51
|
Bischoff A, Bucher M, Gekle M, Sauvant C. Differential effect of COX1 and COX2 inhibitors on renal outcomes following ischemic acute kidney injury. Am J Nephrol 2014; 40:1-11. [PMID: 24943263 DOI: 10.1159/000363251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 04/23/2014] [Indexed: 01/11/2023]
Abstract
BACKGROUND/AIMS We have previously shown that 1 mg/kg indomethacin improves expression and functionality of renal organic anion transporters Oat1 and Oat3 after renal ischemia and furthermore improves renal outcome after ischemia. As we detected differential effects of COX1 or COX2 inhibitors on organic anion transport after ischemia and reperfusion in culture, we investigated the effect of the SC560 (COX1 inhibitor) and SC58125 (COX2 inhibitor) on expression of Oat1/3 and renal outcome after ischemic acute kidney injury (iAKI). METHODS iAKI was induced in rats by bilateral clamping of renal arteries for 45 min. SC560 or SC58125 (1 mg/kg each) were given intraperitoneally as soon as reperfusion started. Sham-treated animals served as controls. Oat1/3 were determined by qPCR and Western blot. Glomerular filtration rate (GFR), p-aminohippurate (PAH) clearance and PAH extraction ratio was determined. All parameters were detected 24 h after ischemia. Renal plasma flow was calculated. RESULTS In clamped animals SC560 (COX1 inhibitor) restored expression of Oat1/3, as well as renal perfusion. Additionally, SC560 substantially improved kidney function as measured by GFR. Application of the COX2 inhibitor SC58125 did not exert these beneficial effects. CONCLUSION Our study indicates that COX1 inhibitor SC560 applied after ischemia prevents ischemia-induced downregulation of Oat1/3 during reperfusion and has a substantial protective effect on kidney function. Whether and to what particular extent this apparent improvement of function is mechanistically due to beneficial effects on tubular function, renal perfusion or glomerular filtration will be the scope of future studies.
Collapse
Affiliation(s)
- Ariane Bischoff
- Klinik für Anästhesie und Operative Intensivmedizin, Universität Halle-Wittenberg, Halle, Germany
| | | | | | | |
Collapse
|
52
|
DiRocco DP, Bisi J, Roberts P, Strum J, Wong KK, Sharpless N, Humphreys BD. CDK4/6 inhibition induces epithelial cell cycle arrest and ameliorates acute kidney injury. Am J Physiol Renal Physiol 2013; 306:F379-88. [PMID: 24338822 DOI: 10.1152/ajprenal.00475.2013] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute kidney injury (AKI) is common and urgently requires new preventative therapies. Expression of a cyclin-dependent kinase (CDK) inhibitor transgene protects against AKI, suggesting that manipulating the tubular epithelial cell cycle may be a viable therapeutic strategy. Broad spectrum small molecule CDK inhibitors are protective in some kidney injury models, but these have toxicities and epithelial proliferation is eventually required for renal repair. Here, we tested a well-tolerated, novel and specific small molecule inhibitor of CDK4 and CDK6, PD 0332991, to investigate the effects of transient cell cycle inhibition on epithelial survival in vitro and kidney injury in vivo. We report that CDK4/6 inhibition induced G0/G1 cycle arrest in cultured human renal proximal tubule cells (hRPTC) at baseline and after injury. Induction of transient G0/G1 cycle arrest through CDK4/6 inhibition protected hRPTC from DNA damage and caspase 3/7 activation following exposure to the nephrotoxins cisplatin, etoposide, and antimycin A. In vivo, mice treated with PD 0332991 before ischemia-reperfusion injury (IRI) exhibited dramatically reduced epithelial progression through S phase 24 h after IRI. Despite reduced epithelial proliferation, PD 0332991 ameliorated kidney injury as reflected by improved serum creatinine and blood urea nitrogen levels 24 h after injury. Inflammatory markers and macrophage infiltration were significantly decreased in injured kidneys 3 days following IRI. These results indicate that induction of proximal tubule cell cycle arrest with specific CDK4/6 inhibitors, or "pharmacological quiescence," represents a novel strategy to prevent AKI.
Collapse
Affiliation(s)
- Derek P DiRocco
- Brigham and Women's Hospital, Harvard Institutes of Medicine, Rm. 550, 4 Blackfan Circle, Boston, MA 02115.
| | | | | | | | | | | | | |
Collapse
|
53
|
Hadjzadeh MAR, Rajaei Z, Keshavarzi Z, Shirazi MG, Toosi V. Effect of aqueous extract of Rheum ribes on cisplatin-induced nephrotoxicity in rat. J Pharm Bioallied Sci 2013; 5:309-13. [PMID: 24302839 PMCID: PMC3831744 DOI: 10.4103/0975-7406.120083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Revised: 05/13/2012] [Accepted: 08/07/2012] [Indexed: 11/10/2022] Open
Abstract
Objective: The purpose of the present study was to examine whether Rheum ribes extract prevents cisplatin-induced nephrotoxicity. Materials and Methods: The animals were divided into three groups: Group A considered as control group, group B were treated with cisplatin (3 mg/kg B.W. for 3 alternative days), and group C further to cisplatin received the aqueous extract of Rheum ribes (150 mg/rat). Results: Blood urea nitrogen (BUN) level increased in group B on days 14 and 42 compared to day 0 (P < 0.001); it was also increased in group B vs. group A on day 14 (P < 0.001). Rheum ribes extract decreased the serum BUN level on day 14 compared to group B (P < 0.001). Serum creatinine level in group B had a similar profile as serum BUN level but Rheum ribes had no effect on blood creatinine level. Serum cholesterol level was increased in group B on days 14 and 42 compared to day 0 (P < 0.001). Also, cholesterol level was significantly increased in group B when compared to group A on day 14 (P < 0.001). Rheum ribes decreased the blood cholesterol level on day 42 in comparison to group B (P < 0.001). Serum glucose level was increased in group B on days 14 and 42 vs. day 0 (P < 0.001). Also, glucose level was significantly increased in group B when compared to group A on day 42 (P < 0.001). Rheum ribes increased the serum glucose level on days 14 and 42 compared to day 0 (P < 0.05). Histology of kidneys exposed to cisplatin showed renal injury, but Rheum ribes had no effect on the kidney architecture. Conclusion: Cisplatin-induced nephrotoxicity was confirmed in our study. Although Rheum ribes had some effects on biochemical parameters; its effect on renal histology in injured kidney was insignificant.
Collapse
Affiliation(s)
- Mousa-Al-Reza Hadjzadeh
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran ; Neuroscience Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | | | | |
Collapse
|
54
|
Kaur S, Jain S, Saha A, Chawla D, Parmar VR, Basu S, Kaur J. Evaluation of glomerular and tubular renal function in neonates with birth asphyxia. ACTA ACUST UNITED AC 2013; 31:129-34. [DOI: 10.1179/146532811x12925735813922] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
55
|
Chang CF, Liu XM, Peyton KJ, Durante W. Heme oxygenase-1 counteracts contrast media-induced endothelial cell dysfunction. Biochem Pharmacol 2013; 87:303-11. [PMID: 24239896 DOI: 10.1016/j.bcp.2013.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/01/2013] [Accepted: 11/05/2013] [Indexed: 02/07/2023]
Abstract
Endothelial cell (EC) dysfunction is involved in the pathogenesis of contrast-induced acute kidney injury, which is a major adverse event following coronary angiography. In this study, we evaluated the effect of contrast media (CM) on human EC proliferation, migration, and inflammation, and determined if heme oxygenase-1 (HO-1) influences the biological actions of CM. We found that three distinct CM, including high-osmolar (diatrizoate), low-osmolar (iopamidol), and iso-osmolar (iodixanol), stimulated the expression of HO-1 protein and mRNA. The induction of HO-1 was associated with an increase in NF-E2-related factor-2 (Nrf2) activity and reactive oxygen species (ROS). CM also stimulated HO-1 promoter activity and this was prevented by mutating the antioxidant responsive element or by overexpressing dominant-negative Nrf2. In addition, the CM-mediated induction of HO-1 and activation of Nrf2 was abolished by acetylcysteine. Finally, CM inhibited the proliferation and migration of ECs and stimulated the expression of intercellular adhesion molecule-1 and the adhesion of monocytes on ECs. Inhibition or silencing of HO-1 exacerbated the anti-proliferative and inflammatory actions of CM but had no effect on the anti-migratory effect. Thus, induction of HO-1 via the ROS-Nrf2 pathway counteracts the anti-proliferative and inflammatory actions of CM. Therapeutic approaches targeting HO-1 may provide a novel approach in preventing CM-induced endothelial and organ dysfunction.
Collapse
Affiliation(s)
- Chao-Fu Chang
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Nephrology, Department of Medicine, Taipei City Hospital, Heping Fuyou Branch, Taipei, Taiwan
| | - Xiao-Ming Liu
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri-Columbia, Columbia, MO, USA
| | - Kelly J Peyton
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri-Columbia, Columbia, MO, USA
| | - William Durante
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri-Columbia, Columbia, MO, USA.
| |
Collapse
|
56
|
Mahadevappa R, Nielsen R, Christensen EI, Birn H. Megalin in acute kidney injury: foe and friend. Am J Physiol Renal Physiol 2013; 306:F147-54. [PMID: 24197071 DOI: 10.1152/ajprenal.00378.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The kidney proximal tubule is a key target in many forms of acute kidney injury (AKI). The multiligand receptor megalin is responsible for the normal proximal tubule uptake of filtered molecules, including nephrotoxins, cytokines, and markers of AKI. By mediating the uptake of nephrotoxins, megalin plays an essential role in the development of some types of AKI. However, megalin also mediates the tubular uptake of molecules implicated in the protection against AKI, and changes in megalin expression have been demonstrated in AKI in animal models. Thus, modulation of megalin expression in response to AKI may be an important part of the tubule cell adaption to cellular protection and regeneration and should be further investigated as a potential target of intervention. This review explores current evidence linking megalin expression and function to the development, diagnosis, and progression of AKI as well as renal protection against AKI.
