)
- Daisuke Ishii
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio,Department of Immunology, Cleveland Clinic, Cleveland, Ohio,Department of Urology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Austin D. Schenk
- Department of Immunology, Cleveland Clinic, Cleveland, Ohio,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Shiro Baba
- Department of Urology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Robert L. Fairchild
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, Ohio,Department of Immunology, Cleveland Clinic, Cleveland, Ohio,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
41
|
García-Criado FJ, Rodriguez-Barca P, García-Cenador MB, Rivas-Elena JV, Grande MT, Lopez-Marcos JF, Mourelle M, López-Novoa JM. Protective effect of new nitrosothiols on the early inflammatory response to kidney ischemia/reperfusion and transplantation in rats. J Interferon Cytokine Res 2009; 29:441-50. [PMID: 19514843 DOI: 10.1089/jir.2008.0100] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Renal ischemia/reperfusion (I/R) is characterized by severe inflammatory damage. We assessed the effect of administrating recently developed nitrosothiol compounds acting as nitric oxide (NO) donors on the production of cytokines and other markers of acute inflammatory reaction in an experimental model of warm (I/R), and in a model of cold ischemia and transplant in rats. Warm ischemia was achieved by ligation of left renal pedicle for 60 min, followed by contralateral nephrectomy. NO-donors LA-803, LA-807, LA-810 were administered i.v. (1.8 micromol/kg) during 30 min before reperfusion. Cold ischemia was achieved by preservating the kidney for 24 h in Euro Collins and grafting it in consanguineous Fisher 344/Ico rats. LA-803 was administered in the preservation fluid and in the recipient rat. Reperfusion time was 4 h in warm ischemia and 3 h in cold ischemia + transplantation. Administration of LA-803, LA-807 and, in a lower proportion, LA-810 prevented from the enhanced production of tumor necrosis factor (TNF), interferon-gamma (IFN-gamma), and interleukin-1beta (IL-1beta), the decrease in interleukin-6 (IL-6) and interleukin-10 (IL-10), the increase in tissue level of superoxide anion (SOA) and superoxide dismutase (SOD), and the increase in neutrophil infiltration induced by warm I/R. Treatment with LA-803 in animals with renal transplantation after cold ischemia was also associated with reduced plasma levels of TNF, IFN-gamma, and IL-1beta, increased plasma levels of IL-6 and IL-10, reduced renal levels of SOA and SOD, and reduced neutrophil infiltration. These data demonstrate that systemic administration of new NO-donors with nitrosothiol structure diminished inflammatory responses in a kidney subjected to warm I/R or cold ischemia and transplantation.
Collapse
Affiliation(s)
- F Javier García-Criado
- Departamento de Cirugía, Facultad de Medicina, Edificio Departamental, Campus Miguel de Unamuno, Universidad de Salamanca, Salamanca, Spain
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Kucuk A, Kabadere S, Tosun M, Koken T, Kinaci MK, Isikli B, Erkasap N. Protective effects of doxycycline in ischemia/reperfusion injury on kidney. J Physiol Biochem 2009; 65:183-91. [PMID: 19886397 DOI: 10.1007/bf03179069] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Renal ischemia and reperfusion injury is the major cause of acute renal failure and may also be involved in the development and progression of some forms of chronic kidney disease. The aim of this study was to evaluate whether doxycycline, a member of the tetracycline family of antibiotics, protects kidney tissue or not. 36 Sprague-Dawley rats (200-250 g) were used. The animals were divided into three groups: control, ischemia/reperfusion and ischemia/reperfusion+doxycycline group. Rats were subjected to renal ischemia by clamping the left pedicle for 1 h, and then reperfused for 1 h. The ischemia/reperfusion+doxycycline group were pretreated intraperitoneally with doxycycline suspension (10 mg/kg) 2 h before the induction of ischemia. Our results indicate that malondialdehyde, matrix-metalloproteinase-2, interleukin-2, interleukin-6, interleukin-10, interleukin 1-beta and tumor necrosis factor-alpha levels were significantly higher in the ischemia/reperfusion group than those in the control group. Doxycycline administration significantly decreased these parameters. Tissue inhibitor of metalloproteinases-1 levels also increased after ischemia/reperfusion and decreased with doxycycline pretreatment, but these changes were not significantly different. Glutathione levels significantly decreased after ischemia/reperfusion injury when compared with the control group and doxycycline pretreatment significantly increased glutathione levels when compared with the ischemia/reperfusion group. Apoptotic cells and p53 positive cells were significantly decreased in doxycycline treated group. These results suggest that doxycycline reduces renal oxidative injury and facilitates repair. Doxycycline may play a role in a renoprotective therapeutic regimen.
