Comprehensive Physiology Wiley Online Library

Pathophysiology of Acute Kidney Injury

Full Article on Wiley Online Library



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. © 2012 American Physiological Society. Compr Physiol 2:1303‐1353, 2012.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1.

Relationship between the clinical phases and the cellular phases of ischemic acute kidney injury (AKI), and the temporal impact on organ function as represented by glomerular filtration rate (GFR). Prerenal azotemia exists when a reduction in renal blood flow causes a reduction in GFR. A variety of cellular and vascular adaptations maintain renal epithelial integrity during these phases. The initiation phase ossuces when a further reduction in renal blood flow results in cellular injury, particularly the renal tubular epithelial cells, and a continued decline in GFR. Vascular and inflammatory processes that contribute to further cell injury and a further decline in GFR usher in the extension phase. During the maintenance phase, GFR reaches a stable nadir as cellular repair processes are initiated to maintain and reestablish organ integrity. The recovery phases in marked by a return of normal cell and organ function than results in an improvement in GFR. Adapted, with permission, from reference 532.

Figure 2. Figure 2.

Regional blood flow is altered following injury in ischemic acute kidney injury (AKI). Immediately following ischemic injury total renal blood flow is reduced but more striking are the regional deficits in blood flow that exist in the cortex, outer stripe of outer medulla and inner stripe of the outer medulla as indicated in (A) [data adapted, with permission, from reference 251]. As overall blood flow starts to recover in the ensuing hours after injury, profound regional alterations in blood flow remain with progressive and profound reduction of the blood flow to the outer stripe of the outer medulla as indicated in (B) [data adapted, with permission, from reference 202].

Figure 3. Figure 3.

Interplay between tubular and vascular injury leading to sustained reductions of glomerular filtration rate (GFR) in the extension phase of acute kidney injury (AKI). Injury induced by ischemia can result in damage to both the tubular as well as the microvascular compartment. Resolution of vasoconstriction appears effective at reducing injury when administered prophylactically, but not following established injury. Resistance may be due to exacerbated inflammation, which may impart reductions in renal blood flow (RBF) and GFR insensitive to vasodilator therapies. Of central importance in this process is the activation of inflammatory processes that are influenced by factors released by damaged proximal tubules as well as adhesion of damaged microvascular cells. Infiltrating leukocytes may impinge on RBF either by secreting vasoactive factors, or by contributing to the disruption of flow by physical interference. In addition, exacerbated hypoxia leading to tubular obstruction may contribute to reductions in GFR independent of vasodilator therapy.

Reprinted from Microvascular Research 77: 4‐7, 2009 531 with permission from Elsevier.
Figure 4. Figure 4.

Organization of vascular compartment in the kidney. (A) The medulla is arterial supplied from the efferent arterioles of the juxtamedullary glomeruli, giving supply to the descending arterial vasa recta, and further to the ascending venous vasa recta, draining into the arcuate veins. OSOM, outer stripe of outer medulla; ISOM, inner stripe of outer medulla; IM, inner medulla. (B) Very strong expression of b130‐1, 2 h after ischemia/reperfusion (I/R) injury of the kidney, at the level of the ascending vasa recta. (C) Detailed expression of b130‐1, 2 h after I/R injury of the rat kidney, at the level of the ascending venous vasa recta. (D) Detailed expression of b130‐1, 2 h after I/R injury of the human kidney, at the level of the ascending venous vasa recta. (E) Trapping of CD28‐expressing T cells in the ascending vasa recta (HIS‐17 staining). (F) Trapping of monocytes/macrophages in the ascending vasa recta (ED‐1 staining). (G) This trapping of leukocytes in the ascending vasa recta results in an upstream congestion at the ascending arterial vasa recta. This congestion, or no‐reflow, represents a well‐known phenomenon in acute ischemic injury, exacerbating during reperfusion the ischemic damage.

Reprinted, with permission from Macmillan Publishers Ltd; Kidney International, 2004 626.
Figure 5. Figure 5.

A proposed model for the dephosphorylation, activation, and translocation of actin depolymerizing factor (ADF) to the apical microvilli during ischemia. Under physiological conditions, the distribution of ADF and phosphorylated ADF (pADF) in proximal tubule cells is diffused throughout the cytoplasm with little or no localization to the apical microvillar region. With ischemia, pADF is dephosphorylated and, therefore, activated. In addition, the diffused cytoplasmic localization of ADF changes with ADF now concentrating at the apical membrane region of the cell. It is hypothesized that ADF relocalizes to the apical microvillar region and binds the microfilament core, resulting in markedly enhanced filament severing and depolymerization. Breakdown of the microfilament core is accompanied by dramatic changes in the overlying microvillar membrane. The microvillar membrane is internalized or extruded as membrane vesicle or blebs. These vesicles contain both ADF and monomeric actin. Figure and legend adapted, ith permission, from reference 15.

Figure 6. Figure 6.

Evidence of mitochondrial depolarization in kidney tissue slices by chemical anoxia using multiphoton imaging. PTs loaded with tetramethylrhodamine methylester (TMRM) showed rapid depolarization of Δψm after chemical anoxia. Bar = 20 μm. (B) In the diphtheria toxin (DT), the decrease was slower and Δψm was not completely depolarized after 60 min of anoxia; however, in the presence of oligomycin (5 μg/mL), Δψm depolarized rapidly in distal tubular cells when exposed to anoxia. Data are means ± SE signal per tubule from a total of 15 PTs, 15 DTs without oligomycin, and 29 DTs with oligomycin from three separate slices for each experiment. The data were normalized from 1 (value at t = 0, taken as resting Δψm) to 0 (minimum value after FCCP, taken as 0 mV).

Reprinted, with permission, by the American Society of Nephrology from reference 185.
Figure 7. Figure 7.

Illustration of the various stages of apoptotic cell death. (A) Depiction of the stereotypical changes including condensation, changes in nuclear structure, and fragmentation of the cell into small apoptotic bodies. In vivo, the apoptotic bodies are phagocytosed by neighboring cells, whereas in vitro they undergo swelling and eventual lysis (secondary necrosis). (B) Photographs of LLC‐PK1 cells undergoing apoptosis at the corresponding stages as shown in (A). Apoptosis was induced by overnight exposure of the cells to 50 μmol/L cisplatin. The cells in the first three photographs were stained with Hoechst dye, and the cells in the last photograph were stained with acrydine orange and ethidium bromide. In the last photograph, viable cells appear green, whereas the apoptotic cells with intact plasma membrane appear green with yellowish dots representing condensed chromatin; apoptotic cells and bodies that are undergoing secondary necrosis appear bright orange or red due to the plasma membrane damage and entry of ethidium bromide. Illustration adapted, with permission, from reference 415.

Figure 8. Figure 8.

The continuum of renal cell damage. Individual renal tubular cells are likely to respond in different ways to injury depending upon the severity of the noxious stimulus. The majority of cells presumably remain viable, either because they escape injury altogether, or because they are only sublethally injured and able to recover. More severe injury likely results in apoptosis, whereas necrosis only occurs when cells are subjected to extremely severe injury that leads to critical energy depletion and subsequent metabolic collapse. Legend and figure adapted, with permission, from reference 320.

Figure 9. Figure 9.

Overview of death‐signaling pathways in mammalian cells. The death receptor pathway (left) is initiated by the binding of a ligand (Eg: FasL) to its receptor Fas, which results in the sequential recruitment of FADD and procaspase‐8. c‐FLIP can block the recruitment of procaspase‐8 to the complex. The proximity of several procaspase‐8 molecules results in its activation. Caspase‐8 can proteolytically activate caspase‐3, or it can cleave Bid to its truncated form t‐Bid, which binds to Bax and gets integrated into the mitochondrial membrane to release cytochrome c. In response to various cellular stress‐induced apoptotic stimuli, the intrinsic mitochondrial pathway is activated. This pathway involves the translocation of proapoptotic molecules such as Bax from the cytosol to the mitochondrial membrane. Bax can release cytochromec from the mitochondria into the cytosol. Cytochromec associates with Apaf‐1 and caspase‐9 to form the apoptosome and subsequent activation of caspase‐3. Mitochondria also release apoptosis‐inducing factor (AIF) and Endo G, which may exert their effects on the nuclei. Mitochondria released Smac/Diablo and Omi/HtrA2 sequesters inhibitors of apoptosis (IAPs) to prevent them from inhibiting caspase‐3. BNIP3 is a Bcl‐2 family member that is translocated and integrated into the mitochondria. Unlike other Bcl‐2 family members, BNIP3 can induce necrotic cell death in response to death stimuli. Activation of poly (ADP‐ribose) polymerase (PARP) leads to NAD+ depletion and may induce mitochondrial depolarization to release AIF. ROS, reactive oxygen species. Legend and figure adapted, with permission, from reference 320.

Figure 10. Figure 10.

Control of heat shock protein (HSP) expression in response to cell stress. Shown are the known actions of the constitutively expressed Hsps, primarily of the Hsp70 family called heat shock congnates (HSC) in processing cellular functions. Cell stress increases denatured proteins increasing the demand for HSC. Heat shock transcription factor (HSF), reversibly binds to HSC and is released with the increased demand for HSC. HSF then rapidly initiates transcription for all inducible Hsps including Hsp70 and Hsp25/27. Adapted, with kind permission, from Springer Science + Business media. Pediatric Nephrology (6th ed.), edited by Avner E, Harmon W, Niaudet P and Yoshikawa N. Heidleberg: Chap 64; Pathogenesis of acute renal failure. Sreedharan R, Devarajan P, and Van Why S, 1579‐1602, reference 521.

Figure 11. Figure 11.

Repair and regeneration of renal proximal tubule cells following acute sublethal injury. Sublethally injured renal proximal tubule cells (RPTCs) either repair physiological functions and restore normal tubular function or dedifferentiate, migrate, and/or proliferate to replace lost cells, then differentiate and resume normal function. The processes of repair and regeneration work in concert to ensure relining of the damaged nephron and restoration of renal function.

Reprinted, with permission by The American Society of Pharmacology and Experimental Therapeutics, from reference 402.
Figure 12. Figure 12.

Evidence that sublethally damaged proximal tubules are the source of dividing cells during recovery from acute kidney injury (AKI) periodic acid‐Schiff (PAS) staining and immunostaining were performed in the kidney sections of creksp;Z/EG mice with renal IRI. (A) PAS staining of the kidney 2 days after IRI. Tubular injury is shown by the loss of brush border membrane, cell detachment from the basement membrane, and nuclear condensation in some cells (arrows). (B) Expression of PCNA in tubular cells (red, arrows). (C) Low‐power image of BrdU incorporation in renal tubules. Some BrdU‐containing cells (red, arrows) colocalized with enhanced Green fluorescent protein (EGFP)‐expressing cells (green). The arrowhead indicates BrdU incorporation in an EGFP‐negative cell. (D) BrdU incorporation (red, arrow) in the epithelial cells expressing EGFP (green). Note: the cre transgene labels tubular cells and their progeny with EGFP. The nuclei were counterstained with 4′,6‐diamidino‐2‐phenylindole (DAPI), and images were merged (B‐D). Scale bars: 20 μm.

Reprinted, with permission by the American Society of Clinical Investigation, from reference 325.
Figure 13. Figure 13.

Gross renal morphology and capillary filling in normal and postischemic kidneys. Representative stereoscopic views of 20‐μm microfil‐infused kidney section. Shown are microfil‐infused kidneys from a sham‐operated rat (A) at 4 (B) and 8 week (C) postischemic injury. In this stereoscopic view, microfil appears as bright yellow against a dark background. A reduction in microfil‐infused structures in recovered postischemic kidneys is evident. c, cortex; os, outer stripe of the outer medulla; is, inner stripe of the outer medulla; im, inner medulla. Magnification is shown.

Adapted, with permission, from reference 32.
Figure 14. Figure 14.

A potential role for vascular dropout in promoting the development of CKD following acute kidney injury (AKI). Acute injury has the potential to affect both tubular and vascular compartments. In addition to direct injury to the microvascular compartment, tubular injury may compromise normal vascular support, shifting the environment to one that promotes vascular impairment rather than vascular stability, including the loss of VEGF expression, the increase in tubular glomerular feedback‐β (TGF‐β) expression as well as several other angio inhibitory compounds. The resultant decrease in capillary structures has a number of potential consequences on renal function including the exacerbation of hypoxia and the impairment of Na handling hemodynamic responses. Hypoxia, along with the potential endothelial mesenchymal transition, is likely to participate in the development of fibrosis, which is also influenced by sustained immune/inflammatory activity. Figure modified, with permission, from and earlier version published in reference 29.



Figure 1.

Relationship between the clinical phases and the cellular phases of ischemic acute kidney injury (AKI), and the temporal impact on organ function as represented by glomerular filtration rate (GFR). Prerenal azotemia exists when a reduction in renal blood flow causes a reduction in GFR. A variety of cellular and vascular adaptations maintain renal epithelial integrity during these phases. The initiation phase ossuces when a further reduction in renal blood flow results in cellular injury, particularly the renal tubular epithelial cells, and a continued decline in GFR. Vascular and inflammatory processes that contribute to further cell injury and a further decline in GFR usher in the extension phase. During the maintenance phase, GFR reaches a stable nadir as cellular repair processes are initiated to maintain and reestablish organ integrity. The recovery phases in marked by a return of normal cell and organ function than results in an improvement in GFR. Adapted, with permission, from reference 532.



Figure 2.

Regional blood flow is altered following injury in ischemic acute kidney injury (AKI). Immediately following ischemic injury total renal blood flow is reduced but more striking are the regional deficits in blood flow that exist in the cortex, outer stripe of outer medulla and inner stripe of the outer medulla as indicated in (A) [data adapted, with permission, from reference 251]. As overall blood flow starts to recover in the ensuing hours after injury, profound regional alterations in blood flow remain with progressive and profound reduction of the blood flow to the outer stripe of the outer medulla as indicated in (B) [data adapted, with permission, from reference 202].



Figure 3.

Interplay between tubular and vascular injury leading to sustained reductions of glomerular filtration rate (GFR) in the extension phase of acute kidney injury (AKI). Injury induced by ischemia can result in damage to both the tubular as well as the microvascular compartment. Resolution of vasoconstriction appears effective at reducing injury when administered prophylactically, but not following established injury. Resistance may be due to exacerbated inflammation, which may impart reductions in renal blood flow (RBF) and GFR insensitive to vasodilator therapies. Of central importance in this process is the activation of inflammatory processes that are influenced by factors released by damaged proximal tubules as well as adhesion of damaged microvascular cells. Infiltrating leukocytes may impinge on RBF either by secreting vasoactive factors, or by contributing to the disruption of flow by physical interference. In addition, exacerbated hypoxia leading to tubular obstruction may contribute to reductions in GFR independent of vasodilator therapy.

Reprinted from Microvascular Research 77: 4‐7, 2009 531 with permission from Elsevier.


Figure 4.

Organization of vascular compartment in the kidney. (A) The medulla is arterial supplied from the efferent arterioles of the juxtamedullary glomeruli, giving supply to the descending arterial vasa recta, and further to the ascending venous vasa recta, draining into the arcuate veins. OSOM, outer stripe of outer medulla; ISOM, inner stripe of outer medulla; IM, inner medulla. (B) Very strong expression of b130‐1, 2 h after ischemia/reperfusion (I/R) injury of the kidney, at the level of the ascending vasa recta. (C) Detailed expression of b130‐1, 2 h after I/R injury of the rat kidney, at the level of the ascending venous vasa recta. (D) Detailed expression of b130‐1, 2 h after I/R injury of the human kidney, at the level of the ascending venous vasa recta. (E) Trapping of CD28‐expressing T cells in the ascending vasa recta (HIS‐17 staining). (F) Trapping of monocytes/macrophages in the ascending vasa recta (ED‐1 staining). (G) This trapping of leukocytes in the ascending vasa recta results in an upstream congestion at the ascending arterial vasa recta. This congestion, or no‐reflow, represents a well‐known phenomenon in acute ischemic injury, exacerbating during reperfusion the ischemic damage.