Collapse
Affiliation(s)
- Ravikiran Mahadevappa
- Dept. of Biomedicine, Aarhus Univ., Wilhelm Meyers Allé 3, Bldg. 1234, Aarhus DK-8000, Denmark.
| | | | | | | |
Collapse
|
57
|
Brede C, Labhasetwar V. Applications of nanoparticles in the detection and treatment of kidney diseases. Adv Chronic Kidney Dis 2013; 20:454-65. [PMID: 24206598 DOI: 10.1053/j.ackd.2013.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/13/2013] [Accepted: 07/29/2013] [Indexed: 12/17/2022]
Abstract
Nanoparticles have emerged in the medical field as a technology well suited for the diagnosis and treatment of various disease states. They have been heralded as efficacious in terms of improved therapeutic efficacy and reduction of treatment side effects in some cases. Various nanomaterials have been developed that can be tagged with targeting moieties as well as with drug delivery and imaging capability or a combination of both as a theranostic agent. These nanomaterials have been investigated for treatment and detection of various pathological conditions. The emphasis of this review is to demonstrate current research and clinical applications for nanoparticles in the diagnosis and treatment of kidney diseases.
Collapse
|
58
|
Cao Z, Yu W, Li W, Cheng F, Xia Y, Rao T, Yao X, Zhang X, Larré S. Acute kidney injuries induced by various irrigation pressures in rat models of mild and severe hydronephrosis. Urology 2013; 82:1453.e9-16. [PMID: 24144540 DOI: 10.1016/j.urology.2013.08.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 07/22/2013] [Accepted: 08/09/2013] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To clarify whether tolerance to irrigation pressure could be modified over varying degrees of kidney obstruction during the endoscopic treatment of kidney stones in a rat model. METHODS A total of 126 rats were randomly allocated into 2 experimental groups and a control group. The experimental groups underwent a surgical procedure to induce mild (group M, n = 60) or severe (group S, n = 60) hydronephrosis. In each group, the rats were then randomly allocated into 4 subgroups (M0 to M3 and S0 to S3) of respectively 6, 18, 18, and 18 rats. Groups 0 to 3 were respectively perfused with 0 (no irrigation), 20, 60, and 100 mm Hg pressure fluid. The control group underwent no surgical procedures and was only perfused with 100 mm Hg pressure fluid. Acute kidney injuries were assessed by analyzing the kidney microstructure, tubular cell apoptosis, kidney injury molecule-1, and cysteine-rich 61 (Cyr61/CCN1) expression using immunohistochemistry. RESULTS No abnormalities were observed for the control group, groups 0, or 1. In group 2, abnormalities were observed only in the S group, whereas all kidneys in group 3 suffered acute kidneys injuries, along with occurrence of tubular cells necrosis, increased apoptosis, and increased expression of kidney injury molecule-1 and Cyr61. CONCLUSION Rats with severely obstructed kidneys were more likely to suffer acute kidney injuries than those with less obstructed kidneys when exposed to higher kidney irrigation pressures. This suggests that the pressure should be controlled and reduced when performing endourologic procedures in the context of kidney obstruction.
Collapse
Affiliation(s)
- Zhixiu Cao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | | | | | | | | | | | | | | | | |
Collapse
|
59
|
Ham A, Kim M, Kim JY, Brown KM, Fruttiger M, D'Agati VD, Lee HT. Selective deletion of the endothelial sphingosine-1-phosphate 1 receptor exacerbates kidney ischemia-reperfusion injury. Kidney Int 2013; 85:807-23. [PMID: 24025642 PMCID: PMC3952061 DOI: 10.1038/ki.2013.345] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 07/08/2013] [Accepted: 07/12/2013] [Indexed: 12/26/2022]
Abstract
The role for the endothelial sphingosine-1-phosphate 1 receptor (S1P1R) in acute kidney injury (AKI) remains unclear as germline endothelial S1P1R deletion is embryonically lethal. Here, we generated conditional endothelial S1P1R deficiency by crossing mice with floxed S1P1R with mice expressing a tamoxifen-inducible form of Cre recombinase under the transcriptional control of the platelet-derived growth factor-β gene. Mice with tamoxifen-induced deletion of endothelial S1P1R had increased renal tubular necrosis, inflammation, impaired vascular permeability as well as exacerbated renal tubular apoptosis after ischemic AKI compared to tamoxifen-treated wild type mice. Moreover, endothelial S1P1R deletion resulted in increased hepatic injury after ischemic AKI. As a potential mechanism for exacerbated renal injury, conditional endothelial S1P1R null mice had markedly reduced endothelial HSP27 expression compared to wild type mice. Cultured glomerular endothelial cells treated with a specific S1P1R antagonist (W146) for 3 days also showed reduced HSP27 expression compared to vehicle treated cells. Finally, mice treated with W146 for 3 days also showed reduced endothelial HSP27 expression as well as exacerbated renal and hepatic injury after ischemic AKI. Thus, our studies demonstrate a protective role for endothelial S1P1R against ischemic AKI most likely by regulating endothelial barrier integrity and endothelial HSP27 expression.
Collapse
Affiliation(s)
- Ahrom Ham
- Department of Anesthesiology, Anesthesiology Research Laboratories, College of Physicians and Surgeons of Columbia University, Columbia University, New York, New York, USA
| | - Mihwa Kim
- Department of Anesthesiology, Anesthesiology Research Laboratories, College of Physicians and Surgeons of Columbia University, Columbia University, New York, New York, USA
| | - Joo Yun Kim
- Department of Anesthesiology, Anesthesiology Research Laboratories, College of Physicians and Surgeons of Columbia University, Columbia University, New York, New York, USA
| | - Kevin M Brown
- Department of Anesthesiology, Anesthesiology Research Laboratories, College of Physicians and Surgeons of Columbia University, Columbia University, New York, New York, USA
| | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Vivette D D'Agati
- Department of Pathology, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - H Thomas Lee
- Department of Anesthesiology, Anesthesiology Research Laboratories, College of Physicians and Surgeons of Columbia University, Columbia University, New York, New York, USA
| |
Collapse
|
60
|
Preventive Effects of a Natural Anti-Inflammatory Agent, Astragaloside IV, on Ischemic Acute Kidney Injury in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:284025. [PMID: 23853656 PMCID: PMC3703719 DOI: 10.1155/2013/284025] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/12/2013] [Accepted: 05/12/2013] [Indexed: 02/07/2023]
Abstract
This study investigated the anti-inflammatory effects of astragaloside IV(AS-IV) on ischemia/reperfusion (IR) induced acute kidney injury (AKI) in rats. Experimental model of ischemic AKI was induced in rats by bilateral renal artery clamp for 45 min followed by reperfusion of 12 h and 24 h, respectively. AS-IV was orally administered once a day to rats at 10 and 20 mg·kg−1·d−1 for 7 days prior to ischemia. AS-IV pretreatment significantly decreased serum urea, creatinine, and cystatin C levels at 12 h and 24 h of reperfusion in AKI rats. AS-IV pretreatment also ameliorated tubular damage and suppressed the phosphorylation of p65 subunit of NF-κB in AKI rats. Moreover, NF-κB and MPO activity as well as serum and tissue levels of TNF-α, MCP-1, and ICAM-1 were elevated in AKI rats. All of these abnormalities were prevented by AS-IV. Furthermore, AS-IV downregulated the mRNA expression of NF-κB, TNF-α, MCP-1, and ICAM-1 in AKI rats. These results suggest that AS-IV might be developed as a novel therapeutic approach to prevent ischemic AKI through inhibition of NF-κB mediated inflammatory genes expression.
Collapse
|
61
|
Kim JI, Choi SH, Jung KJ, Lee E, Kim HY, Park KM. Protective role of methionine sulfoxide reductase A against ischemia/reperfusion injury in mouse kidney and its involvement in the regulation of trans-sulfuration pathway. Antioxid Redox Signal 2013; 18:2241-50. [PMID: 22657153 PMCID: PMC3638512 DOI: 10.1089/ars.2012.4598] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Methionine sulfoxide reductase A (MsrA) and methionine metabolism are associated with oxidative stress, a principal cause of ischemia/reperfusion (I/R) injury. Herein, we investigated the protective role of MsrA against kidney I/R injury and the involvement of MsrA in methionine metabolism and the trans-sulfuration pathway during I/R. RESULTS We found that MsrA gene-deleted mice (MsrA(-/-)) were more susceptible to kidney I/R injury than wild-type mice (MsrA(+/+)). Deletion of MsrA enhanced renal functional and morphological impairments, congestion, inflammatory responses, and oxidative stress under I/R conditions. Concentrations of homocysteine and H(2)S in the plasma of control MsrA(-/-) mice were significantly lower than those in control MsrA(+/+) mice. I/R reduced the levels of homocysteine and H(2)S in both MsrA(+/+) and MsrA(-/-) mice, and these reductions were significantly more profound in MsrA(-/-) than in MsrA(+/+) mice. I/R reduced the expression and activities of cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CSE), both of which are H(2)S-producing enzymes, in the kidneys. These reductions were more profound in the MsrA(-/-) mice than in the MsrA(+/+)mice. INNOVATION The data provided herein constitute the first in vivo evidence for the involvement of MsrA in regulating methionine metabolism and the trans-sulfuration pathway under normal and I/R conditions. CONCLUSION Our data demonstrate that MsrA protects the kidney against I/R injury, and that this protection is associated with reduced oxidative stress and inflammatory responses. The data indicate that MsrA regulates H(2)S production during I/R by modulating the expression and activity of the CBS and CSE enzymes.