Collapse
Affiliation(s)
- A Kucuk
- Dumlupinar University, Medical Faculty, Physiology Department, Kutahya, Turkey.
| | | | | | | | | | | | | |
Collapse
|
43
|
Matthijsen RA, Derikx JPM, Kuipers D, van Dam RM, Dejong CHC, Buurman WA. Enterocyte shedding and epithelial lining repair following ischemia of the human small intestine attenuate inflammation. PLoS One 2009; 4:e7045. [PMID: 19753114 PMCID: PMC2737143 DOI: 10.1371/journal.pone.0007045] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Accepted: 07/28/2009] [Indexed: 12/13/2022] Open
Abstract
Background Recently, we observed that small-intestinal ischemia and reperfusion was found to entail a rapid loss of apoptotic and necrotic cells. This study was conducted to investigate whether the observed shedding of ischemically damaged epithelial cells affects IR induced inflammation in the human small gut. Methods and Findings Using a newly developed IR model of the human small intestine, the inflammatory response was studied on cellular, protein and mRNA level. Thirty patients were consecutively included. Part of the jejunum was subjected to 30 minutes of ischemia and variable reperfusion periods (mean reperfusion time 120 (±11) minutes). Ethical approval and informed consent were obtained. Increased plasma intestinal fatty acid binding protein (I-FABP) levels indicated loss in epithelial cell integrity in response to ischemia and reperfusion (p<0.001 vs healthy). HIF-1α gene expression doubled (p = 0.02) and C3 gene expression increased 4-fold (p = 0.01) over the course of IR. Gut barrier failure, assessed as LPS concentration in small bowel venous effluent blood, was not observed (p = 0.18). Additionally, mRNA expression of HO-1, IL-6, IL-8 did not alter. No increased expression of endothelial adhesion molecules, TNFα release, increased numbers of inflammatory cells (p = 0.71) or complement activation, assessed as activated C3 (p = 0.14), were detected in the reperfused tissue. Conclusions In the human small intestine, thirty minutes of ischemia followed by up to 4 hours of reperfusion, does not seem to lead to an explicit inflammatory response. This may be explained by a unique mechanism of shedding of damaged enterocytes, reported for the first time by our group.