Reprinted, with permission from Macmillan Publishers Ltd; Kidney International, 2004 626.


Figure 5.

A proposed model for the dephosphorylation, activation, and translocation of actin depolymerizing factor (ADF) to the apical microvilli during ischemia. Under physiological conditions, the distribution of ADF and phosphorylated ADF (pADF) in proximal tubule cells is diffused throughout the cytoplasm with little or no localization to the apical microvillar region. With ischemia, pADF is dephosphorylated and, therefore, activated. In addition, the diffused cytoplasmic localization of ADF changes with ADF now concentrating at the apical membrane region of the cell. It is hypothesized that ADF relocalizes to the apical microvillar region and binds the microfilament core, resulting in markedly enhanced filament severing and depolymerization. Breakdown of the microfilament core is accompanied by dramatic changes in the overlying microvillar membrane. The microvillar membrane is internalized or extruded as membrane vesicle or blebs. These vesicles contain both ADF and monomeric actin. Figure and legend adapted, ith permission, from reference 15.



Figure 6.

Evidence of mitochondrial depolarization in kidney tissue slices by chemical anoxia using multiphoton imaging. PTs loaded with tetramethylrhodamine methylester (TMRM) showed rapid depolarization of Δψm after chemical anoxia. Bar = 20 μm. (B) In the diphtheria toxin (DT), the decrease was slower and Δψm was not completely depolarized after 60 min of anoxia; however, in the presence of oligomycin (5 μg/mL), Δψm depolarized rapidly in distal tubular cells when exposed to anoxia. Data are means ± SE signal per tubule from a total of 15 PTs, 15 DTs without oligomycin, and 29 DTs with oligomycin from three separate slices for each experiment. The data were normalized from 1 (value at t = 0, taken as resting Δψm) to 0 (minimum value after FCCP, taken as 0 mV).

Reprinted, with permission, by the American Society of Nephrology from reference 185.


Figure 7.

Illustration of the various stages of apoptotic cell death. (A) Depiction of the stereotypical changes including condensation, changes in nuclear structure, and fragmentation of the cell into small apoptotic bodies. In vivo, the apoptotic bodies are phagocytosed by neighboring cells, whereas in vitro they undergo swelling and eventual lysis (secondary necrosis). (B) Photographs of LLC‐PK1 cells undergoing apoptosis at the corresponding stages as shown in (A). Apoptosis was induced by overnight exposure of the cells to 50 μmol/L cisplatin. The cells in the first three photographs were stained with Hoechst dye, and the cells in the last photograph were stained with acrydine orange and ethidium bromide. In the last photograph, viable cells appear green, whereas the apoptotic cells with intact plasma membrane appear green with yellowish dots representing condensed chromatin; apoptotic cells and bodies that are undergoing secondary necrosis appear bright orange or red due to the plasma membrane damage and entry of ethidium bromide. Illustration adapted, with permission, from reference 415.



Figure 8.

The continuum of renal cell damage. Individual renal tubular cells are likely to respond in different ways to injury depending upon the severity of the noxious stimulus. The majority of cells presumably remain viable, either because they escape injury altogether, or because they are only sublethally injured and able to recover. More severe injury likely results in apoptosis, whereas necrosis only occurs when cells are subjected to extremely severe injury that leads to critical energy depletion and subsequent metabolic collapse. Legend and figure adapted, with permission, from reference 320.



Figure 9.

Overview of death‐signaling pathways in mammalian cells. The death receptor pathway (left) is initiated by the binding of a ligand (Eg: FasL) to its receptor Fas, which results in the sequential recruitment of FADD and procaspase‐8. c‐FLIP can block the recruitment of procaspase‐8 to the complex. The proximity of several procaspase‐8 molecules results in its activation. Caspase‐8 can proteolytically activate caspase‐3, or it can cleave Bid to its truncated form t‐Bid, which binds to Bax and gets integrated into the mitochondrial membrane to release cytochrome c. In response to various cellular stress‐induced apoptotic stimuli, the intrinsic mitochondrial pathway is activated. This pathway involves the translocation of proapoptotic molecules such as Bax from the cytosol to the mitochondrial membrane. Bax can release cytochromec from the mitochondria into the cytosol. Cytochromec associates with Apaf‐1 and caspase‐9 to form the apoptosome and subsequent activation of caspase‐3. Mitochondria also release apoptosis‐inducing factor (AIF) and Endo G, which may exert their effects on the nuclei. Mitochondria released Smac/Diablo and Omi/HtrA2 sequesters inhibitors of apoptosis (IAPs) to prevent them from inhibiting caspase‐3. BNIP3 is a Bcl‐2 family member that is translocated and integrated into the mitochondria. Unlike other Bcl‐2 family members, BNIP3 can induce necrotic cell death in response to death stimuli. Activation of poly (ADP‐ribose) polymerase (PARP) leads to NAD+ depletion and may induce mitochondrial depolarization to release AIF. ROS, reactive oxygen species. Legend and figure adapted, with permission, from reference 320.



Figure 10.

Control of heat shock protein (HSP) expression in response to cell stress. Shown are the known actions of the constitutively expressed Hsps, primarily of the Hsp70 family called heat shock congnates (HSC) in processing cellular functions. Cell stress increases denatured proteins increasing the demand for HSC. Heat shock transcription factor (HSF), reversibly binds to HSC and is released with the increased demand for HSC. HSF then rapidly initiates transcription for all inducible Hsps including Hsp70 and Hsp25/27. Adapted, with kind permission, from Springer Science + Business media. Pediatric Nephrology (6th ed.), edited by Avner E, Harmon W, Niaudet P and Yoshikawa N. Heidleberg: Chap 64; Pathogenesis of acute renal failure. Sreedharan R, Devarajan P, and Van Why S, 1579‐1602, reference 521.



Figure 11.

Repair and regeneration of renal proximal tubule cells following acute sublethal injury. Sublethally injured renal proximal tubule cells (RPTCs) either repair physiological functions and restore normal tubular function or dedifferentiate, migrate, and/or proliferate to replace lost cells, then differentiate and resume normal function. The processes of repair and regeneration work in concert to ensure relining of the damaged nephron and restoration of renal function.

Reprinted, with permission by The American Society of Pharmacology and Experimental Therapeutics, from reference 402.


Figure 12.

Evidence that sublethally damaged proximal tubules are the source of dividing cells during recovery from acute kidney injury (AKI) periodic acid‐Schiff (PAS) staining and immunostaining were performed in the kidney sections of creksp;Z/EG mice with renal IRI. (A) PAS staining of the kidney 2 days after IRI. Tubular injury is shown by the loss of brush border membrane, cell detachment from the basement membrane, and nuclear condensation in some cells (arrows). (B) Expression of PCNA in tubular cells (red, arrows). (C) Low‐power image of BrdU incorporation in renal tubules. Some BrdU‐containing cells (red, arrows) colocalized with enhanced Green fluorescent protein (EGFP)‐expressing cells (green). The arrowhead indicates BrdU incorporation in an EGFP‐negative cell. (D) BrdU incorporation (red, arrow) in the epithelial cells expressing EGFP (green). Note: the cre transgene labels tubular cells and their progeny with EGFP. The nuclei were counterstained with 4′,6‐diamidino‐2‐phenylindole (DAPI), and images were merged (B‐D). Scale bars: 20 μm.

Reprinted, with permission by the American Society of Clinical Investigation, from reference 325.


Figure 13.

Gross renal morphology and capillary filling in normal and postischemic kidneys. Representative stereoscopic views of 20‐μm microfil‐infused kidney section. Shown are microfil‐infused kidneys from a sham‐operated rat (A) at 4 (B) and 8 week (C) postischemic injury. In this stereoscopic view, microfil appears as bright yellow against a dark background. A reduction in microfil‐infused structures in recovered postischemic kidneys is evident. c, cortex; os, outer stripe of the outer medulla; is, inner stripe of the outer medulla; im, inner medulla. Magnification is shown.

Adapted, with permission, from reference 32.


Figure 14.

A potential role for vascular dropout in promoting the development of CKD following acute kidney injury (AKI). Acute injury has the potential to affect both tubular and vascular compartments. In addition to direct injury to the microvascular compartment, tubular injury may compromise normal vascular support, shifting the environment to one that promotes vascular impairment rather than vascular stability, including the loss of VEGF expression, the increase in tubular glomerular feedback‐β (TGF‐β) expression as well as several other angio inhibitory compounds. The resultant decrease in capillary structures has a number of potential consequences on renal function including the exacerbation of hypoxia and the impairment of Na handling hemodynamic responses. Hypoxia, along with the potential endothelial mesenchymal transition, is likely to participate in the development of fibrosis, which is also influenced by sustained immune/inflammatory activity. Figure modified, with permission, from and earlier version published in reference 29.