Collapse
Affiliation(s)
- Jee In Kim
- Department of Anatomy, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | | | | | | | | | | |
Collapse
|
62
|
Parida S, Badhe AS. Cardiac surgery-associated acute kidney injury. J Anesth 2013; 27:433-446. [PMID: 23179740 DOI: 10.1007/s00540-012-1523-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 11/04/2012] [Indexed: 12/22/2022]
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
|
63
|
Zhen-Qiang F, Bing-Wei Y, Yong-Liang L, Xiang-Wei W, Shan-Hong Y, Yuan-Ning Z, Wei-Sheng J, Wei C, Ye G. Localized expression of human BMP-7 by BM-MSCs enhances renal repair in an in vivo model of ischemia-reperfusion injury. Genes Cells 2013; 17:53-64. [PMID: 22212474 DOI: 10.1111/j.1365-2443.2011.01572.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Ischemia and subsequent reperfusion (I/R) damage kidney tubular cells and consequently impair renal function. Rabbit bone marrow mesenchymal stem cells (BM-MSCs) expressing human bone morphogenic protein-7 (hBMP-7) regenerated tubular cells and improved renal function in a kidney I/R model. Rabbits were injected immediately after I/R with one of the following: (i) hBMP-7-transduced BM-MSCs (BM-MSCshBMP-7); (ii) enhanced green fluorescent protein-transduced BM-MSCs (BM-MSCsEGFP); or (iii) PBS. The activity of superoxide dismutase (SOD) was higher, and the amount of malondialdehyde (MDA) was lower in the BM-MSCshBMP-7 group than in the BM-MSCsEGFP group. Both the BM-MSCshBMP-7 group and the BM-MSCsEGFP group had higher SOD activity and lower amounts of MDA than the PBS group. Bcl-2- and Bcl-2-associated X protein levels, and other variables, indicated the regeneration of the kidney in both experimental groups. However, the BM-MSCs (hBMP-7) group showed higher activity than the BM-MSCsEGFP group, indicating that the combined strategy of BM-MSC transplantation with hBMP-7 gene therapy could be a useful approach for the treatment of renal IRI.
Collapse
Affiliation(s)
- Fang Zhen-Qiang
- Department of Urology, Center of Nephrology, The Second affiliated Hospital of the Third Military Medical University, Chongqing 400037, China
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Caine ST, Mclaughlin KA. Regeneration of functional pronephric proximal tubules after partial nephrectomy in Xenopus laevis. Dev Dyn 2013; 242:219-29. [PMID: 23233460 DOI: 10.1002/dvdy.23916] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Revised: 11/02/2012] [Accepted: 11/26/2012] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND While the renal system is critical for maintaining homeostatic equilibrium within the body, it is also susceptible to various kinds of damage. Tubule dysfunction in particular contributes to acute renal injury and chronic kidney disease in millions of patients worldwide. Because current treatments are highly invasive and often unavailable, gaining a better understanding of the regenerative capacity of renal structures is vital. Although the effects of various types of acute damage have been previously studied, the ability of the excretory system to repair itself after dramatic tissue loss due to mechanical damage is less well characterized. RESULTS A novel unilateral nephrectomy technique was developed to excise pronephric proximal tubules from Xenopus laevis tadpoles to study tubule repair after injury. Immunohistochemical detection of protein expression and renal uptake assays demonstrated that X. laevis larvae have the capacity to regenerate functional proximal tubules following resection. CONCLUSIONS We have validated the renal identity of the restored tubules and demonstrated their ability to functional normally providing the first evidence of regeneration of renal tissue in an amphibian system. Importantly, this tubule restoration occurs by means of a process involving an early apoptotic event and the biphasic expression of the matrix metalloproteinase, Xmmp-9.
Collapse
|
65
|
Chang CF, Lu TM, Yang WC, Lin SJ, Lin CC, Chung MY. Gene polymorphisms of interleukin-10 and tumor necrosis factor-α are associated with contrast-induced nephropathy. Am J Nephrol 2013; 37:110-7. [PMID: 23363891 DOI: 10.1159/000346528] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 12/17/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Contrast-induced nephropathy (CIN) is the third most common cause of hospital-acquired acute renal failure. However, the pathogenesis of CIN remains unclear. This study evaluated the role of anti-inflammatory cytokine interleukin-10 (IL-10) and pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) gene polymorphisms as CIN susceptibility markers after percutaneous coronary intervention (PCI). METHODS Four IL-10 tag SNPs (rs1554286, rs3021094, rs3790622, rs1800896) and three TNF-α tag SNPs (rs1799964, rs1800630, rs1800629) were analyzed by MALDI-TOF mass spectrometry in 53 CIN patients and 455 control subjects. Serum IL-10 and TNF-α were detected using ELISA. RESULTS When compared to controls, the CIN patients showed increased frequencies of CC (rs1554286) and AG+GG (rs1800896) genotypes in IL-10 and GA+AA (rs1800629) genotype in TNF-α (OR = 2.24 (1.13-4.44), p = 0.018; OR = 2.61 (1.30-5.26), p = 0.005, and OR = 2.11 (1.08-4.09), p = 0.025, respectively). Baseline serum IL-10 levels in CIN patients were significantly lower (1.02 ± 1.14 vs. 2.78 ± 4.73 pg/ml, p = 0.008). Patients with CIN had a higher rate of decline in renal function than those without CIN (0.89 ± 1.67 vs. 0.30 ± 0.95 ml/min/1.73 m(2) per month, p = 0.002). Significantly higher rates of decline in creatinine clearance were noted in patients with TNF-α (rs1800629) GA+AA than GG genotype (0.88 ± 1.83 vs. 0.36 ± 0.70, p = 0.03), and with IL-10 (rs1800896) AG+GG than AA genotype (1.28 ± 2.14 vs. 0.33 ± 0.90, p < 0.001). CONCLUSIONS Gene polymorphisms of IL-10 and TNF-α are associated with CIN risk and long-term renal outcome after PCI. More prospective studies are needed to confirm our results.
Collapse
Affiliation(s)
- Chao-Fu Chang
- School of Medicine, Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
66
|
Ardalan MR, Estakhri R, Hajipour B, Ansarin K, Asl NA, Nasirizade MR, Azar AN, Ghorbanihaghjou A, Vatankhah AM, Esmaili HA. Erythropoietin ameliorates oxidative stress and tissue injury following renal ischemia/reperfusion in rat kidney and lung. Med Princ Pract 2013; 22:70-4. [PMID: 23006583 PMCID: PMC5586709 DOI: 10.1159/000340060] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2011] [Accepted: 06/10/2012] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE To study the effect of erythropoietin (EPO) treatment on renal and lung injury following renal ischemia/reperfusion (I/R). MATERIALS AND METHODS Thirty male Wistar rats were assigned to three groups of 10 rats each. The first group was sham-operated, the second was subjected to renal I/R (30 min of ischemia followed by 24 h of reperfusion). The third group was subjected to renal I/R and treated with EPO in two doses: the first dose 1 h prior to ischemia (1,000 U/kg) and the second dose 6 h after ischemia (1,000 U/kg). RESULTS The renal and lung tissue injury index, tissue serum blood urea nitrogen and creatinine (Cr) were higher in the renal I/R group compared to the renal I/R + EPO group; the difference was statistically significant (p < 0.05). Kidney and lung tissue glutathione peroxidase and superoxide dismutase levels were higher in the renal I/R + EPO group than the renal I/R group; the difference was also statistically significant (p < 0.05). CONCLUSION The data showed that EPO pretreatment could be effective in reducing renal and lung injury following renal I/R and could improve the cellular antioxidant defense system. Hence EPO pretreatment may be effective for attenuating renal and lung injury after renal I/R-induced injury during surgical procedures, hypotension, renal transplantation and other conditions inducing renal I/R.