Collapse
Affiliation(s)
- Robert A. Matthijsen
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Joep P. M. Derikx
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Dian Kuipers
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Ronald M. van Dam
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Cornelis H. C. Dejong
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Wim A. Buurman
- Department of Surgery, School for Nutrition & Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
- * E-mail:
| |
Collapse
|
44
|
Feng L, Ke N, Cheng F, Guo Y, Li S, Li Q, Li Y. The protective mechanism of ligustrazine against renal ischemia/reperfusion injury. J Surg Res 2009; 166:298-305. [PMID: 19592024 DOI: 10.1016/j.jss.2009.04.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Revised: 04/02/2009] [Accepted: 04/06/2009] [Indexed: 02/05/2023]
Abstract
BACKGROUND Ischemia/reperfusion (I/R) injury is unavoidable in renal transplantation, and represents an additional risk factor for the late renal allograft failure. Our study focused on the effects of ligustrazine on oxidative stress, apoptosis, neutrophils recruitment, the expression of proinflammatory mediators and adhesion molecules caused by renal I/R injury. MATERIALS AND METHODS Renal warm I/R was induced in male C57BL/6 mice by clamping the left renal artery and vein non-traumatically. Group I was sham-operated animals; group II, nontreated animals; and group III, ligustrazine-treated animals (80 mg/kg, i.p. 30 min before I/R). Mice were sacrificed 4 and 24h post reperfusion. The effects of ligustrazine on oxidative stress, neutrophils recruitment, proinflammatory mediators, and adhesion molecules caused by renal I/R injury were assayed. RESULTS Ligustrazine pretreatment attenuated dramatically the injuries in mice kidneys caused by warm I/R (histological scores of untreated versus treated, 4.2 ± 0.4 versus 0.9 ± 0.3; P<0.01). Administration of ligustrazine significantly reduced myeloperoxidase (MPO) activity by 38.6% and decreased malondialdehye (MDA) level by 19.2%, while superoxide dismutase (SOD) activity increased by 39.6% (P<0.01), suggesting an effective reduction of oxidative stress following ligustrazine treatment. Moreover, ligustrazine also inhibited cell apoptosis, abrogated neutrophils recruitment, and suppressed the over expression of TNF-α and ICAM-1. CONCLUSIONS In conclusion, ligustrazine protects murine kidney from warm ischemia/reperfusion injury, probably via reducing oxidative stress, inhibiting cell apoptosis, decreasing neutrophils infiltration, and suppressing the overexpression of TNF-α and ICAM-1 levels.
Collapse
Affiliation(s)
- Li Feng
- Key Laboratory of Transplant Engineering and Immunology of Health Ministry of China, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China.
| | | | | | | | | | | | | |
Collapse
|
45
|
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: 3.4] [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
|
46
|
Kadkhodaee M, Golab F, Zahmatkesh M, Ghaznavi R, Hedayati M, Arab HA, Ostad SN, Soleimani M. Effects of different periods of renal ischemia on liver as a remote organ. World J Gastroenterol 2009; 15:1113-8. [PMID: 19266605 PMCID: PMC2655196 DOI: 10.3748/wjg.15.1113] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [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 assess the hepatic changes after induction of different periods of renal ischemia.
METHODS: Rats were subjected to either sham operation or ischemia (30, 45 and 60 min) followed by 60 min reperfusion. Liver and renal functional indices were measured. Hepatic glutathione (GSH) and ferric reducing antioxidant power levels and the concentration of interleukin (IL)-10 and tumor necrosis factor (TNF-α) were evaluated. Portions of liver and kidney tissues were fixed for histological evaluation.
RESULTS: Forty-five minutes renal ischemia followed by 60 min reperfusion caused significant changes in liver structure and a significant reduction in renal function. These rats showed a significant decrease in liver GSH, as well as a significant increase in TNF-α and IL-10 concentrations. These results demonstrated that renal ischemia caused changes in liver histology, function, oxidative stress and inflammatory status, which led to a reduction in hepatic antioxidant capacity. With 30 min ischemia, the magnitude of these changes was less than those with 45 or 60 min ischemia.
CONCLUSION: A minimum of 45 min ischemia is needed to study the effects of renal injury on the liver as a remote organ.