References
 1. Abul‐Ezz SR, Walker PD, Shah SV. Role of glutathione in an animal model of myoglobinuric acute renal failure. Proc Natl Acad Sci U S A 88: 9833‐9837, 1991.
 2. Adabag AS, Ishani A, Koneswaran S, Johnson DJ, Kelly RF, Ward HB, McFalls EO, Bloomfield HE, Chandrashekhar Y. Utility of N‐acetylcysteine to prevent acute kidney injury after cardiac surgery: A randomized controlled trial. Am Heart J 155: 1143‐1149, 2008.
 3. Agarwal A, Balla J, Alam J, Croatt AJ, Nath KA. Induction of heme oxygenase in toxic renal injury: A protective role in cisplatin nephrotoxicity in the rat. Kidney Int 48: 1298‐1307, 1995.
 4. Aki Y, Nishiyama A, Miyatake A, Kimura S, Kohno M, Abe Y. Role of adenosine A1 receptor in angiotensin II‐ and norepinephrine‐induced renal vasoconstriction. J Pharmacol Exp Ther 303: 117‐123, 2002.
 5. Akira S, Takeda K. Toll‐like receptor signalling. Nat Rev Immunol 4: 499‐511, 2004.
 6. Alejandro V, Scandling JD Jr, Sibley RK, Dafoe D, Alfrey E, Deen W, Myers BD. Mechanisms of filtration failure during postischemic injury of the human kidney. A study of the reperfused renal allograft. J Clin Invest 95: 820‐831, 1995.
 7. Alejandro VS, Nelson WJ, Huie P, Sibley RK, Dafoe D, Kuo P, Scandling JD Jr, Myers BD. Postischemic injury, delayed function and Na+/K(+)‐ATPase distribution in the transplanted kidney. Kidney Int 48: 1308‐1315, 1995.
 8. Alon US. Neonatal acute renal failure: The need for long‐term follow‐up. Clin Pediatr (Phila) 37: 387‐390, 1998.
 9. Amdur RL, Chawla LS, Amodeo S, Kimmel PL, Palant CE. Outcomes following diagnosis of acute renal failure in U.S. veterans: Focus on acute tubular necrosis. Kidney Int 76: 1089‐1097, 2009.
 10. Arany I, Megyesi JK, Kaneto H, Tanaka S, Safirstein RL. Activation of ERK or inhibition of JNK ameliorates H2O2 cytotoxicity in mouse renal proximal tubule cells. Kidney Int 65: 1231‐1239, 2004.
 11. Argaud L, Gateau‐Roesch O, Muntean D, Chalabreysse L, Loufouat J, Robert D, Ovize M. Specific inhibition of the mitochondrial permeability transition prevents lethal reperfusion injury. J Mol Cell Cardiol 38: 367‐374, 2005.
 12. Arora P, Rajagopalam S, Ranjan R, Kolli H, Singh M, Venuto R, Lohr J. Preoperative use of angiotensin‐converting enzyme inhibitors/angiotensin receptor blockers is associated with increased risk for acute kidney injury after cardiovascular surgery. Clin J Am Soc Nephrol 3: 1266‐1273, 2008.
 13. Arriero M, Brodsky SV, Gealekman O, Lucas PA, Goligorsky MS. Adult skeletal muscle stem cells differentiate into endothelial lineage and ameliorate renal dysfunction after acute ischemia. Am J Physiol Renal Physiol 287: F621‐F627, 2004.
 14. Ashworth SL, Molitoris BA. Pathophysiology and functional significance of apical membrane disruption during ischemia. Curr Opin Nephrol Hypertens 8: 449‐458, 1999.
 15. Ashworth SL, Sandoval RM, Hosford M, Bamburg JR, Molitoris BA. Ischemic injury induces ADF relocalization to the apical domain of rat proximal tubule cells. Am J Physiol Renal Physiol 280: F886‐F894, 2001.
 16. Askenazi D, Feig D, Graham N, Hui‐Stickle S, Goldstein SL. 3‐5 year longitudinal follow‐up of pediatric patients after acute renal failure. Kidney Int 69: 184‐189, 2006.
 17. Ates E, Geno E, Erkasap N, Erkasap S, Akman S, Firat P, Emre Sk, Kiper H. Renal protection by brief liver ischemia in rats. Transplantation 74: 1247‐1251, 2002.
 18. Atkinson SJ, Hosford MA, Molitoris BA. Mechanism of actin polymerization in cellular ATP depletion. J Biol Chem 279: 5194‐5199, 2004.
 19. Aufricht C, Lu E, Thulin G, Kashgarian M, Siegel NJ, Van Why SK. ATP releases HSP‐72 from protein aggregates after renal ischemia. Am J Physiol 274: F268‐F274, 1998.
 20. Bacallao R, Fine LG. Molecular events in the organization of renal tubular epithelium: From nephrogenesis to regeneration. Am J Physiol 257: F913‐F924, 1989.
 21. Bacallao R, Garfinkel A, Monke S, Zampighi G, Mandel L. ATP depletion: A novel method to study junctional properties in epithelial tissues. I. Rearrangement of the actin cytoskeleton. J Cell Sci 107: 3301‐3313, 1994.
 22. Badr KF, Ichikawa I. Prerenal failure: A deleterious shift from renal compensation to decompensation. N Engl J Med 319: 623‐629, 1988.
 23. Bae EH, Lee J, Ma SK, Kim IJ, Frøkiaer J, Nielsen S, Kim SY, Kim SW. Alpha‐lipoic acid prevents cisplatin‐induced acute kidney injury in rats. Nephrol Dial Transplant 24: 2692‐2700, 2009.
 24. Bagnasco S, Good D, Balaban R, Burg M. Lactate production in isolated segments of the rat nephron. Am J Physiol 248: F522‐F526, 1985.
 25. Bagshaw SM, Laupland KB, Doig CJ, Mortis G, Fick GH, Mucenski M, Godinez‐Luna T, Svenson LW, Rosenal T. Prognosis for long‐term survival and renal recovery in critically ill patients with severe acute renal failure: A population‐based study. Crit Care 9: R700‐R709, 2005.
 26. Baldwin DD, Maynes LJ, Berger KA, Desai PJ, Zuppan CW, Zimmerman GJ, Winkielman AM, Sterling TH, Tsai CK, Ruckle HC. Laparoscopic warm renal ischemia in the solitary porcine kidney model. Urology 64: 592‐597, 2004.
 27. Baliga R, Zhang Z, Baliga M, Ueda N, Shah SV. In vitro and in vivo evidence suggesting a role for iron in cisplatin‐induced nephrotoxicity. Kidney Int 53: 394‐401, 1998.
 28. Bard JBL, McConnell JE, Davies JA. Towards a genetic basis for kidney development. Mech Dev 48: 3‐11, 1994.
 29. Basile DP. The endothelial cell in ischemic acute kidney injury: Implications for acute and chronic function. Kidney Int 72: 151‐156, 2007.
 30. Basile DP, Donohoe DL, Cao X, Van Why S. Resistance to ischemic acute renal failure in the Brown Norway rat: A new model to study cytoprotection. Kidney Int 65: 2201‐2211, 2004.
 31. Basile DP, Donohoe DL, Roethe K, Mattson DL. Chronic renal hypoxia following ischemia/reperfusion injury: Effects of L‐Arginine on hypoxia and secondary damage. Am J Physiol Renal Physiol 284: F338‐F348, 2003.
 32. Basile DP, Donohoe DL, Roethe K, Osborn JL. Renal ischemic injury results in permanent damage to peritubular capillaries and influences long‐term function. Am J Physiol 281: F887‐F899, 2001.
 33. Basile DP, Fredrich K, Chelladurai B, Leonard EC, Parrish AR. Renal ischemia reperfusion inhibits VEGF expression and induces ADAMTS‐1, a novel VEGF inhibitor. Am J Physiol Renal Physiol 294: F928‐F936, 2008
 34. Basile DP, Fredrich K, Weihrauch DW, Hattan N, Chilian WM. Angiostatin and matrix metalloprotease expression following ischemic acute renal failure. Am J Physiol Renal Physiol 286: F893‐F902, 2004.
 35. Basile DP, Friedrich JL, Spahic J, Knipe NL, Mang HE, Leonard EC, Ashtiyani SC, Bacallao RL, Molitoris BA, Sutton TA. Impaired endothelial proliferation and mesenchymal transition contribute to vascular rarefaction following acute kidney injury. Am J Physiol Renal Physiol 300: F721‐F733, 2011.
 36. Basile DP, Liapis H, Hammerman MR. Expression of bcl‐2 and bax in regenerating rat renal tubules following ischemic injury. Am J Physiol 41: F640‐F647, 1997.
 37. Basile DP, Martin DR, Hammerman MH. Extracellular matrix‐related genes in kidney post‐ischemic injury: Potential role for TGF‐ß in repair. Am J Physiol 275: F894‐F903, 1998.
 38. Basile DP, Rovak JM, Martin DR, Hammerman MR. Increased transforming growth factor‐ß expression in regenerating rat renal tubules following ischemic injury. Am J Physiol 270: F500‐F509, 1996.
 39. Basnakian AG, Apostolov EO, Yin X, Napirei M, Mannherz HG, Shah SV. Cisplatin nephrotoxicity is mediated by deoxyribonuclease I. J Am Soc Nephrol 16: 697‐702, 2005.
 40. Basnakian AG, Ueda N, Hong X, Galitovsky VE, Yin X, Shah SV. Ceramide synthase is essential for endonuclease‐mediated death of renal tubular epithelial cells induced by hypoxia‐reoxygenation. Am J Physiol Renal Physiol 288: F308‐F314, 2005.
 41. Basnakian AG, Ueda N, Kaushal GP, Mikhailova MV, Shah SV. DNase I‐like endonuclease in rat kidney cortex that is activated during ischemia/reperfusion injury. J Am Soc Nephrol 13: 1000‐1007, 2002.
 42. Bastin J, Cambon N, Thompson M, Lowry OH, Burch HB. Change in energy reserves in different segments of the nephron during brief ischemia. Kidney Int 31: 1239‐1247, 1987.
 43. Bayati A, Hellberg O, Odlind B, Wolgast M. Prevention of ischaemic acute renal failure with superoxide dismutase and sucrose. Acta Physiol Scand 130: 367‐372, 1987.
 44. Bayir H, Kagan V. Bench‐to‐bedside review: Mitochondrial injury, oxidative stress and apoptosis ‐ there is nothing more practical than a good theory. Critical Care 12: 206, 2008.
 45. Bellomo R, Chapman M, Hickling K, Myburgh J. Low‐dose dopamine in patients with early renal dysfunction: A placebo‐controlled randomised trial. The Lancet 356: 2139‐2143, 2000.
 46. Benjamin IJ, McMillan DR. Stress (heat shock) proteins: Molecular chaperones in cardiovascular biology and disease. Circ Res 83: 117‐132, 1998.
 47. Bennett M, Dent CL, Ma Q, Dastrala S, Grenier F, Workman R, Syed H, Ali S, Barasch J, Devarajan P. Urine NGAL predicts severity of acute kidney injury after cardiac surgery: A prospective study. Clin J Am Soc Nephrol 3: 665‐673, 2008.
 48. Bernard GR, Vincent J‐L, Laterre P‐F, LaRosa SP, Dhainaut J‐F, Lopez‐Rodriguez A, Steingrub JS, Garber GE, Helterbrand JD, Ely EW, Fisher CJ. Efficacy and safety of recombinant human activated protein C for severe sepsis. New Eng J Med 344: 699‐709, 2001.
 49. Bia M, Tyler K. Effect of cyclosporine on renal ischemic injury. Transplantation 43: 800‐804, 1987.
 50. Bidmon B, Endemann M, Muller T, Arbeiter K, Herkner K, Aufricht C. Heat shock protein‐70 repairs proximal tubule structure after renal ischemia. Kidney Int 58: 2400‐2407, 2000.
 51. Biju MP, Akai Y, Shrimanker N, Haase VH. Protection of HIF‐1‐deficient primary renal tubular epithelial cells from hypoxia‐induced cell death is glucose dependent. Am J Physiol Renal Physiol 289: F1217‐F1226, 2005.
 52. Bishop AL, Hall A. Rho GTPases and their effector proteins. Biochem J 348: 241‐255, 2000.
 53. Blantz RC, Pelayo JC. A functional role for the tubuloglomerular feedback mechanism. Kidney Int 25: 739‐746, 1984.
 54. Blazer‐Yost BL, Watanabe M, Haverty TP, Ziyadeh FN. Role of insulin and IGF1 receptors in proliferation of cultured renal proximal tubule cells. Biochim Biophys Acta 1133: 329‐335, 1992.
 55. Blydt‐Hansen TD, Katori M, Lassman C, Ke B, Coito AJ, Iyer S, Buelow R, Ettenger R, Busuttil RW, Kupiec‐Weglinski JW. Gene transfer‐induced local heme oxygenase‐1 overexpression protects rat kidney transplants from ischemia/reperfusion injury. J Am Soc Nephrol 14: 745‐754, 2003.
 56. Bohle A, Jahnecke J, Meyer D, Schubert GE. Morphology of acute renal failure: Comparative data from biopsy and autopsy. Kidney Int Suppl 6: S9‐S16, 1976.
 57. Bolisetty S, Agarwal A. Neutrophils in acute kidney injury: Not neutral any more. Kidney Int 75: 674‐676, 2009.
 58. Bonomini V, Stefoni S, Vagelista A. Long‐term patient and renal prognosis in acute renal failure. Nephron 36: 169‐172, 1984.
 59. Bonventre JV. Mechanisms of ischemic acute renal failure. Kidney Int 43: 1160‐1178, 1993.
 60. Bonventre JV. Kidney ischemic preconditioning. Curr Opin Nephrol Hypertens 11: 43‐48, 2002.
 61. Bonventre JV, Brezis M, Siegel NJ, Rosen S, Portilla D, Venkatachalam MA. Acute renal failure I: Relative importance of proximal vs. distal tubular injury. Am J Physiol 275: F623‐F632: 1998.
 62. Bonventre JV, Weinberg JM. Recent advances in the pathophysiology of ischemic acute renal failure. J Am Soc Nephrol 14: 2199‐2210, 2003.
 63. Bonventre JV, Zuk A. Ischemic acute renal failure: An inflammatory disease? Kidney Int 66: 480‐485, 2004.
 64. Bor M, Durmuş O, Bilgihan A, Çevik C, Türközkan N. The beneficial effect of 2′‐deoxycoformycin in renal ischemia‐reperfusion is mediated both by preservation of tissue ATP and inhibition of lipid peroxidation. Int J Clin Lab Res 29: 75‐79, 1999.
 65. Borkan SC, Gullans SR. Molecular chaperones in the kidney. Ann Rev Physiol 64: 503‐527, 2002.
 66. Borthwick E, Ferguson A. Perioperative acute kidney injury: Risk factors, recognition, management, and outcomes. BMJ 341: c3365, 2010.
 67. Bosch X, Poch E, Grau JM. Rhabdomyolysis and acute kidney injury. New Eng J Med 361: 62‐72, 2009.
 68. Brady H, Brenner B, Clarkson M, Lieberthal W. Acute renal failure. In: Brenner B, editor. The Kidney. Philadelphia: WB Saunders, 2000, p. 1201‐1262.
 69. Brady HR, Kone BC, Stromski ME, Zeidel ML, Giebisch G, Gullans SR. Mitochondrial injury: An early event in cisplatin toxicity to renal proximal tubules. Am J Physiol Renal Physiol 258: F1181‐F1187, 1990.
 70. Brezis M, Rosen S. Hypoxia of the renal medulla‐its implications for disease. New Eng J Med 332: 647‐657, 1995.
 71. Briggs J, Kennedy A, Young L, Luke R, Gray M. Renal function after acute tubular necrosis. Br Med J 3: 513‐516, 1967.
 72. Bright R. Reports of Medical Cases, Selected with a View of Illustrating the Symptoms and Cure of Diseases by a Reference to Morbid Anatomy. London: Richard Taylor for Longman, Rees, Orme, Browne, and Greene, 1827.
 73. Brivet FG, Kleinknecht DJ, Loirat P, Landais PJ. Acute renal failure in intensive care units–causes, outcome, and prognostic factors of hospital mortality; a prospective, multicenter study. French Study Group on Acute Renal Failure. Crit Care Med 24: 192‐198, 1996.
 74. Brodsky SV, Yamamoto T, Tada T, Kim B, Chen J, Kajiya F, Goligorsky MS. Endothelial dysfunction in ischemic acute renal failure: Rescue by transplanted endothelial cells. Am J Physiol Renal Physiol 282: F1140‐F1149, 2002.
 75. Broekema M, Harmsen MC, van Luyn M, Koerts J, Persersen AH, Kooten TG, van Goor H, Navis G, Popa ER. Bone marrow‐derived myofibroblasts contribute to renal interstital myofibroblasts population and produce procollagen I after ischemia reperfusion in rats. J Am Soc Nephrol 18: 165‐175, 2007.