Collapse
Affiliation(s)
| | | | - Babak Hajipour
- Urmia University of Medical Sciences, Urmia, Iran
- *Babak Hajipou, Young Researchers Club, Tabriz Branch, Islamic Azad University, Postbox 51385-3633, Tabriz (Iran), Tel. +98 914 107 5936, E-Mail
| | - Khalil Ansarin
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Urmia, Iran
| | | | - Mohammad Reza Nasirizade
- Department of Physiology, Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Urmia, Iran
| | - Alireza Nour Azar
- Department of Physiology, Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Urmia, Iran
| | | | | | | |
Collapse
|
67
|
Transcriptome analysis of renal ischemia/reperfusion injury and its modulation by ischemic pre-conditioning or hemin treatment. PLoS One 2012; 7:e49569. [PMID: 23166714 PMCID: PMC3498198 DOI: 10.1371/journal.pone.0049569] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 10/10/2012] [Indexed: 01/06/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) is a leading cause of acute renal failure. The definition of the molecular mechanisms involved in renal IRI and counter protection promoted by ischemic pre-conditioning (IPC) or Hemin treatment is an important milestone that needs to be accomplished in this research area. We examined, through an oligonucleotide microarray protocol, the renal differential transcriptome profiles of mice submitted to IRI, IPC and Hemin treatment. After identifying the profiles of differentially expressed genes observed for each comparison, we carried out functional enrichment analysis to reveal transcripts putatively involved in potential relevant biological processes and signaling pathways. The most relevant processes found in these comparisons were stress, apoptosis, cell differentiation, angiogenesis, focal adhesion, ECM-receptor interaction, ion transport, angiogenesis, mitosis and cell cycle, inflammatory response, olfactory transduction and regulation of actin cytoskeleton. In addition, the most important overrepresented pathways were MAPK, ErbB, JAK/STAT, Toll and Nod like receptors, Angiotensin II, Arachidonic acid metabolism, Wnt and coagulation cascade. Also, new insights were gained about the underlying protection mechanisms against renal IRI promoted by IPC and Hemin treatment. Venn diagram analysis allowed us to uncover common and exclusively differentially expressed genes between these two protective maneuvers, underscoring potential common and exclusive biological functions regulated in each case. In summary, IPC exclusively regulated the expression of genes belonging to stress, protein modification and apoptosis, highlighting the role of IPC in controlling exacerbated stress response. Treatment with the Hmox1 inducer Hemin, in turn, exclusively regulated the expression of genes associated with cell differentiation, metabolic pathways, cell cycle, mitosis, development, regulation of actin cytoskeleton and arachidonic acid metabolism, suggesting a pleiotropic effect for Hemin. These findings improve the biological understanding of how the kidney behaves after IRI. They also illustrate some possible underlying molecular mechanisms involved in kidney protection observed with IPC or Hemin treatment maneuvers.
Collapse
|
68
|
|
69
|
Cantaluppi V, Biancone L, Quercia A, Deregibus MC, Segoloni G, Camussi G. Rationale of mesenchymal stem cell therapy in kidney injury. Am J Kidney Dis 2012; 61:300-9. [PMID: 22938846 DOI: 10.1053/j.ajkd.2012.05.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 05/23/2012] [Indexed: 01/06/2023]
Abstract
Numerous preclinical and clinical studies suggest that mesenchymal stem cells, also known as multipotent mesenchymal stromal cells (MSCs), may improve pathologic conditions involving different organs. These beneficial effects initially were ascribed to the differentiation of MSCs into organ parenchymal cells. However, at least in the kidney, this is a very rare event and the kidney-protective effects of MSCs have been attributed mainly to paracrine mechanisms. MSCs release a number of trophic, anti-inflammatory, and immune-modulatory factors that may limit kidney injury and favor recovery. In this article, we provide an overview of the biologic activities of MSCs that may be relevant for the treatment of kidney injury in the context of a case vignette concerning a patient at high immunologic risk who underwent a second kidney transplantation followed by the development of ischemia-reperfusion injury and acute allograft rejection. We discuss the possible beneficial effect of MSC treatment in the light of preclinical and clinical data supporting the regenerative and immunomodulatory potential of MSCs.
Collapse
Affiliation(s)
- Vincenzo Cantaluppi
- Nephrology, Dialysis and Renal Transplantation Unit, Centre for Experimental Medical Research (CeRMS) and Department of Internal Medicine, University of Torino, Torino, Italy
| | | | | | | | | | | |
Collapse
|
70
|
Reel B, Guzeloglu M, Bagriyanik A, Atmaca S, Aykut K, Albayrak G, Hazan E. The effects of PPAR-γ agonist pioglitazone on renal ischemia/reperfusion injury in rats. J Surg Res 2012; 182:176-84. [PMID: 22981741 DOI: 10.1016/j.jss.2012.08.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 07/20/2012] [Accepted: 08/09/2012] [Indexed: 01/19/2023]
Abstract
BACKGROUND Acute renal failure due to renal ischemia/reperfusion (IR) injury is a significant clinical problem in cardiovascular surgery. Reactive oxygen species and inflammation play essential roles in the pathophysiology of IR injury. Matrix metalloproteinases (MMPs) are enzymes that play important roles in inflammation and mediate extracellular matrix degradation. It is known that peroxisome proliferator-activated receptor-γ agonists have antiinflammatory and antioxidant effects. In the present study, we aimed to investigate the effects of pioglitazone, a synthetic peroxisome proliferator-activated receptor-γ agonist, on MMPs and oxidative stress in a renal IR injury model in rats. MATERIALS AND METHODS Male Wistar albino rats were divided into three groups: control (n = 7), placebo (n = 7; saline/p.o.), and pioglitazone (n = 7; 5 mg/kg/day/p.o.). In the control group, a right nephrectomy was conducted without left renal IR injury. In the placebo and pioglitazone groups, pretreatments were started 3 d before operation. In both groups, left renal pedicles were clamped for 60 min and then reperfused for 60 min. Paraffinized renal sections were evaluated histopathologically. Furthermore, expressions of MMP-2, MMP-9, tissue inhibitor of metalloproteinase (TIMP)-2, superoxide dismutase 1 (SOD1), and p47-phox/p67-phox subunits of NADPH oxidase were determined by immunostaining and scoring. RESULTS In the placebo group, renal IR injury induced diffuse tubular necrosis and intense acute inflammation, but pioglitazone inhibited these effects. MMP-2, MMP-9, and TIMP-2 expression increased in the placebo group. However, while MMP-2 and -9 expression decreased, TIMP-2 expression did not change in the pioglitazone group. p47-phox/p67-phox expression increased in the placebo group, but SOD1 expression did not change. Pioglitazone diminished p47-phox/p67-phox expression, whereas it enhanced SOD1 expression. CONCLUSION Our results suggest that pioglitazone might be helpful to reduce renal IR injury because of its antiinflammatory and antioxidant effects.
Collapse
Affiliation(s)
- Buket Reel
- Faculty of Pharmacy, Department of Pharmacology, Ege University, Izmir, Turkey
| | | | | | | | | | | | | |
Collapse
|
71
|
Extracellular ATP attenuates ischemia-induced caspase-3 cleavage in human endothelial cells. Biochem Biophys Res Commun 2012; 425:230-6. [PMID: 22828512 DOI: 10.1016/j.bbrc.2012.07.073] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 07/15/2012] [Indexed: 01/31/2023]
Abstract
BACKGROUND Apoptotic death of endothelial cells (EC) plays a crucial role for the development of ischemic injury. In the present study we investigated the impact of extracellular Adenosine-5'-triphosphate (ATP), either released from cells or exogenously added, on ischemia-induced apoptosis of human EC. METHODS AND RESULTS To simulate ischemic conditions, cultured human umbilical vein endothelial cells (HUVEC) were exposed to 2 h of hypoxia (Po(2)<4mm Hg) in serum-free medium. Ischemia led to a 1.7-fold (+/-0.4; P<0.05) increase in EC apoptosis compared to normoxic controls as assessed by immunoblotting and immunocytochemistry of cleaved caspase-3. Ischemia-induced apoptosis was accompanied by a 2.3-fold (+/-0.5; P<0.05) increase of extracellular ATP detected by using a luciferin/luciferase assay. Addition of the soluble ecto-ATPase apyrase, enhancing ATP degradation, increased ischemia-induced caspase-3 cleavage. Correspondingly, inhibition of ATP breakdown by addition of the selective ecto-ATPase inhibitor ARL67156 significantly reduced ischemia-induced apoptosis. Extracellular ATP acts on membrane-bound P2Y- and P2X-receptors to induce intracellular signaling. Both, ATP and the P2Y-receptor agonist UTP significantly reduced ischemia-induced apoptosis in an equipotent manner, whereas the P2X-receptor agonist αβ-me-ATP did not alter caspase-3 cleavage. The anti-apoptotic effects of ARL67156 and UTP were abrogated when P2-receptors were blocked by Suramin or PPADS. Furthermore, extracellular ATP led to an activation of MEK/ERK- and PI3K/Akt-signaling pathways. Accordingly, inhibition of MEK/ERK-signaling by UO126 or inhibition of PI3K/Akt-signaling by LY294002 abolished the anti-apoptotic effects of ATP. CONCLUSION The data of the present study indicate that extracellular ATP counteracts ischemia-induced apoptosis of human EC by activating a P2Y-receptor-mediated signaling reducing caspase-3 cleavage.
Collapse
|
72
|
Eadon MT, Hack BK, Xu C, Ko B, Toback FG, Cunningham PN. Endotoxemia alters tight junction gene and protein expression in the kidney. Am J Physiol Renal Physiol 2012; 303:F821-30. [PMID: 22791339 DOI: 10.1152/ajprenal.00023.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Intact tight junctional (TJ) proteins are required for tubular ion transport and waste excretion. Disruption of TJs may contribute to a decreased glomerular filtration rate in acute kidney injury (AKI) via tubular backleak. The effect of LPS-mediated AKI on murine TJs has not been studied extensively. We hypothesized LPS endotoxin administration to mice would disrupt tubular TJ proteins including zonula occludens-1 (ZO-1), occludin, and claudins. ZO-1 and occludin immunofluorescence 24 h post-LPS revealed a marked change in localization from the usual circumferential fencework pattern to one with substantial fragmentation. Renal ZO-1 expression was significantly reduced 24 h after LPS (decrease of 56.1 ± 7.4%, P < 0.001), with subsequent recovery. ZO-1 mRNA expression was increased 24 h post-LPS (4.34 ± 0.87-fold, P = 0.0019), suggesting disruption of ZO-1 protein is not mediated by transcriptional regulation, but rather by degradation or changes in translation. Similarly, claudin-4 protein expression was decreased despite elevated mRNA. LPS administration resulted in dephosphorylation of occludin and fragmented tubular redistribution. Protein expression of claudin-1, and -3 was increased after LPS. ZO-1, occludin, and claudin-1, -3, and -4 gene expression were increased 48 h after LPS, suggesting a renal response to strengthen TJs following injury. Interestingly, reduced mRNA expression was found only for claudin-8. This study provides further support that LPS-induced AKI is associated with structural injury and is not merely due to hemodynamic changes.