Collapse
|
47
|
Jia RP, Xie JJ, Luo FY, Zhu JG. Ischemic preconditioning improves rat kidney allograft function after ischemia/reperfusion injury: the role of tumor necrosis factor-alpha. Transplant Proc 2009; 40:3316-20. [PMID: 19100380 DOI: 10.1016/j.transproceed.2008.06.113] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Revised: 05/03/2008] [Accepted: 06/23/2008] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the early protection of ischemic preconditioning (IPC) and its mechanisms in transplanted rat kidneys. MATERIALS AND METHODS Thirty-six male Sprague-Dawley (SD) rat donors and recipients were randomly divided into the following groups: sham-operated group (A; n = 6); untreated transplantation group (B; n = 6); and treatment group (C; n = 6). Group A was subjected to exploratory laparotomy. Group B received orthotopic transplantation. Group C underwent a 15-minute period of ischemia followed by a 10-minute reperfusion before orthotopic transplantation. We assessed the serum creatinine (SCr), blood urea nitrogen (BUN), and to evaluate the degree of kidney graft ischemia/reperfusion injury: tumor necrosis factor-alpha (TNF-alpha), IkappaB kinase-beta (IKK-beta), and nuclear factor-kappa B (NF-kappaB) P65 subunit mRNA expressions. RESULTS The levels of SCr and BUN in groups C and B were greater than in the sham-operated group (P < .01), but there was no significant difference between the C and B groups at 24 hours after transplantation (P > .05). The degree of renal graft tubular injury in group C was significantly less compared with group B (P < .01). TNF-alpha transcription levels at 24 hours after transplantation were significantly less compared with the non-IPC group (P < .01). However, no significant difference was observed in IKK-beta mRNA and P65 mRNA expressions between groups C and B (P > .05). CONCLUSIONS A 1-cycle schedule of preconditioning (15 min/10 min) attenuated renal graft ischemia/reperfusion injury in the early phase. IPC can improve rat kidney allograft function after ischemia/reperfusion injury. The inhibitory effects on TNF-alpha and on positive feedback signaling of TNF-alpha/NF-kappaB pathways may play important roles in renal graft protection in the early stage.
Collapse
Affiliation(s)
- R P Jia
- Department of Urology and Renal Transplantation, Nanjing First Hospital Affiliated to Nanjing Medical University, Nanjing, Pepole's Republic of China.
| | | | | | | |
Collapse
|
48
|
Shaw DM, Sutherland AM, Russell JA, Lichtenstein SV, Walley KR. Novel polymorphism of interleukin-18 associated with greater inflammation after cardiac surgery. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2009; 13:R9. [PMID: 19178691 PMCID: PMC2688121 DOI: 10.1186/cc7698] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2008] [Revised: 12/01/2008] [Accepted: 01/29/2009] [Indexed: 02/06/2023]
Abstract
Introduction Interleukin (IL)-18 is a key modulator of the cytokine response that leads to organ dysfunction and prolonged intensive care unit (ICU) stay after cardiopulmonary bypass surgery. We hypothesised that variation in the pro-inflammatory gene IL-18 is associated with adverse clinical outcome because of a more intense inflammatory response. Methods Haplotypes of the IL-18 gene were inferred from genotypes of 23 Coriell Registry subjects. Four haplotype tag single nucleotide polymorphisms (-607 C/A, -137 G/C, 8148 C/T and 9545 T/G) identified four major haplotype clades. These polymorphisms were genotyped in 658 Caucasian patients undergoing cardiopulmonary bypass surgery. Clinical phenotypes were collected by retrospective chart review. Results Patients homozygous for the T allele of the 9545 T/G polymorphism had an increased occurrence of prolonged ICU stay (6.8% for TT genotype versus 2.7% for GG or GT genotype; p = 0.015). Patients homozygous for the T allele also had increased occurrence of low systemic vascular resistance index (62%) compared with the GG and GT genotypes (53%; p = 0.045). Patients homozygous for the T allele had increased serum IL-18 concentrations 24 hours post-surgery (p = 0.018), increased pro-inflammatory tumour necrosis factor alpha concentrations (p = 0.014) and decreased anti-inflammatory serum IL-10 concentrations (p = 0.018) 24 hours post-surgery. Conclusions The TT genotype of the IL-18 9545 T/G polymorphism is associated with an increased occurrence of prolonged ICU stay post-surgery and greater post-surgical inflammation. These results may be explained by greater serum IL-18, leading to greater pro-versus anti-inflammatory cytokine expression.
Collapse
Affiliation(s)
- David M Shaw
- Critical Care Research Laboratories, Heart + Lung Institute, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | |
Collapse
|
49
|
Experimental Study on Early Protective Effect of Ischemic Preconditioning on Rat Kidney Graft. Transplant Proc 2009; 41:69-72. [DOI: 10.1016/j.transproceed.2008.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 10/27/2008] [Accepted: 11/05/2008] [Indexed: 02/04/2023]
|
50
|
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: 124] [Impact Index Per Article: 7.8] [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
|