 76. Brooks C, Wei Q, Cho S, Dong Z. Regulation of mitochondrial dynamics in acute kidney injury in cell culture and rodent models. J Clin Invest 119: 1275‐1285, 2009.
 77. Bulent Gul C, Gullulu M, Oral B, Aydinlar A, Oz O, Budak F, Yilmaz Y, Yurtkuran M. Urinary IL‐18: A marker of contrast‐induced nephropathy following percutaneous coronary intervention? Clin Biochem 41: 544‐547, 2008.
 78. Burne‐Taney MJ, Rabb H. The role of adhesion molecules and T cells in ischemic renal injury. Curr Opin Nephrol Hypertens 12: 85‐90, 2003.
 79. Burne‐Taney MJ, Yokota N, Rabb H. Persistent renal and extrarenal immune changes after severe ischemic injury. Kidney Int 67: 1002‐1009, 2005.
 80. Bush KT, Tsukamoto T, Nigam SK. Selective degradation of E‐cadherin and dissolution of E‐cadherin‐catenin complexes in epithelial ischemia. Am J Physiol Renal Physiol 278: F847‐F852, 2000.
 81. Bussolati B, Bruno S, Grange C, Buttiglieri S, Deregibus MC, Cantino D, Camussi G. Isolation of renal progenitor cells from adult human kidney. Am J Pathol 166: 545‐555, 2005.
 82. Bussolati B, Tetta C, Camussi G. Contribution of stem cells to kidney repair. Am J Nephrol 28: 813‐822, 2008.
 83. Büyükgebiz O, Aktan AÖ, Haklar G, Yalçin AS, Yeğen C, Yalin R, Ercan ZS. BQ‐123, a specific endothelin (ET1) receptor antagonist, prevents ischemia‐reperfusion injury in kidney transplantation. Transplant Int 9: 201‐207, 1996.
 84. Bywaters E, Beal D. Crush injuries and renal function. Br Med J 1: 427‐432, 1941.
 85. Castaneda MP, Swiatecka‐Urban A, Mitsnefes MM, Feuerstein D, Kaskel FJ, Tellis V, Devarajan P. Activation of mitochondrial apoptotic pathways in human renal allografts after ischemiareperfusion injury. Transplantation 76: 50‐54, 2003.
 86. Chandraker A, Takada M, Nadeau KC, Peach R, Tilney NL, Sayegh MH. CD28‐b7 blockade in organ dysfunction secondary to cold ischemia/reperfusion injury. Kidney Int 52: 1678‐1684, 1997.
 87. Chatterjee PK, Cuzzocrea S, Brown PA, Zacharowski K, Stewart KN, Mota‐Filipe H, Thiemermann C. Tempol, a membrane‐permeable radical scavenger, reduces oxidant stress‐mediated renal dysfunction and injury in the rat. Kidney Int 58: 658‐673, 2000.
 88. Chen C‐F, Tsai S‐Y, Ma M‐C, Wu M‐S. Hypoxic preconditioning enhances renal superoxide dismutase levels in rats. J Physiol 552: 561‐569, 2003.
 89. Chen G, Bridenbaugh EA, Akintola AD, Catania JM, Vaidya VS, Bonventre JV, Dearman AC, Sampson HW, Zawieja DC, Burghardt RC, Parrish AR. Increased susceptibility of aging kidney to ischemic injury: Identification of candidate genes changed during aging, but corrected by caloric restriction. Am J Physiol Renal Physiol 293: F1272‐F1281, 2007.
 90. 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 79: 288‐299, 2011.
 91. Chen Y‐F, Li P‐L, Zou A‐P. Oxidative stress enhances the production and actions of adenosine in the kidney. Am J Physiol Regul Integr Comp Physiol 281: R1808‐R1816, 2001.
 92. Chen Z, Chua CC, Ho Y‐S, Hamdy RC, Chua BHL. Overexpression of Bcl‐2 attenuates apoptosis and protects against myocardial I/R injury in transgenic mice. Am J Physiol Heart Circ Physiol 280: H2313‐H2320, 2001.
 93. Chertow GM, Burdick E, Honour M, Bonventre JV, Bates DW. Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. J Am Soc Nephrol 16: 3365‐3370, 2005.
 94. Chertow GM, Christiansen CL, Cleary PD, Munro C, Lazarus JM. Prognostic stratification in critically ill patients with acute renal failure requiring dialysis. Arch Intern Med 155: 1505‐1511, 1995.
 95. Choudhury D, Ahmed Z. Drug‐associated renal dysfunction and injury. Nat Clin Pract Nephrol 2: 80‐91, 2006.
 96. Coca SG, Yalavarthy R, Concato J, Parikh CR. Biomarkers for the diagnosis and risk stratification of acute kidney injury: A systematic review. Kidney Int 73: 1008‐1016, 2008.
 97. Coca SG, Yusuf B, Shlipak MG, Garg AX, Parikh CR. Long‐term risk of mortality and other adverse outcomes after acute kidney injury: A systematic review and meta‐analysis. Am J KidDiseases 53: 961‐973, 2009.
 98. Conesa ELp, Valero F, Nadal JC, Fenoy FJ, LÃ3pez B, Arregui Ba, Salom MGa. N‐acetyl‐l‐cysteine improves renal medullary hypoperfusion in acute renal failure. Am J Physiol Regul Integr Comp Physiol 281: R730‐R737, 2001.
 99. Conger JD, Robinette JB, Guggenheim SJ. Effect of acetylcholine on the early phase of reversible norepinephrine‐induced acute renal failure. Kidney Int 19: 399‐409, 1981.
 100. Conger JD, Robinette JB, Hammond WS. Differences in vascular reactivity in models of ischemic acute renal failure. Kidney Int 39: 1087‐1097, 1991.
 101. Conger JD, Robinette JB, Schrier RW. Smooth muscle calcium and endothelium derived relaxing factor in the abnormal vascular responses of acute renal failure. J Clin Invest 82: 532‐537, 1988.
 102. Conger JD, Weil JV. Abnormal vascular function following ischemia‐reperfusion injury. J Invest Med 43: 431‐442, 1995.
 103. Coopersmith CM, O'Donnell D, Gordon JI. Bcl‐2 inhibits ischemia‐reperfusion‐induced apoptosis in the intestinal epithelium of transgenic mice. Am J Physiol Gastrointest Liver Physiol 276: G677‐G686, 1999.
 104. Cowley AW Jr Role of the renal medulla in volume and arterial pressure regulation. Am J Physiol R1‐15, 1997 .
 105. Crompton M. The mitochondrial permeability transition pore and its role in cell death. Biochem J 341: 233‐249, 1999.
 106. Cruthirds DL, Novak L, Akhi KM, Sanders PW, Thompson JA, MacMillan‐Crow LA. Mitochondrial targets of oxidative stress during renal ischemia/reperfusion. Arch Biochem Biophys 412: 27‐33, 2003.
 107. Cunningham PN, Dyanov HM, Park P, Wang J, Newell KA, Quigg RJ. Acute renal failure in endotoxemia is caused by TNF acting directly on TNF receptor‐1 in kidney. J Immunol 168: 5817‐5823, 2002.
 108. Cunningham PN, Wang Y, Guo R, He G, Quigg RJ. Role of toll‐like receptor 4 in endotoxin‐induced acute renal failure. J Immunol 172: 2629‐2635, 2004.
 109. Cuppage FE, Chiga M, Tate A. Cell cycle studeis in the regenerating rat nephron following injury with mercuric chloride. Lab Invest 26: 122‐126, 1972.
 110. Cuppage FE, Cunningham N, Tate A. Nucleic acid synthesis in the regenerating nephron following injury with mercuric chloride. Lab Invest 21: 449‐457, 1969.
 111. Cuttle L, Zhang X‐J, Endre ZH, Winterford C, Gobe GC. Bcl‐XL translocation in renal tubular epithelial cells in vitro protects distal cells from oxidative stress. Kidney Int 59: 1779‐1788, 2001.
 112. Dagher F, Pollina RM, Rogers DM, Gennaro M, Ascer E. The value and limitations of L‐arginine infusion on glomerular and tubular function in the ischemic/reperfused kidney. J Vasc Surg 21: 453‐458, 1995.
 113. Dagher PC, Basile DP. An expanding role of Toll‐like receptors in sepsis‐induced acute kidney injury. Am J Physiol Renal Physiol 294: F1048‐F1049, 2008.
 114. Dagher PC, Herget‐Rosenthal S, Ruehm SG, Jo S‐K, Star RA, Agarwal R, Molitoris BA. Newly developed techniques to study and diagnose acute renal failure. J Am Soc Nephrol 14: 2188‐2198, 2003.
 115. Dagher Pierre C. Apoptosis in ischemic renal injury: Roles of GTP depletion and p53. Kidney Int 66: 506‐509, 2004.
 116. Dawidson I, Rooth P. Effects of calcium antagonist in ameliorating cyclosporine A nephrotoxicity and post‐transplant (ATN). In: M Epstein, R Loutzenhiser, editors. Calcium antagonists and the kidney. Philadelphia: Hanley & Belfus, Inc, 1990, p. 233‐246.
 117. Day Y‐J, Huang L, Ye H, Li L, Linden J, Okusa MD. Renal ischemia‐reperfusion injury and adenosine 2A receptor‐mediated tissue protection: The role of CD4+ T Cells and IFN‐α. J Immunol 176: 3108‐3114, 2006.
 118. De Greef KE, Ysebaert DK, Dauwe S, Persy V, Vercauteren SR, Mey D, De Broe ME. Anti‐B7‐1 blocks mononuclear cell adherence in vasa recta after ischemia. Kidney Int 60: 1415‐1427, 2001.
 119. de la Coste A, Fabre M, McDonell N, Porteu A, Gilgenkrantz H, Perret C, Kahn A, Mignon A. Differential protective effects of Bcl‐xL and Bcl‐2 on apoptotic liver injury in transgenic mice. Am J Physiol Gastrointest Liver Physiol 277: G702‐G708, 1999.
 120. Dear JW, Yasuda H, Hu X, Hieny S, Yuen PST, Hewitt SM, Sher A, Star RA. Sepsis‐induced organ failure is mediated by different pathways in the kidney and liver: Acute renal failure is dependent on MyD88 but not renal cell apoptosis. Kidney Int 69: 832‐836, 2006.
 121. Denton Mark D, Chertow GM, Brady HR. [ldquo]Renal‐dose[rdquo] dopamine for the treatment of acute renal failure: Scientific rationale, experimental studies and clinical trials. Kidney Int 50: 4‐14, 1996.
 122. Devalaraja‐Narashimha K, Diener AM, Padanilam BJ. Cyclophilin D gene ablation protects mice from ischemic renal injury. Am J Physiol Renal Physiol 297: F749‐F759, 2009.
 123. Devarajan P, Mishra J, Supavekin S, Patterson LT, Steven Potter S. Gene expression in early ischemic renal injury: Clues towards pathogenesis, biomarker discovery, and novel therapeutics. Mol Genet Metabol 80: 365‐376, 2003.
 124. di Mari JF, Davis R, Safirstein RL. MAPK activation determines renal epithelial cell survival during oxidative injury. Am J Physiol 277: F195‐F203, 1999.
 125. DiMari J, Megyesi J, Udvarhelyi N, Price P, Davis R, Safirstein R. N‐acetyl cysteine ameliorates ischemic renal failure. Am J Physiol 272: F292‐F298, 1997.
 126. Djamali A, Sadowski E, Muehrer RJ, reese S, Smavatkul C, Vidyasagar A, Fain SB, Lipscomb RC, Hullett DH, Samaniego‐Picota M, Grist TM, Becker BN. BOLD MRI assessment of intrarenal oxygenation and oxidative stress in patients with chronic kidney alograft dysfunction. Am J Physiol 292: F513‐F522, 2007.
 127. Dobashi K, Ghosh B, Orak JK, Singh I, Singh AK. Kidney ischemia‐reperfusion: Modulation of antioxidant defenses. Mol Cell Biochem 205: 1‐11, 2000.
 128. Doctor R, Mandel L. Minimal role of xanthine oxidase and oxygen free radicals in rat renal tubular reoxygenation injury. J Am Soc Nephrol 1: 959‐969, 1991.
 129. Dong X, Swaminathan S, Bachman LA, Croatt AJ, Nath KA, Griffin MD. Resident dendritic cells are the predominant TNF‐secreting cell in early renal ischemia‐reperfusion injury. Kidney Int 71: 619‐628, 2007.
 130. Dong Z. Response to “Autophagy: A protective mechanism against nephrotoxicant‐induced renal injury”. Kidney Int 75: 119‐119, 2009.
 131. Donnahoo KK, Meng X, Ao L, Ayala A, Shames BD, Cain MP, Harken AH, Meldrum DR. Differential cellular immunolocalization of renal tumour necrosis factor‐α production during ischaemia versus endotoxaemia. Immunology 102: 53‐58, 2001.
 132. Donnahoo KK, Meng X, Ayala A, Cain MP, Harken AH, Meldrum DR. Early kidney TNF–alpha expression mediates neutrophil infiltration and injury after renal ischemia‐reperfusion. Am J Physiol 277: R922‐R929, 1999.
 133. Donohoe JF, Venkatchalam MA, Bernard DB, Levinsky NG. Tubular leakage and obstruction after renal ischemia: Structural‐functional correlations. Kidney Int 13: 208‐222, 1978.
 134. Du C, Guan Q, Diao H, Yin Z, Jevnikar AM. Nitric oxide induces apoptosis in renal tubular epithelial cells through activation of caspase‐8. Am J Physiol Renal Physiol 290: F1044‐F1054, 2006.
 135. Du C, Guan Q, Yin Z, Zhong R, Jevnikar AM. IL‐2‐mediated apoptosis of kidney tubular epithelial cells is regulated by the caspase‐8 inhibitor c‐FLIP. Kidney Int 67: 1397‐1409, 2005.
 136. Duffield JS. Macrophages and Immunologic Inflammation of the Kidney. Semin Nephrol 30: 234‐254, 2010.
 137. Dunnill MS. A review of the pathology and pathogenesis of acute renal failure due to acute tubular necrosis. J Clin Pathol 27: 2‐13, 1974.
 138. Edelstein CL. Biomarkers of acute kidney injury. Adv Chronic Kidney Dis 15: 222‐234, 2008.
 139. Eickelberg O, Seebach FA, Riordan M, Thulin G, Mann A, Reidy K, Van Why SK, Kashgarian M, Siegel NJ. Functional activation of heat shock factor and hypoxia‐inducible factor in the kidney. J Am Soc Nephrol 13: 2094‐2101, 2002.
 140. Emami A, Schwartz JH, Borkan SC. Transient ischemia or heat stress induces a cytoprotectant protein in rat kidney. Am J Physiol 260: F479‐F485, 1991.
 141. Enestrom S, Druid H, Rammer L. Fibrin deposition in the kidney in post‐ischaemic renal damage. Br J Exp Pathol 69: 387‐394, 1988.
 142. Erley CM, Duda SH, Schlepckow S, Koehler J, Huppert PE, Strohmaier WL, Bohle A, Risler T, Osswald H. Adenosine antagonist theophylline prevents the reduction of glomerular filtration rate after contrast media application. Kidney Int 45: 1425‐1431, 1994.
 143. Faubel S, Ljubanovic D, Reznikov L, Somerset H, Dinarello CA, Edelstein CL. Caspase‐1‐deficient mice are protected against cisplatin‐induced apoptosis and acute tubular necrosis. Kidney Int 66: 2202‐2213, 2004.
 144. Feldkamp T, Kribben A, Weinber JM. Assessment of mitochondrial membrane potential in proximal tubules after hypoxia‐reoxygenation injury. Am J Physiol Renal Physiol 288: F1092‐F1102, 2005.
 145. Feldkamp T, Park JS, Pasupulati R, Amora D, Roeser NF, Venkatachalam MA, Weinberg JM. Regulation of the mitochondrial permeability transition in kidney proximal tubules and its alteration during hypoxia‐reoxygenation. Am J Physiol Renal Physiol 297: F1632‐F1646, 2009.
 146. Fernando AR, Armstrong DMG, Griffiths JR, Hendry WF, O'Donoghue EPN, Perrett D, Ward JP, Wickham JEA. Inosine in experimental acute renal failure. Lancet 308: 1302‐1303, 1976.
 147. Finn WF. Recovery from acute renal failure. In: Lazarus JM, Brenner BM, editors. Acute Renal Failure. New York: Churchill Livingstone, 1993, p. 553‐596.
 148. Finn WF, Hak LJ, Grossman SH. Protective effect of prostacyclin on postischemic acute renal failure in the rat. Kidney Int 32: 479‐487, 1987.
 149. Firth JD, Raine AEG, Ratcliffe PJ, Ledingham JGG. Endothelin: An important factor in acute renal failure? Lancet 332: 1179‐1182, 1988.