Collapse
Affiliation(s)
- Michael T Eadon
- Section of Nephrology, The University of Chicago, Illinois, USA.
| | | | | | | | | | | |
Collapse
|
73
|
Lésions d’ischémie-reperfusion rénale. MEDECINE INTENSIVE REANIMATION 2012. [DOI: 10.1007/s13546-012-0475-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
74
|
Girardi ACC, Di Sole F. Deciphering the mechanisms of the Na+/H+ exchanger-3 regulation in organ dysfunction. Am J Physiol Cell Physiol 2012; 302:C1569-87. [DOI: 10.1152/ajpcell.00017.2012] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Na+/H+ exchanger-3 (NHE3) belongs to the mammalian NHE protein family and catalyzes the electro-neutral exchange of extracellular sodium for intracellular proton across cellular membranes. Its transport function is of essential importance for the maintenance of the body's salt and water homeostasis as well as acid-base balance. Indeed, NHE3 activity is finely regulated by a variety of stimuli, both acutely and chronically, and its transport function is fundamental for a multiplicity of severe and world-wide infection-pathological conditions. This review aims to provide a concise overview of NHE3 physiology and discusses the role of NHE3 in clinical conditions of prominent importance, specifically in hypertension, diabetic nephropathy, heart failure, acute kidney injury, and diarrhea. Study of NHE3 function in models of these diseases has contributed to the deciphering of mechanisms that control the delicate ion balance disrupted in these disorders. The majority of the findings indicate that NHE3 transport function is activated before the onset of hypertension and inhibited thereafter; NHE3 transport function is also upregulated in diabetic nephropathy and heart failure, while it is reported to be downregulated in acute kidney injury and in diarrhea. The molecular mechanisms activated during these pathological conditions to regulate NHE3 transport function are examined with the aim of linking NHE3 dysfunction to the analyzed clinical disorders.
Collapse
Affiliation(s)
| | - Francesca Di Sole
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; and
- Center of Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
75
|
Microvesicles derived from endothelial progenitor cells protect the kidney from ischemia-reperfusion injury by microRNA-dependent reprogramming of resident renal cells. Kidney Int 2012; 82:412-27. [PMID: 22495296 DOI: 10.1038/ki.2012.105] [Citation(s) in RCA: 418] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Endothelial progenitor cells are known to reverse acute kidney injury by paracrine mechanisms. We previously found that microvesicles released from these progenitor cells activate an angiogenic program in endothelial cells by horizontal mRNA transfer. Here, we tested whether these microvesicles prevent acute kidney injury in a rat model of ischemia-reperfusion injury. The RNA content of microvesicles was enriched in microRNAs (miRNAs) that modulate proliferation, angiogenesis, and apoptosis. After intravenous injection following ischemia-reperfusion, the microvesicles were localized within peritubular capillaries and tubular cells. This conferred functional and morphologic protection from acute kidney injury by enhanced tubular cell proliferation, reduced apoptosis, and leukocyte infiltration. Microvesicles also protected against progression of chronic kidney damage by inhibiting capillary rarefaction, glomerulosclerosis, and tubulointerstitial fibrosis. The renoprotective effect of microvesicles was lost after treatment with RNase, nonspecific miRNA depletion of microvesicles by Dicer knock-down in the progenitor cells, or depletion of pro-angiogenic miR-126 and miR-296 by transfection with specific miR-antagomirs. Thus, microvesicles derived from endothelial progenitor cells protect the kidney from ischemic acute injury by delivering their RNA content, the miRNA cargo of which contributes to reprogramming hypoxic resident renal cells to a regenerative program.
Collapse
|
76
|
Abstract
PURPOSE OF REVIEW Acute kidney injury (AKI) is a major clinical problem without effective therapy. Development of AKI among hospitalized patients drastically increases mortality and morbidity. With increases in complex surgical procedures together with a growing elderly population, the incidence of AKI is rising. Renal adenosine receptor manipulation may have great therapeutic potential in mitigating AKI. In this review, we discuss renal adenosine receptor biology and potential clinical therapies for AKI. RECENT FINDINGS The four adenosine receptor subtypes (A(1)AR, A(2A)AR, A(2B)AR, and A(3)AR) have diverse effects on the kidney. The pathophysiology of AKI may dictate the specific adenosine receptor subtype activation needed to produce renal protection. The A(1)AR activation in renal tubules and endothelial cells produces beneficial effects against ischemia and reperfusion injury by modulating metabolic demand, decreasing necrosis, apoptosis, and inflammation. The A(2A)AR protects against AKI by modulating leukocyte-mediated renal and systemic inflammation, whereas the A(2B)AR activation protects by direct activation of renal parenchymal adenosine receptors. In contrast, the A(1)AR antagonism may play a protective role in nephrotoxic AKI and radiocontrast induced nephropathy by reversing vascular constriction and inducing naturesis and diuresis. Furthermore, as the A(3)AR activation exacerbates apoptosis and tissue damage due to renal ischemia and reperfusion, selective A(3)AR antagonism may hold promise to attenuate renal ischemia and reperfusion injury. Finally, renal A(1)AR activation also protects against renal endothelial dysfunction caused by hepatic ischemia and reperfusion injury. SUMMARY Despite the current lack of therapies for the treatment and prevention of AKI, recent research suggests that modulation of renal adenosine receptors holds promise in treating AKI and extrarenal injury.
Collapse
|
77
|
Lee PY, Chien Y, Chiou GY, Lin CH, Chiou CH, Tarng DC. Induced pluripotent stem cells without c-Myc attenuate acute kidney injury via downregulating the signaling of oxidative stress and inflammation in ischemia-reperfusion rats. Cell Transplant 2012; 21:2569-85. [PMID: 22507855 DOI: 10.3727/096368912x636902] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Induced pluripotent stem (iPS) cells have potential for multilineage differentiation and provide a resource for stem cell-based treatment. However, the therapeutic effect of iPS cells on acute kidney injury (AKI) remains uncertain. Given that the oncogene c-Myc may contribute to tumorigenesis by causing genomic instability, herein we evaluated the therapeutic effect of iPS cells without exogenously introduced c-Myc on ischemia-reperfusion (I/R)-induced AKI. As compared with phosphate-buffered saline (PBS)-treated group, administration of iPS cells via intrarenal arterial route into kidneys improved the renal function and attenuated tubular injury score at 48 h after ischemia particularly at the dose of 5 × 10(5) iPS cells. However, a larger number of iPS cells (5 × 10(7) per rat) diminished the therapeutic effects for AKI and profoundly reduced renal perfusion detected by laser Doppler imaging in the reperfusion phase. In addition, the green fluorescence protein-positive iPS cells mobilized to the peritubular area at 48 h following ischemia, accompanied by a significant reduction in infiltration of macrophages and apoptosis of tubular cells, and a remarkable enhancement in endogenous tubular cell proliferation. Importantly, transplantation of iPS cells reduced the expression of oxidative substances, proinflammatory cytokines, and apoptotic factors in I/R kidney tissues and eventually improved survival in rats of ischemic AKI. Six months after transplantation in I/R rats, engrafted iPS cells did not result in tumor formation in kidney and other organs. In summary, considering the antioxidant, anti-inflammatory, and antiapoptotic properties of iPS cells without c-Myc, transplantation of such cells may be a treatment option for ischemic AKI.
Collapse
Affiliation(s)
- Pei-Ying Lee
- Department and Institute of Physiology, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
78
|
A critical evaluation of in vitro cell culture models for high-throughput drug screening and toxicity. Pharmacol Ther 2012; 134:82-106. [DOI: 10.1016/j.pharmthera.2012.01.001] [Citation(s) in RCA: 276] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 12/22/2011] [Indexed: 01/10/2023]
|
79
|
Abstract
Acute kidney injury (AKI) is the leading cause of nephrology consultation and is associated with high mortality rates. The primary causes of AKI include ischemia, hypoxia, or nephrotoxicity. An underlying feature is a rapid decline in glomerular filtration rate (GFR) usually associated with decreases in renal blood flow. Inflammation represents an important additional component of AKI leading to the extension phase of injury, which may be associated with insensitivity to vasodilator therapy. It is suggested that targeting the extension phase represents an area potential of treatment with the greatest possible impact. The underlying basis of renal injury appears to be impaired energetics of the highly metabolically active nephron segments (i.e., proximal tubules and thick ascending limb) in the renal outer medulla, which can trigger conversion from transient hypoxia to intrinsic renal failure. Injury to kidney cells can be lethal or sublethal. Sublethal injury represents an important component in AKI, as it may profoundly influence GFR and renal blood flow. The nature of the recovery response is mediated by the degree to which sublethal cells can restore normal function and promote regeneration. The successful recovery from AKI depends on the degree to which these repair processes ensue and these may be compromised in elderly or chronic kidney disease (CKD) patients. Recent data suggest that AKI represents a potential link to CKD in surviving patients. Finally, earlier diagnosis of AKI represents an important area in treating patients with AKI that has spawned increased awareness of the potential that biomarkers of AKI may play in the future.