 150. Fissell WH, Dyke DB, Weitzel WF, Buffington DA, Westover AJ, MacKay SM, Gutierrez JM, Humes HD. Bioartificial kidney alters cytokine response and hemodynamics in endotoxin‐challenged uremic animals. Blood Purif 20(1): 55‐60, 2002.
 151. Floege J, Burns MW, Alpers CE, Yoshimura A, Pritzl P, Gordon K, Seifert RA, Bowen‐Pope DF, Couser WG, Johnson RJ. Glomerular cell proliferation and PDGF expression precede glomerulosclerosis in the remnant kidney model. Kidney Int 41: 297‐309, 1992.
 152. Flores J, DiBona DR, Beck CH, Leaf A. The role of cell swelling in ischemic renal damage and the protective effect of hypertonic solute. J Clin Invest 51: 118‐126, 1972.
 153. Forbes JM, Hewitson TD, Becker GJ, Jones CL. Simultaneous blockade of endothelin A and B receptors in ischemic acute renal failure is detrimental to long‐term kidney function. Kidney Int 59: 1333‐1341, 2001.
 154. Fowler B. Mechanisms of kidney cell injury from metals. Environ Health Perspect 100: 57‐63, 1993.
 155. Friedewald JJ, Rabb H. Inflammatory cells in ischemic acute renal failure. Kidney Int 66: 486‐491, 2004.
 156. Fujii T, Kurata H, Takaoka M, Muraoka T, Fujisawa Y, Shokoji T, Nishiyama A, Abe Y, Matsumura Y. The role of renal sympathetic nervous system in the pathogenesis of ischemic acute renal failure. Eur J Pharmacol 481: 241‐248, 2003.
 157. Fuller TF, Rose F, Singleton KD, Linde Y, Hoff U, Freise CE, Dragun D, Niemann CU. Glutamine donor pretreatment in rat kidney transplants with severe preservation reperfusion injury. J Surg Res 140: 77‐83, 2007.
 158. Fuller TF, Sattler B, Binder L, Vetterlein F, Ringe B, Lorf T. Reduction of severe ischemia/reperfusion injury in rat idney grafts by a soluble P‐Selectin glycprotein ligand. Tranplantation 72: 216‐222, 2001.
 159. Funk JA, Odejinmi S, Schnellmann RG. SRT1720 induces mitochondrial biogenesis and rescues mitochondrial function after oxidant injury in renal proximal tubule cells. J Pharmacol Exp Ther 333: 593‐601.
 160. Furuichi K, Wada T, Iwata Y, Sakai N, Yoshimoto K, Kobayashi K, Mukaida N, Matsushima K, Yokoyama H. Administration of FR167653, a new anti‐inflammatory compound, prevents renal ischaemia/reperfusion injury in mice. Nephrol Dial Transplant 17: 399‐407, 2002.
 161. Garcia‐Criado FJ, Eleno N, Santos‐Benito F, Valdunciel JJ, Reverte M, Lozano‐Sanchez FS, Ludena MD, Gomez‐Alonso A, Lopez‐Novoa JM. Protective effect of exogenous nitric oxide on the renal function and inflammatory response in a model of ischemia‐reperfusion. Transplantation 66: 982‐990, 1998.
 162. Gaudio KM, Ardito TA, Reilly HF, Kashgarian M, Siegel NJ. Accelerated cellular recovery after an ischemic renal injury. Am J Pathol 112: 338‐346, 1983.
 163. Gaudio KM, Taylor MR, Chaudry IH, Kashgarian M, Siegel NJ. Accelerated recovery of single nephron function by the postischemic infusion of ATP‐MgCl2. Kidney Int 22: 13‐20, 1982.
 164. Gaudio KM, Thulin G, Ardito T, Kashgarian M, Siegel NJ. Redistribution of cellular energy following renal ischemia. Pediatr Nephrol 5: 591‐596, 1991.
 165. Gellai M, Jugus M, Fletcher T, DeWolf R, Nambi P. Reversal of postischemic acute renal failure with a selective endothelinA receptor antagonist in the rat. J Clin Invest 93: 900‐906, 1994.
 166. Genescà M, Sola A, Hotter G. Actin cytoskeleton derangement induces apoptosis in renal ischemia/reperfusion. Apoptosis 11: 563‐571, 2006.
 167. Geng H, Lan R, Wang G, Siddiqi AR, Naski MC, Brooks AI, Barnes JL, Saikumar P, Weinberg JM, Venkatachalam MA. Inhibition of autoregulated TGF‐B signaling simultaneously enhances proliferation and differentiation of kidney epithelium and promotes repair following renal ischemia. Am J Pathol 174: 1291‐1308, 2009.
 168. Ghielli M, Verstrepen W, Nouwen E, De Broe ME. Regeneration processes in the kidney after acute injury: Role of infiltrating cells. Exp Nephrol 6: 502‐507, 1998.
 169. Glaumann B, Glaumann H, Berezesky I, Trump BF. Studies on the pathogenesis of ischemic cell injury II. Morphological changes of the pars convolutia (P1 and P2) of the proximal tubule of the rat kidney made ischemic in vivo. Virchows Arch B Cell Pathol 19: 281‐302, 1975.
 170. Glaumann B, Glaumann H, Berezesky I, Trump BF. Studies on the cellular recovery from injury II. Ultrastructural studies on the recovery of the pars convoluta of the proximal tubule of the rate kidney from temporary ischemia. Virchows Arch B Cell Pathol 24: 1‐18, 1977.
 171. Glaumann B, Trump BF. Studies on the pathogenesis of ischemic cell injury. III. Morphological changes of the proximal pars recta tubules (P3) of the rat kidney made ischemic in vivo. Virchows Arch B Cell Pathol 19: 303‐332, 1975.
 172. Gobe G, Zhang XJ, Cuttle L, Pat B, Willgoss D, Hancock J, Barnard R, Endre RB. Bcl‐2 genes and growth factors in the pathology of ischaemic acute renal failure. Immunol Cell Biol 77: 279‐286, 1999.
 173. Goligorsky MS, Lieberthal W, Racusen L, Simon EE. Integrin receptors in renal tubular epithelium: New insights into pathophysiology of acute renal failure. Am J Physiol 264: F1‐F8, 1993.
 174. Gomes M, Cancherini V, Reboucas MA, Reboucas NA. Ischemic preconditioning of renal tissue: Identification of early up‐regulated genes. Nephron Exp Nephrol 93: e107‐e116, 2003.
 175. Gonzalez‐Flecha B, Boveris A. Mitochondrial sites of hydrogen peroxide production in reperfused rat kidney cortex. Biochim Biophys Acta 1243: 361‐366, 1995.
 176. Gopalakrishnan S, Hallett MA, Atkinson SJ, Marrs JA. Differential regulation of junctional complex assembly in renal epithelial cell lines. Am J Physiol Cell Physiol 285: C102‐C111, 2003.
 177. Grams ME, Rabb H. The distant organ effects of acute kidney injury. Kidney Int 2011 Aug 3. doi: 10.1038/ki.2011.241. [Epub ahead of print].
 178. Gunter TE, Pfeiffer DR. Mechanisms by which mitochondria transport calcium. Am J Physiol Cell Physiol 258: C755‐C786, 1990.
 179. Guo R, Wang Y, Minto AW, Quigg RJ, Cunningham PN. Acute renal failure in endotoxemia is dependent on caspase activation. J Am Soc Nephrol 15: 3093‐3102, 2004.
 180. Guo S, Wharton W, Moseley P, Shi H. Heat shock protein 70 regulates cellular redox status by modulating glutathione‐related enzyme activities. Cell Stress Chaperones 12: 245‐254, 2007.
 181. Gupta S, Verfaillie C, Chmielewski D, Kim Y, Rosenberg ME. A role for extrarenal cells in the regeneration following acute renal failure. Kidney Int 62: 1285‐1290, 2002.
 182. Gupta S, Verfaillie C, Chmielewski D, Kren S, Eidman K, Connaire J, Heremans Y, Lund T, Blackstad M, Jiang Y, Luttun A, Rosenberg ME. Isolation and characterization of kidney‐derived stem cells. J Am Soc Nephrol 17: 3028‐3040, 2006.
 183. Haase VH. Hypoxia‐inducible factors in the kidney. Am J Physiol Renal Physiol 291: F271‐F281, 2006.
 184. Hagen TM, Aw TY, Jones DP. Glutathione uptake and protection against oxidative injury in isolated kidney cells. Kidney Int 34: 74‐81, 1988.
 185. Hall AM, Unwin RJ, Parker N, Duchen MR. Multiphoton imaging reveals differences in mitochondrial function between nephron segments. J Am Soc Nephrol 20: 1293‐1302, 2009.
 186. Hallett MA, Dagher PC, Atkinson SJ. Rho GTPases show differential sensitivity to nucleotide triphosphate depletion in a model of ischemic cell injury. Am J Physiol Cell Physiol 285: C129‐C138, 2003.
 187. Hamerski DA, Santoro SA. Integrins and the kidney: Biology and pathobiology. Curr Opin Nephrol Hypertens 8: 9‐14, 1999.
 188. Hammerman MR, Rogers SA, Ryan G. Growth factors and metanephrogenesis. Am J Physiol 262: F523‐F532, 1992.
 189. Hammerschmidt E, Loeffler I, Wolf G. Morg1 heterozygous mice are protected from acute renal ischemia‐reperfusion injury. Am J Physiol Renal Physiol 297: F1273‐F1287, 2009.
 190. Haq M, Norman J, Saba S, Ramirez G, Rabb H. Role of IL‐1 in renal ischemic reperfusion injury. J Am Soc Nephrol 9: 614‐619, 1998.
 191. Harris RC. Growth factors and cytokines in acute renal failure. Adv Ren Replace Ther 4: 43‐53, 1997.
 192. Harris RC, Breyer MD. Physiological regulation of cyclooxygenase‐2 in the kidney. Am J Physiol Renal Physiol 281: F1‐F11, 2001.
 193. Hasegawa K, Wakino S, Yoshioka K, Tatematsu S, Hara Y, Minakuchi H, Sueyasu K, Washida N, Tokuyama H, Tzukerman M, Skorecki K, Hayashi K, Itoh H. Kidney‐specific overexpression of Sirt1 protects against acute kidney injury by retaining peroxisome function. J Biol Chem 285: 13045‐13056, 2010.
 194. Hashimoto‐Uoshima M, Yan Y, Schneider G, Aukhil I. The alternatively spliced domains EIIIB and EIIIA of human fibronectin affect cell adhesion and spreading. J Cell Sci 110: 2271‐2280, 1997.
 195. Haug CE, Colvin RB, Delmonico FL, Auchincloss HJ, Tolkoff‐Rubin N, Preffer FI, Rothlein R, Norris S, Scharschmidt L, Cosimi AB. A phase I trial of immunosuppression with ANTIIC AM‐1 (CD54) mAb in renal allograft recipients. Transplantation 55: 766‐772, 1993.
 196. Hausenloy DJ, Yellon DM. Remote ischaemic preconditioning: Underlying mechanisms and clinical application. Cardiovasc Res 79: 377‐386, 2008.
 197. Havasi A, Li Z, Wang Z, Martin JL, Botla V, Ruchalski K, Schwartz JH, Borkan SC. Hsp27 inhibits Bax activation and apoptosis via a phosphatidylinositol 3‐kinase‐dependent mechanism. J Biol Chem 283: 12305‐12313, 2008.
 198. Havasi A, Wang Z, Gall JM, Spaderna M, Suri V, Canlas E, Martin JL, Schwartz JH, Borkan SC. Hsp27 inhibits sublethal, Src‐mediated renal epithelial cell injury. Am J Physiol Renal Physiol 297: F760‐F768, 2009.
 199. Healy E, Dempsey M, Lally C, Ryan MP. Apoptosis and necrosis: Mechanisms of cell death induced by cyclosporine A in a renal proximal tubular cell line. Kidney Int 54: 1955‐1966, 1998.
 200. Hegarty NJ, Young LS, Kirwan CN, O'Neill AJ, Bouchier‐Hayes DM, Sweeney P, Watson RWG, Fitzpatrick JM. Nitric oxide in unilateral ureteral obstruction: Effect on regional renal blood flow. Kidney Int 59: 1059‐1065, 2001.
 201. Hellberg PO, Kallskog O, Wolgast M. Nephron function in the early phase of ischemic renal failure. Significance of erythrocyte trapping. Kidney Int 38: 432‐439, 1990.
 202. Hellberg POA, Kallskog O, Wolgast M. Red cell trapping and postischemic renal blood flow. Differences between the cortex, outer and inner medulla. Kidney Int 40: 625‐631, 1991.
 203. Hellberg POA, Kallskog OT, Ojteg G, Wolgast M. Peritubular capillary permeability and intravascular RBC aggregation after ischemia: Effects of neutrophils. Am J Physiol 258: F1018‐F1025, 1990.
 204. Herrera MB, Bussolati B, Bruno S, Morando L, Mauriello‐Romanazzi G, Sanavio F, Stamenkovic I, Biancone L, Camussi G. Exogenous mesenchymal stem cells localize to the kidney by means of CD44 following acute tubular injury. Kidney Int 72: 430‐441, 2007.
 205. Hill P, Shukla D, Tran MGB, Aragones J, Cook HT, Carmeliet P, Maxwell PH. Inhibition of hypoxia inducible factor hydroxylases protects against renal ischemia‐reperfusion injury. J Am Soc Nephrol 19: 39‐46, 2008.
 206. Himmelfarb J, Joannidis M, Molitoris B, Schietz M, Okusa MD, Warnock D, Laghi F, Goldstein SL, Prielipp R, Parikh CR, Pannu N, Lobo SM, Shah S, D'Intini V, Kellum JA. Evaluation and initial management of acute kidney injury. Clin J Am Soc Nephrol 3: 962‐967, 2008.
 207. Hirschberg R. Effects of growth hormone and IGF‐I on glomerular ultrafiltration in growth hormone‐deficient rats. Regul Pept 48: 241‐250, 1993.
 208. Hirschberg R, Kopple J, Lipsett P, Benjamin E, Minei J, Albertson T, Munger M, Metzler M, Zaloga G, Murray M, Lowry S, Conger J, McKeown W, O'Shea M, Baughman R, Wood K, Haupt M, Kaiser R, Simms H, Warnock D, Summer W, Hintz R, Myers B, Haenftling K, Capra W, Pike M, Guler H‐P. Multicenter clinical trial of recombinant human insulin‐like growth factor I in patients with acute renal failure. Kidney Int 55: 2423‐2432, 1999.
 209. Hohenstein B, Kuo M‐C, Addabbo F, Yasuda K, Ratliff B, Schwarzenberger C, Eckardt K‐U, Hugo CPM, Goligorsky MS. Enhanced progenitor cell recruitment and endothelial repair after selective endothelial injury of the mouse kidney. Am J Physiol Renal Physiol 298: F1504‐F1514, 2010.
 210. Hollenberg NK, Epstein FH, Rosen S, Basch R, Oken D, Merrill JP. Acute oliguric renal failure in man: Evidence for preferential renal cortical ischemia. Medicne (Baltimore) 47: 455‐474, 1968.
 211. Homma‐Takeda S, Takenaka Y, Kumagai Y, Shimojo N. Selective induction of apoptosis of renal proximal tubular cells caused by inorganic mercury in vivo. Environ Toxicol Pharmacol 7: 179‐187, 1999.
 212. Horbelt M, Lee S‐Y, Mang HE, Knipe NL, Sado Y, Kribben A, Sutton TA. Acute and chronic microvascular alterations in a mouse model of ischemic acute kidney injury. Am J Physiol Renal Physiol 293: F688‐F695, 2007.
 213. Hou SH, Bushinsky DA, Wish JB, Cohen JJ, Harrington JT. Hospital‐acquired renal insufficiency: A prospective study. Am J Med 74: 243‐248, 1983.
 214. Hsu CY, McCulloch CE, Fan D, Ordonez JD, Chertow GM, Go AS. Community‐based incidence of acute renal failure. Kidney Int 72: 208‐212, 2007.
 215. Huang C, Huang C, Hestin D, Dent PC, Barclay P, Collis M, Johns EJ. The effect of endothelin antagonists on renal ischaemia reperfusion injury and the development of acute renal failure in the rat. Nephrol Dial Transplant 17: 1578‐1585, 2002.
 216. Huang H, He Z, Roberts LJ, Salahudeen AK. Deferoxamine reduces cold‐ischemic renal injury in a syngeneic kidney transplant model. Am J Transplant 3: 1531‐1537, 2003.
 217. Humes D, Cieslinski DA, Coimbra TM, Messana JM, Galvao C. Epidermal growth factor enhaces renal tubule cell regeneration and repair and accelerates the recovery of renal function in postischemic acute renal failure. J Clin Invest 84: 1757‐1761, 1989.
 218. Humphreys BD, Lin S‐L, Kobayashi A, Hudson TE, Nowlin BT, Bonventre JV, Valerius MT, McMahon AP, Duffield JS. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol 176: 85‐97, 2010.
 219. Humphreys BD, Valerius MT, Kobayashi A, Mugford JW, Soeung S, Duffield JS, McMahon AP, Bonventre JV. Intrinsic epithelial cells repair the kidney after injury. Cell Stem Cell 2: 284‐291, 2008.
 220. Hynninen MS, Niemi TT, Poyhia R, Raininko EI, Salmenperä MT, Lepantalo MJ, Railo MJ, Tallgren MK. N‐acetylcysteine for the prevention of kidney injury in abdominal aortic surgery: A randomized, double‐blind, placebo‐controlled trial. Anesth Analg 102: 1638‐1645, 2006.
 221. Ichikawa I, Kiyama S, Yoshioka T. Renal antioxidant enzymes: Their regulation and function. Kidney Int 45: 1‐9, 1994.
 222. Ichikawa I, Purkerson ML, Yates J, Klahr S. Dietary protein intake conditions the degree of renal vasoconstriction in acute renal failure caused by ureteral obstruction. Am J Physiol 249: F54‐F61, 1985.
 223. Igawa T, Matsumoto K, Kanda S, Saito Y, Nakamura T. Hepatocyte growth factor may function as a renotropic factor for regeneration in rats with acute renal injury. Am J Physiol 265: F61‐F69, 1993.
 224. Ilker K, Ilker T, Nigar Y, Recep S, Nermin K, Ahmet O. Iloprost downregulates expression of adhesion molecules and reduces renal injury induced by abdominal aortic ischemia‐reperfusion. Ann Vasc Surg 23: 212‐223, 2009.
 225. Imgrund M, Grone E, Grone H‐J, Kretzler M, Holzman L, Schlondorff D, Rothenpieler UW. Re‐expression of the developmental gene Pax‐2 during experimental acute tubular necrosis in mice1. Kidney Int 56: 1423‐1431, 1999.
 226. Inscho EW, Imig JD, Cook AK, Pollock DM. ETA and ETB receptors differentially modulate afferent and efferent arteriolar responses to endothelin. Br J Pharmacol 146: 1019‐1026, 2005.
 227. Ishani A, Xue JL, Himmelfarb J, Eggers PW, Kimmel PL, Molitoris BA, Collins AJ. Acute kidney injury increases risk of ESRD among elderly. J Am Soc Nephrol 20: 223‐228, 2009.
 228. Ishizuka S, Yano T, Hagiwara K, Sone M, Nihei H, Ozasa H, Horikawa S. Extracellular signal‐regulated kinase mediates renal regeneration in rats with myoglobinuric acute renal injury. Biochem Biophys Res Commun 254: 88‐92, 1999.
 229. Iwano M, Plieth D, Danoff TM, Xue C, Okada H, Neilson EG. Evidence that fibroblasts derive from epithelium during tissue fibrosis. J Clin Invest 110: 341‐350, 2002.
 230. Iwasaki M, Adachi Y, Minamino K, Suzuki Y, Zhang Y, Okigaki M, Nakano K, Koike Y, Wang J, Mukaide H, Taketani S, Mori Y, Takahashi H, Iwasaka T, Ikehara S. Mobilization of bone marrow cells by G‐CSF rescues mice from cisplatin‐induced renal failure, and M‐CSF enhances the effects of G‐CSF. J Am Soc Nephrol 16: 658‐666, 2005.
 231. Jang H‐S, Kim J, Park Y‐K, Park KM. Infiltrated macrophages contribute to recovery after ischemic injury but not to ischemic preconditioning in kidneys. Transplantation 85: 447‐455, 2008.
 232. Janin A, Deschaumes C, Daneshpouy M, Estaquier J, Micic‐Polianski J, Rajagopalan‐Levasseur P, Akarid K, Mounier N, Gluckman E, Socie G, Ameisen JC. CD95 engagement induces disseminated endothelial cell apoptosis in vivo: Immunopathologic implications. Blood 99: 2940‐2947, 2002.
 233. Jefayri MK, Grace PA, Mathie RT. Attenuation of reperfusion injury by renal ischaemic preconditioning: The role of nitric oxide. BJU Int 85: 1007‐1013, 2000.
 234. Jensen AM, Norregaard R, Topcu SO, Frokiaer J, Pedersen M. Oxygen tension correlates with regional blood flow in obstructed rat kidney. J Exp Biol 212: 3156‐3163, 2009.
 235. Jiang M, Pabla N, Murphy RF, Yang T, Yin X‐M, Degenhardt K, White E, Dong Z. Nutlin‐3 protects kidney cells during cisplatin therapy by suppressing Bax/Bak activation. J Biol Chem 282: 2636‐2645, 2007.
 236. Jo S‐K, Cho WY, Sung SA, Kim HK, Won NH. MEK inhibitor, U0126, attenuates cisplatin‐induced renal injury by decreasing inflammation and apoptosis. Kidney Int 67: 458‐466, 2005.
 237. Jo SK, Rosner MH, Okusa MD. Pharmacologic treatment of acute kidney injury: Why drugs haven't worked and what is on the horizon. Clin J Am Soc Nephrol 2: 356‐365, 2007.
 238. Johannes T, Ince C, Klingel K, Unertl KE, Mik EG. Iloprost preserves renal oxygenation and restores kidney function in endotoxemia‐related acute renal failure in the rat. Crit Care Med 37: 1423‐1432, 2009.
 239. Johnson RJ, Schreiner GF. Hypothesis: The role of acquired tubulointerstitial disease in the pathogenesis of salt‐dependent hypertension. Kidney Int 52: 1169‐1179, 1997.
 240. Joyce DE, Gelbert L, Ciaccia A, DeHoff B, Grinnell BW. Gene expression profile of antithrombotic protein C defines new mechanisms modulating inflammation and apoptosis. J Biol Chem 276: 11199‐11203, 2001.
 241. Jung KY, Uchida S, Endou H. Nephrotoxicity assessment by measuring cellular ATP content: I. Substrate specificities in the maintenance of ATP content in isolated rat nephron segments. Toxicol Appl Pharmacol 100: 369‐382, 1989.
 242. Jung YJ, Kim DH, Lee AS, Lee S, Kang KP, Lee SY, Jang KY, Sung MJ, Park SK, Kim W. Peritubular capillary preservation with COMP‐angiopoietin‐1 decreases ischemia‐reperfusion‐induced acute kidney injury. Am J Physiol Renal Physiol 297: F952‐F960, 2009.
 243. Kaizu T, Tamaki T, Tanaka M, Uchida Y, Tsuchihashi S‐I, Kawamura A, Kakita A. Preconditioning with tin‐protoporphyrin IX attenuates ischemia/reperfusion injury in the rat kidney. Kidney Int 63: 1393‐1403, 2003.
 244. Kakoki M, Hirata Y, Hayakawa H, Suzuki E, Nagata D, Tojo A, Nishimatsu H, Nakanishi N, Hattori Y, Kikuchi K, Nagano T, Omata M. Effects of tetrahydrobiopterin on endothelial dysfunction in rats with ischemic acute renal failure. J Am Soc Nephrol 11: 301‐309, 2000.
 245. Kalluri R, Neilson EG. Epithelial‐mesenchymal transition and its implications for fibrosis. J Clin Invest 112: 1776‐1784, 2003.
 246. Kanellis J, Paizis K, Cox AJ, Stacker SA, Gilbert RE, Cooper ME, Power DA. Renal ischemia‐reperfusion increases endothelial VEGFR‐2 without increasing VEGF or VEGFR‐1 expression. Kidney Int 61: 1696‐1706, 2002.
 247. Kang DH, Anderson S, Kim YG, Mazzalli M, Suga S, Jefferson JA, Gordon KL, Oyama TT, Hughes J, Hugo C, Kerjaschki D, Schreiner GF, Johnson RJ. Impaired angiogenesis in the aging kidney: Vascular endothelial growth factor and thrombospondin‐1 in renal disease. Am J Kidney Dis 37: 601‐611, 2001.
 248. Kang DH, Hughes J, Mazzali M, Schreiner GF, Johnson RJ. Impaired angiogenesis in the remnant kidney model: II. Vascular endothelial growth factor administration reduces renal fibrosis and stabilizes renal function. J Am Soc Nephrol 12: 1448‐1457, 2001.
 249. Kang DH, Kanellis J, Hugo C, Truong L, Anderson S, Kerjaschki D, Schreiner GF, Johnson R. Role of the microvascular endothelium in progressive renal disease. J Am Soc Nephrol 13: 806‐816, 2002.
 250. Karlberg L, Kallskog O, Norlen BJ, Wolgast M. Postischemic renal failure. Intrarenal blood flow and functional characteristics in the recovery phase. Acta Physiol Scand 115: 1‐10, 1982.
 251. Karlberg L, Norlen BJ, Ojteg G, Wolgast M. Impaired medullary circulation in postischemic acute renal failure. Acta Physiol Scand 118: 11‐17, 1983.
 252. Kaushal GP, Kaushal V, Hong X, Shah SV. Role and regulation of activation of caspases in cisplatin‐induced injury to renal tubular epithelial cells. Kidney Int 60: 1726‐1736, 2001.
 253. Kellerman PS, Bogusky RT. Microfilament disruption occurs very early in ischemic proximal tubule cell injury. Kidney Int 42: 896‐902, 1992.
 254. Kellum JA. Systematic review: The use of diuretics and dopamine in acute renal failure: A systematic review of the evidence. Crit Care 1: 53‐59, 1997.
 255. Kellum JA, M Decker J. Use of dopamine in acute renal faiulre: A meta analysis. Crit Care Med 29: 1526‐1531, 2001.
 256. Kelly KJ. Distant effects of experimental renal ischemia/reperfusion injury. J Am Soc Nephrol 14: 1549‐1558, 2003.
 257. Kelly KJ, Baird NR, Greene AL. Induction of stress response proteins and experimental renal ischemia/reperfusion. Kidney Int 59: 1798‐1802, 2001.
 258. Kelly KJ, Plotkin Z, Dagher PC. Guanosine supplementation reduces apoptosis and protects renal function in the setting of ischemic injury. J Clin Invest 108: 1291‐1298, 2001.
 259. Kelly KJ, Sutton TA, Weathered N, Ray N, Caldwell EJ, Plotkin Z, Dagher PC. Minocycline inhibits apoptosis and inflammation in a rat model of ischemic renal injury. Am J Physiol 287: F760‐F766, 2004.
 260. Kelly KJ, Williams W, Colvin R, Bonventre JV. Antibody to intercellular adhesion molecule 1 protects the kidney against ischemic injury. Proc Natl Acad Sci U S A 18: 812‐816, 1994.
 261. Kelly KJ, Williams WW Jr, Colvin RB, Bonventre JV. Antibody to intercellular adhesion molecule 1 protects the kidney against ischemic injury. Proc Natl Acad Sci U S A 91: 812‐816, 1994.
 262. Kelly KJ, Williams WW Jr, Colvin RB, Meehan SM, Springer TA, Gutierrez‐Ramos JC, Bonventre JV. Intercellular adhesion molecule‐1‐deficient mice are protected against ischemic renal injury. J Clin Invest 97: 1056‐1063, 1996.
 263. Kerr JFR, Wyllie AH, Currie AR. Apoptosis: A basic biological phenomenon with wide‐ranging implications in tissue kinetics. Br J Cancer 26: 239‐257, 1972.
 264. Kerr LE, Birse‐Archbold JL, Short DM, McGregor AL, Heron I, MacDonald DC, Thompson J, Carlson GJ, Kelly JS, McCulloch J, Sharkey J. Nucleophosmin is a novel Bax chaperone that regulates apoptotic cell death. Oncogene 26: 2554‐2562, 2006.
 265. Kessler RH. Effects of ischemia on the concentration of adenine nucleotides in the kidney of anesthetized dogs. Proc Soc Exp Biol Med 134: 1091‐1095, 1970.
 266. Kielar ML, John R, Bennett M, Richardson JA, Shelton JM, Chen L, Jeyarajah DR, Zhou XJ, Zhou H, Chiquett B, Nagami GT, Lu CY. Maladaptive role of IL‐6 in ischemic acute renal failure. J Am Soc Nephrol 16: 3315‐3325, 2005.
 267. Kim J‐S, He L, Lemasters JJ. Mitochondrial permeability transition: A common pathway to necrosis and apoptosis. Biochem Biophys Res Commun 304: 463‐470, 2003.
 268. Kim J‐S, Jin Y, Lemasters JJ. Reactive oxygen species, but not Ca2+ overloading, trigger pH‐ and mitochondrial permeability transition‐dependent death of adult rat myocytes after ischemia‐reperfusion. Am J Physiol Heart Circ Physiol 290: H2024‐H2034, 2006.
 269. Kim K, Lee K, Han D, Yu E, Cho Y. Adult stem cell like tubular cells reside in the corticomedullary junction of the kidney. Int J Clin Exp Pathol 1: 232‐241, 2007.
 270. Kim M, Park SW, Kim M, Chen SWC, Gerthoffer WT, D'Agati VD, Lee HT. Selective renal overexpression of human heat shock protein 27 reduces renal ischemia‐reperfusion injury in mice. Am J Physiol Renal Physiol 299: F347‐F358, 2010.
 271. Kim YK, Kim HJ, Kwon CH, Kim JH, Woo JS, Jung JS, Kim JM. Role of ERK activation in cisplatin‐induced apoptosis in OK renal epithelial cells. J Appl Toxicol 25: 374‐382, 2005.
 272. Kinsey GR, Li L, Okusa MD. Inflammation in acute kidney injury. Nephron Exp Nephrol 109: e102‐e107, 2008.
 273. Kinsey GR, McHowat J, Patrick KS, Schnellmann RG. Role of Ca2+‐independent phospholipase A2gamma in Ca2+‐induced mitochondrial permeability transition. J Pharmacol Exp Ther 321: 707‐715, 2007.
 274. Kinsey GR, Sharma R, Huang L, Li L, Vergis AL, Ye H, Ju S‐T, Okusa MD. Regulatory T cells suppress innate immunity in kidney ischemia‐reperfusion injury. J Am Soc Nephrol 20: 1744‐1753, 2009.
 275. Kjellstrand CM, Ebben J, Davin T. Time of death, recovery of renal function, development of chronic renal failure and need for chronic hemodialysis in patients with acute tubular necrosis. Trans Am Soc Artif Intern Organs 27: 45‐51, 1981.
 276. Klingebiel T, von Gise H, Bohle A. Morphometric studies on acute renal failure in humans during the oligoanuric and polyuric phases. Clin Nephrol 20: 1‐10, 1983.
 277. Knoll T, Schult S, Birck R, Braun C, Michel MS, Bross S, Juenemann KP, Kirchengast M, Rohmeiss P. Therapeutic administration of an endothelin‐A receptor antagonist after acute ischemic renal failure dose‐dependently improves recovery of renal function. J Cardiovasc Pharmacol 37: 483‐488, 2001.
 278. Konya L, Szenasi G, Bencsath P, Feher J. Study of the effect of superoxide dismutase on acute renal failure in dogs. Acta Med Hung 48: 79‐85, 1991.
 279. Kramer AA, Postler G, Salhab KF, Mendez C, Carey LC, Rabb H. Renal ischemia/reperfusion leads to macrophage‐mediated increase in pulmonary vascular permeability. Kidney Int 55: 2362‐2367, 1999.
 280. Krüger B, Krick S, Dhillon N, Lerner SM, Ames S, Bromberg JS, Lin M, Walsh L, Vella J, Fischereder M, Krämer BK, Colvin RB, Heeger PS, Murphy BT, Schröppel B. Donor toll‐like receptor 4 contributes to ischemia and reperfusion injury following human kidney transplantation. Proc Natl Acad Sci U S A 106: 3390‐3395, 2009.
 281. Kruidering M, Van De Water B, De Heer E, Mulder GJ, Nagelkerke JF. Cisplatin‐induced nephrotoxicity in porcine proximal tubular cells: Mitochondrial dysfunction by inhibition of complexes I to IV of the respiratory chain. J Pharmacol Exp Ther 280: 638‐649, 1997.
 282. Kuida K, Haydar T, Kuan C, Gu Y, Taya C, Karasuyama H, Su M, Rakic P, Flavell R. Reduced apoptosis and cytochrome c‐mediated caspase activation in mice lacking caspase 9. Cell 94: 325‐337, 1998.
 283. Kwon O, Hong S‐M, Sutton TA, Temm CJ. Preservation of peritubular capillary endothelial integrity and increasing pericytes may be critical to recovery from postischemic acute kidney injury. Am J Physiol Renal Physiol 295: F351‐F359, 2008.
 284. Kwon O, Nelson WJ, Sibley R, Huie P, Scandling JD, Dafoe D, Alfrey E, Myers BD. Backleak, tight junctions, and cell‐ cell adhesion in postischemic injury to the renal allograft. J Clin Invest 101: 2054‐2064, 1998.
 285. Kwon O, Phillips CL, Molitoris BA. Ischemia induces alterations in actin filaments in renal vascular smooth muscle cells. Am J Physiol Renal Physiol 282: F1012‐F1019, 2002.