Collapse
Affiliation(s)
- David P Basile
- Department of Cellular & Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | | | | |
Collapse
|
80
|
Mariscalco G, Nicolini F, Scannapieco A, Gherli R, Serraino F, Dominici C, Renzulli A, Gherli T, Sala A, Beghi C. Acute kidney injury after composite valve-graft replacement for ascending aorta aneurysms. Heart Vessels 2012; 28:229-36. [PMID: 22411492 DOI: 10.1007/s00380-012-0239-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 02/03/2012] [Indexed: 10/28/2022]
Abstract
Acute kidney injury (AKI) following cardiac surgery is a continuing source of morbidity and mortality. Although several studies have attempted to determine its etiology and prophylactic measures, limited data exist after thoracic aortic surgery. The aim of this study was to evaluate the incidence and predictors of AKI in patients undergoing aortic root replacement (ARR) with valve conduit for ascending aorta aneurysms. A multi-center observational study of 414 patients undergoing ARR with a valve conduit was conducted, focusing on clinical outcome and AKI defined by consensus RIFLE (risk, injury, failure, loss of function, end-stage renal disease) criteria. Mean age was 62.5 years (range: 21-82 years) with 327 males (79%). Emergent operations were performed in 5% of the cases, while concomitant surgical procedures were performed in 24.9%. Postoperative AKI (all RIFLE classes) occurred in 69 (16.7%) patients, while eight (1.9%) required dialysis. Independent AKI predictors were packed red blood cells (pRBCs) >4 units (OR 2.28; 95% CI 1.20-4.30), cardiopulmonary bypass (CPB) time longer than 180 min (OR, 2.08; 95% CI, 1.16-3.73), and concomitant surgical procedures (OR, 1.85; 95% CI, 1.04-3.29). The severity of RIFLE class was associated with longer ICU stay, hospitalization, and higher hospital mortality (p < 0.001 for each variable). AKI after ARR operations with valve conduit for ascending aorta aneurysms increases utilization of health resources and is associated with adverse events. Concomitant surgical procedures, prolonged CPB-time, and pRBCs >4 units as independent AKI predictors merit further researches enhancing possible preventive strategies.
Collapse
Affiliation(s)
- Giovanni Mariscalco
- Department of Surgical and Morphological Sciences, Cardiac Surgery Unit, Varese University Hospital, University of Insubria, Varese, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Larman BW, Karolak MJ, Lindner V, Oxburgh L. Distinct bone morphogenetic proteins activate indistinguishable transcriptional responses in nephron epithelia including Notch target genes. Cell Signal 2012; 24:257-64. [PMID: 21945409 PMCID: PMC3205934 DOI: 10.1016/j.cellsig.2011.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 08/22/2011] [Accepted: 09/06/2011] [Indexed: 10/17/2022]
Abstract
Endogenous Bone Morphogenetic Protein (BMP) signaling plays a significant role in the kidney's recovery from acute injury and exogenous administration of BMP7 has therapeutic potential in numerous rodent models of renal injury and disease. However, in the healthy kidney endogenous BMP7 ligand is vigorously counteracted by extracellular antagonists such as USAG1 and CHRDL1. Little is known about the degree of BMP signaling and the ligands driving it in the healthy adult kidney. In this study we characterize basal BMP signaling in the healthy tubular nephron, and show that BMP2 is expressed in proximal nephron epithelial cells. Comparative gene profiling of proximal tubule cell responses to BMP2 and BMP7 does not reveal any qualitative difference, suggesting that identical BMP gene targets may be activated in healthy and injured organs. Interestingly, our gene profiling analysis shows that BMP signaling activates a number of Notch regulated transcription factors, including HEY1. As in other biological systems, HEY1 functions as a negative feedback regulator of BMP2 expression in the proximal tubule. In summary, this work reveals endogenous BMP signaling patterns in the healthy human and mouse kidneys, and identifies novel gene targets, some of which are involved in the complex regulation of BMP signaling in the adult kidney.
Collapse
Affiliation(s)
- Barry W Larman
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074, United States
| | | | | | | |
Collapse
|
82
|
Bayir Y, Karagoz Y, Karakus E, Albayrak A, Sengul O, Can I, Yayla N, Kuskun U, Keles MS. Nigella SativaReduces Tissue Damage in Rat Ovaries Subjected to Torsion and Detorsion: Oxidative Stress, Proinflammatory Response and Histopathological Evaluation. Gynecol Obstet Invest 2012; 74:41-9. [DOI: 10.1159/000336295] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 01/03/2012] [Indexed: 12/11/2022]
|
83
|
Abstract
Acute kidney injury (AKI) is recognized as an independent risk factor for morbidity and mortality. Unfortunately, this syndrome was historically underdiagnosed due to inconsistent definition of AKI as well as insensitive and nonspecific diagnostic tools. Recent advances in defining AKI, understanding its pathophysiology, and improving its diagnostic accuracy have an impact in disease management and clinical outcome. Prompt recognition and treatment of AKI still remains the cornerstone of clinical management of this syndrome. This chapter focuses on the recent advances in diagnosis of AKI using novel serum and urine biomarkers. The role of neutrophil gelatinase-associated lipocalin (NGAL) in pathophysiology and diagnosis of AKI is presented. A detailed analysis of the biology of NGAL and presentation of laboratory methods of measurement is also provided. The role of NGAL as biomarker beyond the boundaries of nephrology is also presented.
Collapse
|
84
|
Yoshitomi T, Hirayama A, Nagasaki Y. The ROS scavenging and renal protective effects of pH-responsive nitroxide radical-containing nanoparticles. Biomaterials 2011; 32:8021-8. [DOI: 10.1016/j.biomaterials.2011.07.014] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Accepted: 07/05/2011] [Indexed: 12/19/2022]
|
85
|
Mahmoud MF, Diaai AA, Ahmed F. Evaluation of the efficacy of ginger, Arabic gum, and Boswellia in acute and chronic renal failure. Ren Fail 2011; 34:73-82. [PMID: 22017619 DOI: 10.3109/0886022x.2011.623563] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This study was conducted to evaluate the effects of Zingiber officinale Roscoe (Ginger), Arabic gum (AG), and Boswellia on both acute and chronic renal failure (CRF) and the mechanisms underlying their effects. Acute renal failure was induced by 30 min ischemia followed by 24 h reperfusion, while CRF was induced by adenine feeding for 8 weeks. Prophylactic oral administration of ginger, AG, Boswellia, or vehicle (in control groups) was started 3 days before and along with adenine feeding in different groups or 7 days before ischemia-reperfusion. Ginger and AG showed renoprotective effects in both models of renal failure. These protective effects may be attributed at least in part to their anti-inflammatory properties as evident by attenuating serum C-reactive protein levels and antioxidant effects as evident by attenuating lipid peroxidation marker, malondialdehyde levels, and increasing renal superoxide dismutase activity. Ginger was more potent than AG in both models of renal failure. However, Boswellia showed only partial protective effect against both acute renal failure and CRF and it had no antioxidant effects. Finally, we can say that ginger and AG could be beneficial adjuvant therapy in patients with acute renal failure and CRF to prevent disease progression and delay the need for renal replacement therapy.
Collapse
Affiliation(s)
- Mona Fouad Mahmoud
- Department of Pharmacology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| | | | | |
Collapse
|
86
|
Preconditioning with physiological levels of ethanol protect kidney against ischemia/reperfusion injury by modulating oxidative stress. PLoS One 2011; 6:e25811. [PMID: 22022451 PMCID: PMC3192120 DOI: 10.1371/journal.pone.0025811] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Accepted: 09/11/2011] [Indexed: 01/01/2023] Open
Abstract
Background Oxidative stress due to excessive production of reactive oxygen species (ROS) and subsequent lipid peroxidation plays a critical role in renal ischemia/reperfusion (IR) injury. The purpose of current study is to demonstrate the effect of antecedent ethanol exposure on IR-induced renal injury by modulation of oxidative stress. Materials and Methods Bilateral renal warm IR was induced in male C57BL/6 mice after ethanol or saline administration. Blood ethanol concentration, kidney function, histological damage, inflammatory infiltration, cytokine production, oxidative stress, antioxidant capacity and Aldehyde dehydrogenase (ALDH) enzymatic activity were assessed to evaluate the impact of antecedent ethanol exposure on IR-induced renal injury. Results After bilateral kidney ischemia, mice preconditioned with physiological levels of ethanol displayed significantly preserved renal function along with less histological tubular damage as manifested by the reduced inflammatory infiltration and cytokine production. Mechanistic studies revealed that precondition of mice with physiological levels of ethanol 3 h before IR induction enhanced antioxidant capacity characterized by significantly higher superoxidase dismutase (SOD) activities. Our studies further demonstrated that ethanol pretreatment specifically increased ALDH2 activity, which then suppressed lipid peroxidation by promoting the detoxification of Malondialdehyde (MDA) and 4-hydroxynonenal (HNE). Conclusions Our results provide first line of evidence indicating that antecedent ethanol exposure can provide protection for kidneys against IR-induced injury by enhancing antioxidant capacity and preventing lipid peroxidation. Therefore, ethanol precondition and ectopic ALDH2 activation could be potential therapeutic approaches to prevent renal IR injury relevant to various clinical conditions.