 286. Lau AH. Apoptosis induced by cisplatin nephrotoxic injury. Kidney Int 56: 1295‐1298, 1999.
 287. Laubach VE, French BA, Okusa MD. Targeting of adenosine receptors in ischemia reperfusion injury. Expert Opin Ther Targets 15: 103‐118, 2011.
 288. Lauschke A, Teichgraber UKM, Frei U, Eckardt KU. “Low‐dose' dopamine worsens renal perfusion in patients with acute renal failure. Kidney Int 69: 1669‐1674, 2006.
 289. Leach M, Frank S, Olbrich A, Pfeilschifter J, Thiemermann C. Decline in the expression of copper/zinc superoxide dismutase in the kidney of rats with endotoxic shock: Effects of the superoxide anion radical scavenger, tempol, on organ injury. Br J Pharmacol 125: 817‐825, 1998.
 290. Lee DBN, Huang E, Ward HJ. Tight junction biology and kidney dysfunction. Am J Physiol Renal Physiol 290: F20‐F34, 2006.
 291. Lee HT, Emala CW. Protective effects of renal ischemic preconditioning and adenosine pretreatment: Role of A(1) and A(3) receptors. Am J Physiol 278: F380‐F387, 2000.
 292. Lee HT, Park SW, Kim M, D'Agati VD. Acute kidney injury after hepatic ischemia and reperfusion injury in mice. Lab Invest 89: 196‐208, 2008.
 293. Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL, Semprun‐Prieto L, Delafontaine P, Prockop DJ. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti‐inflammatory protein TSG‐6. Cell Stem Cell 5: 54‐63, 2009.
 294. Lee W‐K, Bork U, Gholamrezaei F, Thevenod F. Cd2+‐induced cytochrome c release in apoptotic proximal tubule cells: Role of mitochondrial permeability transition pore and Ca2+ uniporter. Am J Physiol Renal Physiol 288: F27‐F39, 2005.
 295. Leemans JC, Stokman G, Claessen N, Rouschop KM, Teske GJD, Kirschning CJ, Akira S, van der Poll T, Weening JJ, Florquin S. Renal‐associated TLR2 mediates ischemia/reperfusion injury in the kidney. J Clin Invest 115: 2894‐2903, 2005.
 296. Lei K, Davis RJ. JNK phosphorylation of Bim‐related members of the Bcl2 family induces Bax‐dependent apoptosis. Proc Natl Acad Sci U S A 100: 2432‐2437, 2003.
 297. Lemasters J, Qian T, He L, Kim J, Elmore S, Cascio W, Brenner D. Role of mitochondrial inner membrane permeabilization in necrotic cell death, apoptosis, and autophagy. Antioxid Redox Signal 4: 769‐781, 2002.
 298. Lemasters JJ. Mechanisms of hepatic toxicity V: Necroapoptosis and the mitochondrial permeability transion: Shared pathways to necrosis and apoptosis. Am J Physiol 276: G1‐G6, 1999.
 299. Lemasters JJ, Nieminen A‐L, Qian T, Trost LC, Elmore SP, Nishimura Y, Crowe RA, Cascio WE, Bradham CA, Brenner DA, Herman B. The mitochondrial permeability transition in cell death: A common mechanism in necrosis, apoptosis and autophagy. Biochim Biophys Acta 1366: 177‐196, 1998.
 300. Lemasters JJ, Theruvath TP, Zhong Z, Nieminen A‐L. Mitochondrial calcium and the permeability transition in cell death. Biochim Biophys Acta 1787: 1395‐1401, 2009.
 301. Lemos FBC, Ijzermans JNM, Zondervan PE, Peeters AMA, van den Engel S, Mol WM, Weimar W, Baan CC. Differential expression of heme oxygenase‐1 and vascular endothelial growth factor in cadaveric and living donor kidneys after ischemia‐reperfusion. J Am Soc Nephrol 14: 3278‐3287, 2003.
 302. Leonard EC, Friderich J, Basile DP. VEGF‐121 preserves renal microvessel structure and ameliorates secondary renal disease following acute kidney injury. Am J Physiol Renal Physiol 295: F1648‐F1657, 2008.
 303. Levy EM, Viscoli CM, Horwitz RI. The effect of acute renal failure on mortality. A cohort analysis. JAMA 275: 1489‐1494, 1996.
 304. Lewers D, Mathew TH, Maher JF, Schreiner G. Long‐term follow‐up of renal function and histology after acute tubular necrosis. Ann Int Med 73: 523‐529, 1970.
 305. Lewis J, Salem M, Chertow GM, Weisberg L, McGrew F, Marbury T, Allgren R. Atrial natriuretic factor in oliguric acute renal failure. Am J Kidney Dis 36: 767‐774, 2000.
 306. Lewy PR, Quintanilla A, Levin NW, Kessler RH. Renal energy metabolism and sodium reabsorption. Ann Rev Med 24: 365‐384, 1973.
 307. Li B, Cohen A, Hudson TE, Motlagh D, Amrani DL, Duffield JS. Mobilized human hematopoietic stem/progenitor cells promote kidney repair after ischemia/reperfusion injury. Circulation 121: 2211‐2220, 2010.
 308. Li F, Mao HP, Ruchalski KL, Wang YH, Choy W, Schwartz JH, Borkan SC. Heat stress prevents mitochondrial injury in ATP‐depleted renal epithelial cells. Am J Physiol Cell Physiol 283: C917‐C926, 2002.
 309. Li H‐F, Cheng C‐F, Liao W‐J, Lin H, Yang R‐B. ATF3‐mediated epigenetic regulation protects against acute kidney injury. J Am Soc Nephrol 21: 1003‐1013, 2010.
 310. Li K, Li Y, Shelton J, Richarsdson J, Spencer E, Chen Z, Wang X, Williams R. Cytochrome c deficiency causes embryonic lethality and attenuates stress induced apoptosis. Cell 101: 389‐399, 2000.
 311. Li L, Huang L, Sung S‐sJ, Lobo PI, Brown MG, Gregg RK, Engelhard VH, Okusa MD. NKT cell activation mediates neutrophil IFN‐gamma production and renal ischemia‐reperfusion injury. J Immunol 178: 5899‐5911, 2007.
 312. Li L, Huang L, Sung S‐SJ, Vergis AL, Rosin DL, Rose CE Jr, Lobo PI, Okusa MD. The chemokine receptors CCR2 and CX3CR1 mediate monocyte//macrophage trafficking in kidney ischemia‐reperfusion injury. Kidney Int 74: 1526‐1537, 2008.
 313. Li LY, Luo X, Wang X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature 412: 95‐99, 2001.
 314. Liang H, Arsenault J, Mortensen J, Park F, Johnson C, Nilakantan V. Partial attenuation of cytotoxicity and apoptosis by SOD1 in ischemic renal epithelial cells. Apoptosis 14: 1176‐1189, 2009.
 315. Liangos O, Tighiouart H, Perianayagam MC, Kolyada A, Han WK, Wald R, Bonventre JV, Jaber BL. Comparative analysis of urinary biomarkers for early detection of acute kidney injury following cardiopulmonary bypass. Biomarkers 14: 423‐431, 2009.
 316. Liangos O, Wald R, O'Bell JW, Price L, Pereira BJ, Jaber BL. Epidemiology and outcomes of acute renal failure in hospitalized patients: A national survey. Clin J Am Soc Nephrol 1: 43‐51, 2006.
 317. Liano F, Junco E, Pascual J, Madero R, Verde E. The spectrum of acute renal failure in the intensive care unit compared with that seen in other settings. The Madrid Acute Renal Failure Study Group. Kidney Int Suppl 66: S16‐S24, 1998.
 318. Liano F, Pascual J. Epidemiology of acute renal failure: A prospective, multicenter, community‐based study. The Madrid Acute Renal Failure Study Group. Kidney Int 50: 811‐818, 1996.
 319. Lieberthal W, Koh JS, Levine JS. Necrosis and apoptosis in acute renal failure. Semin Nephrol 18: 505‐518, 1998.
 320. Lieberthal W, Levine JS. Mechanisms of apoptosis and its potential role in renal tubular epithelial cell injury. Am J Physiol 271: F477‐F488, 1996.
 321. Lieberthal W, Menza SA, Levine JS. Graded ATP depletion can cause necrosis or apoptosis of cultured mouse proximal tubular cells. Am J Physiol 274: F315‐F327, 1998.
 322. Lieberthal W, Nigam SK. Acute renal failure. I. Relative importance of proximal vs. distal tubular injury. Am J Physiol 275: F623‐F631, 1998.
 323. Lieberthal W, Nigam SK. Acute renal failure. II. Experimental models of acute renal failure: Imperfect but indispensable. Am J Physiol Renal Physiol 278: F1‐F12, 2000.
 324. Lin F, Cordes K, Li L, Hood L, Couser WG, Shankland SJ, Igarashi P. Hematopoietic stem cells contribute to the regeneration of renal tubules after renal ischemia‐reperfusion injury in mice. J Am Soc Nephrol 14: 1188‐1199, 2003.
 325. Lin F, Moran A, Igarashi P. Intrarenal cells, not bone marrow‐derived cells are the major source of regeneration of the post‐ischemic kidney. J Clin Invest 115: 1756‐1764, 2005.
 326. Lin J‐J, Churchill PC, Bidani AK. Theophylline in rats during maintenance phase of post‐ischemic acute renal failure. Kidney Int 33: 24‐28, 1988.
 327. Lin S‐L, Li B, Rao S, Yeo E‐J, Hudson TE, Nowlin BT, Pei H, Chen L, Zheng JJ, Carroll TJ, Pollard JW, McMahon AP, Lang RA, Duffield JS. Macrophage Wnt7b is critical for kidney repair and regeneration. Proc Natl Acad Sci U S A 107: 4194‐4199, 2010.
 328. Lin S, Chang F, Schrimpf C, Chen Y, Wu C, Wu V, Chiang W, Kuhnert F, Kuo C, Chen Y, Wu K, Tsai T, duffield J. Targetting endothelium pericyte cross talk by inhibiting VEGF receptor signaling attenuates kidney microvascular rarefaction and fibrosis. Am J Pathol 178: 911‐923, 2011.
 329. Linas S, Whittenburg D, Repine JE. Nitric oxide prevents neutrophil‐mediated acute renal failure. Am J Physiol 272: F48‐F54, 1997.
 330. Linas SL, Whittenburg D, Parsons PE, Repine JE. Ischemia increases neutrophil retention and worsens acute renal failure: Role of oxygen metabolites and ICAM 1. Kidney Int 48: 1584‐1591, 1995.
 331. Ling W, Zhaohui N, Ben H, Leyi G, Jianping L, Huili D, Jiaqi Q. Urinary IL‐18 and NGAL as early predictive biomarkers in contrast‐induced nephropathy after coronary angiography. Nephron Clin Pract 108: c176‐c181, 2008.
 332. Liu J‐P, Baker J, Perkins AS, Robertson EJ, Efstratiadis A. Mice carrying null mutations of the genes encoding insulin‐like growth factor I (Igf‐1) and type 1 IGF receptor (Igf1r). Cell 75: 59‐72, 1993.
 333. Liu L, Yang C, Herzog C, Seth R, Kaushal G. Proteosome inhibitors prevent cisplatin induced mitochondrial release of apoptosis inducing factor and markedly ameliorate cisplatin nephrotoxicity. Biochem Pharmacol 79: 137‐146, 2010.
 334. Liu M, Grigoryev DN, Crow MT, Haas M, Yamamoto M, Reddy SP, Rabb H. Transcription factor Nrf2 is protective during ischemic and nephrotoxic acute kidney injury in mice. Kidney Int 76: 277‐285, 2009.
 335. Liu M, Liang Y, Chigurupati S, Lathia JD, Pletnikov M, Sun Z, Crow M, Ross CA, Mattson MP, Rabb H. Acute kidney injury leads to inflammation and functional changes in the brain. J Am Soc Nephrol 19: 1360‐1370, 2008.
 336. Lo LJ, Go AS, Chertow GM, McCulloch CE, Fan D, Ordonez JD, Hsu CY. Dialysis‐requiring acute renal failure increases the risk of progressive chronic kidney disease. Kidney Int 76: 893‐899, 2009.
 337. Lopez‐Novoa JM. Potential role of platelet activating factor in acute renal failure. Kidney Int 55: 1672‐1682, 1999.
 338. Lopez‐Novoa JM, Quiros Y, Vicente L, Morales AI, Lopez‐Hernandez FJ. New insights into the mechanism of aminoglycoside nephrotoxicity: An integrative point of view. Kidney Int 79: 33‐45.
 339. Loukogeorgakis SP, Williams R, Panagiotidou AT, Kolvekar SK, Donald A, Cole TJ, Yellon DM, Deanfield JE, MacAllister RJ. Transient limb ischemia induces remote preconditioning and remote postconditioning in humans by a KATP channel dependent mechanism. Circulation 116: 1386‐1395, 2007.
 340. Lovis C, Mach F, Donati YR, Bonventre JV, Polla BS. Heat shock proteins and the kidney. Ren Fail 16: 179‐192, 1994.
 341. Ma D, Lim T, Xu J, Tang H, Wan Y, Zhao H, Hossain M, Maxwell PH, Maze M. Xenon preconditioning protects against renal ischemic‐reperfusion injury via HIF‐1alpha activation. J Am Soc Nephrol 20: 713‐720, 2009.
 342. Ma H, Saenko M, Opuko A, Togawa A, Soda K, Marlier A, Moeckel GW, Cantley LG, Ishibe S. Deletion of the Met receptor in the collecting duct decreases renal repair following ureteral obstruction. Kidney Int 76: 868‐876, 2009.
 343. MacNider W. The functional and pathological response of the kidney in dogs subjected to a second subcutaneous injection of uranium nitrate. J Exp Med 49: 411‐433, 1929.
 344. Madesh M, Hajnoczky G. VDAC‐dependent permeabilization of the outer mitochondrial membrane by superoxide induces rapid and massive cytochrome c release. J Cell Biol 155: 1003‐1016, 2001.
 345. Maeshima A. Label‐retaining cells in the kidney: Origin of regenerating cells after renal ischemia. Clin Exp Nephrol 11: 269‐274, 2007.
 346. Maeshima A, Yamashita S, Nojima Y. Identification of renal progenitor‐like tubular cells that participate in the regeneration processes of the kidney. J Am Soc Nephrol 14: 3138‐3146, 2003.
 347. Maines MD, Mayer RD, Ewing JF, McCoubrey WK. Induction of kidney heme oxygenase‐1 (HSP32) mRNA and protein by ischemia/reperfusion: Possible role of heme as both promotor of tissue damage and regulator of HSP32. J Pharmacol Exp Ther 264: 457‐462, 1993.
 348. Mandel L, Doctor R, Bacallao R. ATP depletion: A novel method to study junctional properties in epithelial tissues. II. Internalization of Na+,K(+)‐ATPase and E‐cadherin. J Cell Sci 107: 3315‐3324, 1994.
 349. Manley GT, Binder DK, Papadopoulos MC, Verkman AS. New insights into water transport and edema in the central nervous system from phenotype analysis of aquaporin‐4 null mice. Neuroscience 129: 981‐989, 2004.
 350. Mao H, Wang Y, Li Z, Ruchalski KL, Yu X, Schwartz JH, Borkan SC. Hsp72 interacts with paxillin and facilitates the reassembly of focal adhesions during recovery from ATP depletion. J Biol Chem 279: 15472‐15480, 2004.
 351. Martinez‐Salgado C, Eleno N, Tavares P, Rodriguez‐Barbero A, Garcia‐Criado J, Bolanos JP, Lopez‐Novoa JM. Involvement of reactive oxygen species on gentamicin‐induced mesangial cell activation. Kidney Int 62: 1682‐1692, 2002.
 352. Marumo T, Hishikawa K, Yoshikawa M, Fujita T. Epigenetic regulation of BMP7 in the regenerative response to ischemia. J Am Soc Nephrol 19: 1311‐1320, 2008.
 353. Mason J, Beck F, Dorge A, Rick R, Thurau K. Intracellular electrolyte composition following renal ischemia. Kidney Int 20: 61‐70, 1981.
 354. Mason J, Torhorst J, Welsch J. Role of the medullary perfusion defect in the pathogenesis of ischemic renal failure. Kidney Int 26: 283‐293, 1984.
 355. Massicot F, Martin C, Dutertre‐Catella H, Ellouk‐Achard S, Pham‐Huy C, Thevenin M, Rucay P, Warnet J‐M, Claude J‐R. Modulation of energy status and cytotoxicity induced by FK506 and cyclosporin A in a renal epithelial cell line. Arch Toxicol 71: 529‐531, 1997.
 356. Matejka GL, Jennische E. IGF‐I binding and IGF‐I mRNA expression in the post‐ischemic regenerating rat kidney. Kidney Int 42: 1113‐1123, 1992.