Collapse
|
87
|
Mariscalco G, Lorusso R, Dominici C, Renzulli A, Sala A. Acute Kidney Injury: A Relevant Complication After Cardiac Surgery. Ann Thorac Surg 2011; 92:1539-47. [DOI: 10.1016/j.athoracsur.2011.04.123] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2011] [Revised: 04/27/2011] [Accepted: 04/29/2011] [Indexed: 11/25/2022]
|
88
|
Kunin M, Holtzman EJ, Melnikov S, Dinour D. Urinary organic anion transporter protein profiles in AKI. Nephrol Dial Transplant 2011; 27:1387-95. [DOI: 10.1093/ndt/gfr541] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
89
|
Lindoso RS, Araujo DS, Adão-Novaes J, Mariante RM, Verdoorn KS, Fragel-Madeira L, Caruso-Neves C, Linden R, Vieyra A, Einicker-Lamas M. Paracrine interaction between bone marrow-derived stem cells and renal epithelial cells. Cell Physiol Biochem 2011; 28:267-78. [PMID: 21865734 DOI: 10.1159/000331739] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2011] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND/AIMS Renal tubular cells are the main target of ischemic insult associated with acute renal injury. Low oxygen and nutrient supplies result in ATP depletion, leading to cell death and loss of renal function. A possible mechanism by which bone marrow-derived cells support renal tissue regeneration relies on the capacity of mononuclear cells (BMMC), particularly mesenchymal stem cells (MSC), to secrete paracrine factors that mediate support for kidney regeneration. METHODS BMMC/MSC and renal cells (LLC-PK(1) from pig and IRPTC from rat) were co-cultured under stressful conditions (ATP depletion and/or serum free starvation), physically separated by a microporous membrane (0.4 μm), was used to determine whether bone marrow-derived cells can interact with renal cells in a paracrine manner. RESULTS This interaction resulted in stimulation of renal cell proliferation and the arrest of cell death. MSC elicit effective responses in renal cells in terms of stimulating proliferation and protection. Such effects are observed in renal cells co-cultured with rat BMMC/MSC, an indication that paracrine mechanisms are not entirely species-specific. CONCLUSION The paracrine action of BMMC/MSC was influenced by a renal cell stimulus released during stress, indicating that cross-talk with injured cells is required for renal regeneration supported by bone marrow-derived cells.
Collapse
Affiliation(s)
- Rafael S Lindoso
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Lee SY, Kim DH, Sung SA, Kim MG, Cho WY, Kim HK, Jo SK. Sphingosine-1-phosphate reduces hepatic ischaemia/reperfusion-induced acute kidney injury through attenuation of endothelial injury in mice. Nephrology (Carlton) 2011; 16:163-73. [PMID: 21272128 DOI: 10.1111/j.1440-1797.2010.01386.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIM Hepatic ischaemia/reperfusion injury (IRI) frequently complicates acute kidney injury (AKI) during the perioperative period. This study was to determine whether hepatic IRI causes AKI and the effect of the sphingosine-1-phosphate (S1P) on AKI. METHODS S1P and vehicle were given to mice before ischaemia and mice were subjected to hepatic IRI. Plasma creatinine (PCr), alanine transaminase (ALT), urinary neutrophil gelatinase-associated lipocalin (NGAL) and renal histological changes were determined. As a marker of endothelial injury, vascular permeability was measured. The effect of VPC 23019, a S1P(1) receptor antagonist, was also assessed. RESULTS Hepatic IRI resulted in liver injury (increased ALT) and systemic inflammation. Kidneys showed elevated inflammatory cytokines, leucocyte infiltration, increased vascular permeability, tubular cell apoptosis and increased urinary NGAL, although PCr did not increase. Pretreatment with S1P resulted in an attenuation of systemic inflammation and kidney injury without any effect on plasma ALT or peripheral lymphocytes. The protective effect of S1P was partially reversed by VPC 23019, suggesting the important contribution of the S1P/S1P(1) pathway to protect against hepatic IRI-induced AKI. CONCLUSION The study data demonstrate the important contribution of systemic inflammation and endothelial injury to AKI following hepatic IRI. Modulation of the S1P/S1P(1) receptor pathway might have some therapeutic potential in hepatic IRI-induced kidney injury.
Collapse
Affiliation(s)
- So-Young Lee
- Department of Nephrology, Eulji University Hospital, Korea
| | | | | | | | | | | | | |
Collapse
|
91
|
Marley CS, Siegrist T, Kurta J, O'Brien F, Bernstein M, Solomon S, Coleman JA. Cold Intravascular Organ Perfusion for Renal Hypothermia During Laparoscopic Partial Nephrectomy. J Urol 2011; 185:2191-5. [DOI: 10.1016/j.juro.2011.02.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Indexed: 10/18/2022]
Affiliation(s)
- Ciara Siobhan Marley
- Department of Surgery, Urology Service and Department of Radiology, Interventional Service (SS), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Timothy Siegrist
- Department of Surgery, Urology Service and Department of Radiology, Interventional Service (SS), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jordan Kurta
- Department of Surgery, Urology Service and Department of Radiology, Interventional Service (SS), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Frank O'Brien
- Department of Surgery, Urology Service and Department of Radiology, Interventional Service (SS), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Melanie Bernstein
- Department of Surgery, Urology Service and Department of Radiology, Interventional Service (SS), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Steven Solomon
- Department of Surgery, Urology Service and Department of Radiology, Interventional Service (SS), Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jonathan Andrew Coleman
- Department of Surgery, Urology Service and Department of Radiology, Interventional Service (SS), Memorial Sloan-Kettering Cancer Center, New York, New York
| |
Collapse
|
92
|
Abstract
Acute kidney injury (AKI) increases morbidity and mortality, particularly for the critically ill. Recent definitions standardizing AKI to reflect graded changes in serum creatinine and urine output (per the Risk, Injury, Failure, Loss, and End-stage renal failure [RIFLE] and Acute Kidney Injury Network [AKIN] criteria) with severity of renal injury and developments in AKI pathobiology are being utilized to identify biomarkers of early kidney injury. These developments may be useful in the early intervention of preventing AKI. Although there has been progress in the management of AKI, therapeutic challenges include appropriate prophylaxis prior to contrast administration, use of diuretics, vasopressors, and the type and dose of renal replacement therapy. Future use of bioartificial dialyzers, plasma therapies, and the possibility of stem cell regeneration of injured kidney tissue are being actively investigated to provide alternative treatment options for AKI. This review aims to provide an overview of current practices, available therapies, and continued research in AKI therapy.
Collapse
Affiliation(s)
- Devasmita Choudhury
- VA North Texas Health Care Systems, Dallas VA Medical Center, Dallas, TX 75216, USA.
| |
Collapse
|
93
|
Monitoring of plasma creatinine and urinary γ-glutamyl transpeptidase improves detection of acute kidney injury by more than 20%. Crit Care Med 2011; 39:52-6. [PMID: 21178528 DOI: 10.1097/ccm.0b013e3181fa431a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
OBJECTIVES We sought to determine how early we can detect acute kidney injury inpatients at intensive care unit admission by combining the use of plasma creatinine and urinary γ-glutamyl transpeptidase. DESIGN Prospective study including development (n = 100) and validation (n = 56) cohorts. SETTINGS Intensive care unit of a university hospital. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS To determine acute kidney injury, we subtracted measured creatinine clearance from theoretical creatinine clearance with a 25% reduction signifying acute kidney injury. Its incidence in 100 consecutive patients was 36%. An indexed urinary γ-glutamyl transpeptidase-to-urinary creatinine ratio was significantly increased in the patients with acute kidney injury and did not correlate with plasma creatinine (p = .3). Using a predefined threshold of indexed urinary γ-glutamyl transpeptidase-to-urinary creatinine ratio (>12.4 units/mmol) and plasma creatinine (>89 μmol/L), acute kidney injury detection was significantly improved, making it possible to detect 22 (22%) additional patients with acute kidney injury. This finding was confirmed in the validation group. The rates of false-positive results were 30% and 19% in the data development and internal validation cohorts, respectively. CONCLUSIONS The use of low-cost, widely available markers (creatinine and urinary γ-glutamyl transpeptidase) increases the detection of acute kidney injury. Further studies are needed to determine the impact on outcome with the use of these biomarkers.
Collapse
|
94
|
Abstract
Acute kidney injury is commonly encountered in critically ill patients, and is associated with worse outcomes. Fluid therapy is a key component in the management of these patients, often leading to fluid overload, especially in the setting of septic acute kidney injury. Emerging data overwhelmingly suggest that fluid overload in these patients may be associated with adverse outcomes. Management of such patients should include a strategy of early guided resuscitation, followed by careful assessment of fluid status, and early initiation of renal replacement therapy as soon as it is deemed safe, aiming for a neutral or negative fluid balance. This review will focus on the pathophysiological link between fluid overload and acute kidney injury, mechanisms of organ dysfunction in fluid overload, and strategies for management.
Collapse
Affiliation(s)
- Preethi Yerram
- Department of Internal Medicine, Division of Nephrology, University of Missouri-Columbia, Columbia, Missouri, USA
| | | | | |
Collapse
|
95
|
Wan X, Fan L, Hu B, Yang J, Li X, Chen X, Cao C. Small interfering RNA targeting IKKβ prevents renal ischemia-reperfusion injury in rats. Am J Physiol Renal Physiol 2011; 300:F857-63. [PMID: 21289055 DOI: 10.1152/ajprenal.00547.2010] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The transcription factor NF-κB has been found critical to the pathogenesis of renal ischemia-reperfusion injury, which is a major cause of acute kidney injury (AKI). Activation of NF-κB is dependent upon the activation of the specific inhibitory κB kinase (IKK) subunit IKKβ. Here, we investigate whether small interfering RNA (siRNA) targeting IKKβ protects rats from renal ischemia- reperfusion injury in vivo. Renal ischemia-reperfusion injury was induced by clamping the renal artery for 45 min. Rats were treated before ischemia with IKKβ siRNA or scrambled siRNA, administered by renal artery injection. Treated animals were evaluated for renal IKKβ protein and mRNA expression, blood biochemistry, tissue histopathology, NF-κB/DNA binding activity, and expression of two downstream inflammatory cytokines, neutrophil gelatinase-associated lipocalin (NGAL) and IL-18. A local injection of IKKβ siRNA resulted in inhibition of renal IKKβ gene expression, NF-κB/DNA binding activity, and expression of NGAL and IL-18. Rats pretreated with IKKβ siRNA had significantly less blood urea nitrogen and serum creatinine levels and less renal tubular damage scores. Consequently, our data confirm that targeted silencing of IKKβ using siRNA substantially diminishes kidney injury and inflammation following ischemia-reperfusion.
Collapse
Affiliation(s)
- Xin Wan
- Dept. of Nephrology, Nanjing First Hospital affiliated to Nanjing Medical Univ., Nanjing 210006, China
| | | | | | | | | | | | | |
Collapse
|
96
|
Kwak W, Jang HS, Belay T, Kim J, Ha YS, Lee SW, Ahn BC, Lee J, Park KM, Yoo J. Evaluation of kidney repair capacity using 99mTc-DMSA in ischemia/reperfusion injury models. Biochem Biophys Res Commun 2011; 406:7-12. [PMID: 21277288 DOI: 10.1016/j.bbrc.2011.01.085] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Accepted: 01/22/2011] [Indexed: 11/29/2022]
Abstract
Quantitative (99m)Tc-DMSA renal uptake was studied in different renal ischemia/reperfusion (I/R) mice models for the assessment of renal repair capacity. Mice models of nephrectomy, uni- and bi-lateral I/R together with sham-operated mice were established. At 1h, 1d, 4d, 1, 2 and 3 wk after I/R, (99m)Tc-DMSA (27.7 ± 1.3 MBq) was injected via tail vein and after 3h post-injection, the mice were scanned for 30 min with pinhole equipped gamma camera. Higher uptake of (99m)Tc-DMSA was measured in normal kidneys of uni-lateral I/R model and nephrectomized kidney I/R model at 3 wk post-surgery. Comparing the restoration capacities of the affected kidneys of nephrectomy, uni- and bi-lateral I/R models, higher repair capacity was observed in the nephrectomized model followed by bi-lateral then uni-lateral models. The normal kidney may retard the restoration of damaged kidney in uni-lateral I/R model. Moreover, 3 wk after Uni-I/R, the size of injured kidney was significantly smaller than non-ischemic contralateral and sham operated kidneys, while nephrectomy I/R kidneys were significantly enlarged compared to all others at 3 wk post-surgery. Very strong correlation between (99m)Tc-DMSA uptake and weight of dissected kidneys in I/R models was observed. Consistent with (99m)Tc-DMSA uptake results, all histological results indicate that kidney recovery after injury is correlated with the amount of intact tubules and kidney sizes. In summary, our study showed good potentials of (99m)Tc-DMSA scan as a promising non-invasive method for evaluation of kidney restoration after I/R injuries. Interestingly, mice with Bi-I/R injury showed faster repair capacity than those with uni-I/R.
Collapse
Affiliation(s)
- Wonjung Kwak
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Kim JI, Jang HS, Park KM. Endotoxin-induced renal tolerance against ischemia and reperfusion injury is removed by iNOS, but not eNOS, gene-deletion. BMB Rep 2010; 43:629-34. [PMID: 20846496 DOI: 10.5483/bmbrep.2010.43.9.629] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Endotoxin including lipopolysaccharide (LPS) confers organ tolerance against subsequent challenge by ischemia and reperfusion (I/R) insult. The mechanisms underlying this powerful adaptive defense remain to be defined. Therefore, in this study we attempted to determine whether nitric oxide (NO) and its associated enzymes, inducible NOS (iNOS) and endothelial NOS (eNOS, a constitutive NOS), are associated with LPS-induced renal tolerance against I/R injury, using iNOS (iNOS knock-out) or eNOS (eNOS knock-out) gene-deleted mice. A systemic low dose of LPS pretreatment protected kidney against I/R injury. LPS treatment increased the activity and expression of iNOS, but not eNOS, in kidney tissue. LPS pretreatment in iNOS, but not eNOS, knock-out mice did not protect kidney against I/R injury. In conclusion, the kidney tolerance to I/R injury conferred by pretreatment with LPS is mediated by increased expression and activation of iNOS.
Collapse
Affiliation(s)
- Jee In Kim
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Korea
| | | | | |
Collapse
|
98
|
Liao XH, Zhang L, Liu Q, Sun H, Peng CM, Guo H. Augmenter of liver regeneration protects kidneys from ischaemia/reperfusion injury in rats. Nephrol Dial Transplant 2010; 25:2921-2929. [PMID: 20332418 DOI: 10.1093/ndt/gfq151] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Augmenter of liver regeneration (ALR), which was identified originally for its crucial role in promoting hepatocyte proliferation, is expressed in both the liver and kidney. Protective effects of ALR have been demonstrated in experimental models of acute liver failure. In the present study, we investigated the effect of ALR on renal ischaemia/reperfusion (I/R) injury and the possible mechanisms of its action. METHODS Male Sprague-Dawley rats were subjected to renal ischaemia for 60 min and then administered with either saline or recombinant human ALR (rhALR). A sham-operated group served as control. The expression of ALR in the sham-operated and acute kidney injury (AKI) groups was detected by immunohistochemistry and western blotting. Renal dysfunction and injury were assessed by measurement of serum biochemical markers and histological grading. Expression of proliferating cell nuclear antigen (PCNA) was determined by immunohistochemistry. RESULTS Renal ALR expression increased significantly in rats with ischaemic AKI compared with the sham-operated rats. Serum biochemical parameters showed that renal dysfunction was improved by administration of rhALR. Histological analysis revealed that treatment with rhALR also reduced the extent of kidney injury. Intraperitoneal injection of rhALR enhanced the proliferation of renal tubular cells. Conclusions. Administration of rhALR effectively reduces tubular injury and ameliorates the impairment of renal function. The protective effect of rhALR is associated with enhancement of renal tubular cell regeneration.
Collapse
Affiliation(s)
- Xiao-hui Liao
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | | | | | | | | | | |
Collapse
|
99
|
Russo P. Partial nephrectomy for renal cancer (part II): the impact of renal ischaemia, patient preparation, surgical approaches, management of complications and utilization. BJU Int 2010; 105:1494-507. [PMID: 20553456 DOI: 10.1111/j.1464-410x.2010.09388.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Paul Russo
- Department of Surgery, Urology Service, and Weill Medical College, Cornell University, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY, USA.
| |
Collapse
|
100
|
Cheung JS, Fan SJ, Chow AM, Zhang J, Man K, Wu EX. Diffusion tensor imaging of renal ischemia reperfusion injury in an experimental model. NMR IN BIOMEDICINE 2010; 23:496-502. [PMID: 20175152 DOI: 10.1002/nbm.1486] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Renal ischemia reperfusion injury (IRI) is a major cause of acute renal failure. It occurs in various clinical settings such as renal transplantation, shock and vascular surgery. Serum creatinine level has been used as an index for estimating the degree of renal functional loss in renal IRI. However, it only evaluates the global renal function. In this study, diffusion tensor imaging (DTI) was used to characterize renal IRI in an experimental rat model. Spin-echo echo-planar DTI with b-value of 300 s/mm(2) and 6 diffusion gradient directions was performed at 7 T in 8 Sprague-Dawley (SD) with 60-min unilateral renal IRI and 8 normal SD rats. Apparent diffusion coefficient (ADC), directional diffusivities and fractional anisotropy (FA) were measured at the acute stage of IRI. The IR-injured animals were also examined by diffusion-weighted imaging with 7 b-values up to 1000 s/mm(2) to estimate true diffusion coefficient (D(true)) and perfusion fraction (P(fraction)) using a bi-compartmental model. ADC of injured renal cortex (1.69 +/- 0.24 x 10(-3) mm(2)/s) was significantly lower (p < 0.01) than that of contralateral intact cortex (2.03 +/- 0.35 x 10(-3) mm(2)/s). Meanwhile, both ADC and FA of IR-injured medulla (1.37 +/- 0.27 x 10(-3) mm(2)/s and 0.28 +/- 0.04, respectively) were significantly less (p < 0.01) than those of contralateral intact medulla (2.01 +/- 0.38 x 10(-3) mm(2)/s and 0.36 +/- 0.04, respectively). The bi-compartmental model analysis revealed the decrease in D(true) and P(fraction) in the IR-injured kidneys. Kidney histology showed widespread cell swelling and erythrocyte congestion in both cortex and medulla, and cell necrosis/apoptosis and cast formation in medulla. These experimental findings demonstrated that DTI can probe both structural and functional information of kidneys following renal IRI.
Collapse
Affiliation(s)
- Jerry S Cheung
- Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | | | | | | | | |
Collapse
|