 357. Matsuyama M, Yoshimura R, Akioka K, Okamoto M, Ushigome H, Kadotani Y, Nakatani T, Yoshimura M. Tissue factor antisense oligonucleotides prevent renal ischema reperfusion injury. Tranplantation 76: 786‐791, 2003.
 358. Matthys E, Patton MK, Osgood RW, Venkatachalam MA, Stein JH. Alterations in vascular function and morphology in acute ischemic renal failure. Kidney Int 23: 717‐724, 1983.
 359. Mattson DL, Lu S, Cowley AW Jr. Role of nitric oxide in the control of the renal medullary circulation. Clin Exp Pharmacol Physiol 24: 587‐590, 1997.
 360. Megyesi J, Andrade L, Vieira JM Jr, Safirstein RL, Price PM. Positive effect of the induction of p21WAF1/CIP1 on the course of ischemic acute renal failure. Kidney Int 60: 2164‐2172, 2001.
 361. Megyesi J, Di Mari J, Udvarhelyi N, Price PM, Safirstein R. DNA synthesis is dissociated from the immediate‐early gene response in the post‐ischemic kidney. Kidney Int 48: 1451‐1458, 1995.
 362. Megyesi J, Safirstein RL, Price PM. Induction of p21WAF1/CIP1/SDI1 in kidney tubule cells affects the course of cisplatin‐induced acute renal failure. J Clin Invest 101: 777‐782, 1998.
 363. Mehta RL, Chertow GM. Acute renal failure definitions and classification: Time for change? J Am Soc Nephrol 14: 2178‐2187, 2003.
 364. Melk A, Schmidt BMW, Vongwiwatana A, Rayner DC, Halloran PF. Increased expression of senescence‐associated cell cycle inhibitor p16INK4a in deteriorating renal transplants and diseased native kidney. Am J Transplant 5: 1375‐1382, 2005.
 365. Melnikov VY, Ecder T, Fantuzzi G, Siegmund B, Lucia MS, Dinarello CA, Schrier RW, Edelstein CL. Impaired IL‐18 processing protects caspase‐1 deficient mice from ischemic acute renal failure. J Clin Invest 107: 1145‐1152, 2001.
 366. Melnikov VY, Faubel S, Siegmund B, Lucia MS, Ljubanovic D, Edelstein CL. Neutrophil‐independent mechanisms of caspase‐1 and IL‐18 mediated ischemic acute tubular necrosis in mice. J Clin Invest 110: 1083‐1091, 2002.
 367. Michal JT, Julio PJ, Joseph PG, Anthony JC, Allan WA, Govindarajan R, Keith LK, Andrew DB, Matthew DG, Jawed A, Karl AN. Renal hemodynamic, inflammatory, and apoptotic responses to lipopolysaccharide in HO‐1−/− mice. Am J Pathol 170: 1820‐1830, 2007.
 368. Mikhailov V, Mikhailova M, Pulkrabek DJ, Dong Z, Venkatachalam MA, Saikumar P. Bcl‐2 prevents Bax oligomerization in the mitochondrial outer Membrane. J Bioll Chem 276: 18361‐18374, 2001.
 369. Miller SB, Martin DR, Kissane J, Hammerman MR. Insulin‐like growth factor I accelerates recovery from ischemic acute tubular necrosis in the rat. Proc Natl Acad Sciences U S A 89: 11876‐11880, 1992.
 370. Miller SB, Martin DR, Kissane J, Hammerman MR. Hepatocyte growth factor accelerates recovery from acute ischemic renal injury in rats. Am J Physiol 266: F129‐F134, 1994a.
 371. Miller SB, Martin DR, Kissane J, Hammerman MR. Rat models for clinical use of insulin‐like growth factor I in acute renal failure. Am J Physiol266: F949‐F956, 1994b.
 372. Miller TR, Anderson RJ, Linas SL, Henrich WL, Berns AS, Gabow PA, Schrier RW. Urinary diagnostic indices in acute renal failure: A prospective study. Ann Intern Med 89: 47‐50, 1978.
 373. Misao J, Hayakawa Y, Ohno M, Kato S, Fujiwara T, Fujiwara H. Expression of bcl‐2 protein, an inhibitor of apoptosis, and Bax, an accelerator of apoptosis, in ventricular myocytes of human hearts with myocardial infarction. Circulation 94: 1506‐1512, 1996.
 374. Mitazaki S, Kato N, Suto M, Hiraiwa K, Abe S. Interleukin‐6 deficiency accelerates cisplatin‐induced acute renal failure but not systemic injury. Toxicology 265: 115‐121, 2009.
 375. Miyazawa S, Watanabe H, Miyaji C, Hotta O, Abo T. Leukocyte accumulation and changes in extra‐renal organs during renal ischemia reperfusion in mice. J Lab Clin Med 139: 269‐278, 2002.
 376. Molitoris BA. Ischemia‐induced loss of epithelial polarity: Potential role of the actin cytoskeleton. Am J Physiol 260: F769‐F778, 1991.
 377. Molitoris BA. Putting the actin cytoskeleton into perspective: Pathophysiology of ischemic alterations. Am J Physiol 272: F430‐F433, 1997.
 378. Molitoris BA. Actin cytoskeleton in ischemic acute renal failure. Kidney Int 66: 871‐883, 2004.
 379. Molitoris BA, Chan LK, Shapiro JI, Conger JD, Falk SA. Loss of epithelial polarity: A novel hypothesis for reduced proximal tubule Na+ transport following ischemic injury. J Membr Biol 107: 119‐127, 1989.
 380. Molitoris BA, Falk SA, Dahl RH. Ischemia‐induced loss of epithelial polarity. Role of the tight junction. Jo Clin Invest 84: 1334‐1339, 1989.
 381. Molitoris BA, Finn WF. Acute renal failure: A companion to Brenner and rector's. The Kidney. Philadelphia: Saunders, 2001, p. xiv, 535 p., p. 538 of plates.
 382. Molitoris BA, Geerdes A, McIntosh JR. Dissociation and redistribution of Na+,K(+)‐ATPase from its surface membrane actin cytoskeletal complex during cellular ATP depletion. J Cli Invest 88: 462‐469, 1991.
 383. Molitoris BA, Sandoval R, Sutton TA. Endothelial injury and dysfunction in ischemic acute renal failure. Critical Care Medicine 30: S235‐S240, 2002.
 384. Morgera S, Kraft AK, Siebert G, Luft FC, Neumayer HH. Long‐term outcomes in acute renal failure patients treated with continuous renal replacement therapies. Am J Kidney Dis 40: 275‐279, 2002.
 385. Morigi M, Rota C, Montemurro T, Montelatici E, Lo Cicero V, Imberti B, Abbate M, Zoja C, Cassis P, Longaretti L, Rebulla P, Introna M, Capelli C, Benigni A, Remuzzi G, Lazzari L. Life‐sparing effect of human cord blood‐mesenchymal stem cells in experimental acute kidney injury. Stem Cells 28: 513‐522, 2010.
 386. Mortensen J, Shames B, Johnson C, Nilakantan V. MnTMPyP, a superoxide dismutase/catalase mimetic, decreases inflammatory indices in ischemic acute kidney injury. Inflamm Res 60: 299‐307, 2011.
 387. Muramatsu Y, Tsujie M, Kohda Y, Pham B, Perantoni AO, Zhao H, Jo SK, Yuen PS, Craig L, Hu X, Star RA. Early detection of cysteine rich protein 61 (CYR61, CCN1) in urine following renal ischemic reperfusion injury. Kidney Int 62: 1601‐1610, 2002.
 388. Nagothu KK, Bhatt R, Kaushal GP, Portilla D. Fibrate prevents cisplatin‐induced proximal tubule cell death. Kidney Int 68: 2680‐2693, 2005.
 389. Nash K, Hafeez A, Hou S. Hospital‐acquired renal insufficiency. Am J Kidney Dis 39: 930‐936, 2002a.
 390. Nash K, Hafeez A, Hou S. Hospital‐acquired renal insufficiency. Am J Kidney Dis 39: 930‐936, 2002b.
 391. Nath K, Haggard J, Croatt A, Grande J, Poss K, Alam J. The indispensability of heme oxygenase‐1 in protecting against acute heme protein‐induced toxicity in vivo. Am J Pathol 156: 1527‐1535, 2000.
 392. Nath K, Norby S. Reactive oxygen species and acute renal failure. Am J Med 109: 665‐678, 2000.
 393. Nath KA. Heme oxygenase‐1: A provenance for cytoprotective pathways in the kidney and other tissues. Kidney Int 70: 432‐443, 2006.
 394. Nath KA, Croatt AJ, Haggard JJ, Grande JP. Renal response to repetitive exposure to heme proteins: Chronic injury induced by an acute insult. Kidney Int 57: 2423‐2433, 2000.
 395. Nemoto T, Burne MJ, Daniels F, O'Donnell MP, Crosson J, Berens K, Issekutz A, Kasiske BL, Keane WF, Rabb H. Small molecule selectin ligand inhibition improves outcome in ischemic acute renal failure. Kidney Int 60: 2205‐2214, 2001.
 396. Neto JS, Nakao A, Kimizuka K, Romanosky AJ, Stolz DB, Uchiyama T, Nalesnik MA, Otterbein LE, Murase N. Protection of transplant‐induced renal ischemia‐reperfusion injury with carbon monoxide. Am J Physiol Renal Physiol 287: F979‐F989, 2004.
 397. Nicolli A, Basso E, Petronilli V, Wenger RM, Bernardi P. Interactions of cyclophilin with the mitochondrial inner membrane and regulation of the permeability transition pore, a cyclosporin A‐sensitive channel. J Biol Chem 271: 2185‐2192, 1996.
 398. Nitescu N, Ricksten S‐E, Marcussen N, Haraldsson Br, Nilsson U, Basu S, Guron G. N‐acetylcysteine attenuates kidney injury in rats subjected to renal ischaemia‐reperfusion. Nephrol Dial Transplant 21: 1240‐1247, 2006.
 399. Nogae S, Miyazaki M, Kobayashi N, Saito T, Abe K, Saito H, Nakane P, Nakanishi Y, Koji T. Induction of apoptosis in ischemia‐reperfusion model of mouse kidney: Possible involvement of Fas. J Am Soc Nephrol 9: 620‐631, 1998.
 400. Noiri E, Gailit J, Sheth D, Magazine H, Gurrath M, Muller G, Kessler H, Goligorsky MS. Cyclic RGD peptides ameliorate ischemic acute renal failure in rats. Kidney Int 46: 1050‐1058, 1994.
 401. Nolan CV, Shaikh ZA. Lead nephrotoxicity and associated disorders: Biochemical mechanisms. Toxicology 73: 127‐146, 1992.
 402. Nony PA, Schnellmann RG. Mechanisms of renal cell repair and regeneration after acute renal failure. J Pharmacol Exp Ther 304: 905‐912, 2003.
 403. Norman J, Badie‐Dezfooly B, Nord EP, Kurtz I, Schlosser J, Chaudhari A, Fine LG. EGF‐induced mitogenesis in proximal tubular cells: Potentiation by angiotensin II. Am J Physiol 253: F299‐F309, 1987.
 404. Norman J, Fine LG. Intrarenal oxygenation in chronic renal failure. Clin Exp Pharmacol Physiol 33: 989‐996, 2006.
 405. O'Shea MH, Miller SB, Hammerman MR. Effects of IGF‐I on renal function in patients with chronic renal failure. Am J Physiol 264: F917‐F922, 1993.
 406. Ogata M, Iwamoto T, Tazawa N, Nishikawa M, Yamashita J, Takaoka M, Matsumura Y. A novel and selective Na+/Ca2+ exchange inhibitor, SEA0400, improves ischemia/reperfusion‐induced renal injury. Eur J Pharmacol 478: 187‐198, 2003.
 407. Oh D‐J, Dursun B, He Z, Lu L, Hoke TS, Ljubanovic D, Faubel S, Edelstein CL. Fractalkine receptor (CX3CR1) inhibition is protective against ischemic acute renal failure in mice. Am J Physiol Renal Physiol 294: F264‐F271, 2008.
 408. Okada H, Danoff TM, Kalluri R, Neilson EG. Early role of Fsp1 in epithelial‐mesenchymal transformation. Am J Physiol 273: F563‐F574, 1997.
 409. Okusa MD, Linden J, Huang L, Rosin DL, Smith DF, Sullivan G. Enhanced protection from renal ischemia: Reperfusion injury with A2A‐adenosine receptor activation and PDE 4 inhibition. Kidney Int 59: 2114‐2125, 2001.
 410. Oliver J, Mac DM, Tracy A. The pathogenesis of acute renal failure associated with traumatic and toxic injury; renal ischemia, nephrotoxic damage and the ischemic episode. J Clin Invest 30: 1307‐1439, 1951.
 411. Oliver JA, Maarouf O, Cheema FH, Martens TP, Al‐Awqati Q. The renal papilla is a niche for adult kidney stem cells. J Clin Invest 114: 795‐804, 2004.
 412. Oltval ZN, Milliman CL, Korsmeyer SJ. Bcl‐2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programed cell death. Cell 74: 609‐619, 1993.
 413. Ortiz A, Lorz C, Egido J. The Fas ligand/Fas system in renal injury. Nephrol Dial Transplant 14: 1831‐1834, 1999.
 414. Ouellette AJ, Malt RA, Sukhatme VP, Bonventre JV. Expression of two “immediate early” genes, Egr‐1 and c‐fos, in response to renal ischemia and during compensatory renal hypertrophy in mice. J Clin Invest 85: 766‐771, 1990.
 415. Padanilam BJ. Cell death induced by acute renal injury: A perspective on the contributions of apoptosis and necrosis. Am J Physiol Renal Physiol 284: F608‐F627, 2003.
 416. Pagtalunan M, Olson J, Tilney N, Meyer T. Late consequences of acute ischemic injury to a solitary kidney. J Am Soc Nephrol 10: 366‐373, 1999.
 417. Paller MS. Free radical scavengers in mercuric chloride‐induced acute renal failure in the rat. J Lab Clin Med 105: 459‐463, 1985.
 418. Paller MS. Renal work, glutathione and susceptibility to free radical‐mediated postischemic injury. Kidney Int 33: 843‐849, 1988.
 419. Paller MS, Hedlund BE. The role of iron in postischemic renal failure in the rat. Kidney Int 34: 474‐480, 1988.
 420. Paller MS, Hedlund BE. Extracellular iron chelators protect kidney cells from hypoxia/reoxygenation. Free Radic Biol Med 17: 597‐603, 1994.
 421. Paller MS, Hoidal JR, Ferris TF. Oxygen free radicals in ischemic acute renal failure in the rat. J Clin Invest 74: 1156‐1164, 1984.
 422. Parikh CR, Devarajan P. New biomarkers of acute kidney injury. Crit Care Med 36: S159‐S165, 2008.
 423. Park H‐C, Yasuda K, Kuo M‐C, Ni J, Ratliff B, Chander P, Goligorsky MS. Renal capsule as a stem cell niche. Am J Physiol Renal Physiol 298: F1254‐F1262, 2010.
 424. Park K, Chen A, Bonventre JV. Prevention of kidney ischemia/reperfusion induced functional injury and JNK, p38 and MAPK kinase activation by remote ischemic pretreatment. J Biol Chem 276: 11870‐11876, 2001.
 425. Park KM, Kramers C, Vayssier‐Taussat M, Chen A, Bonventre JV. Prevention of kidney ischemia/reperfusion‐induced functional injury, MAPK and MAPK kinase activation, and inflammation by remote transient ureteral obstruction. J Biol Chem 277: 2040‐2049, 2002.
 426. Patschan D, Krupincza K, Patschan S, Zhang Z, Hamby C, Goligorsky MS. Dynamics of mobilzation and homing of endothelial progenitor cells after acute renal ischemia: Modulation by ischemic preconditioning. . Am J Physiol 291: F176‐F185, 2006.
 427. Patschan D, Patschan S, Gobe GG, Chintala S, Goligorsky MS. Uric acid heralds ischemic tissue injury to mobilize endothelial progenitor cells. J Am Soc Nephrol 18: 1516‐1524, 2007.
 428. Pechman K, Basile DP, Lund H, Mattson DL. Immune suppression blocks sodium sensitive hypertension following recovery from acute renal failure. Am J Physiol Regul Integr Comp Physiol 294: R1234‐R1239, 2008
 429. Pechman KR, De Miguel C, Lund H, Leonard EC, Basile DP, Mattson DL. Recovery from renal ischemia‐reperfusion injury is associated with altered renal hemodynamics, blunted pressure natriuresis, and sodium‐sensitive hypertension. Am J Physiol Regul Integr Comp Physiol 297: R1358‐R1363, 2009.
 430. Peralta C, Perales JC, Bartrons R, Mitchell C, Gilgenkrantz H, Xaus C, Prats N, Fernandez L, Gelpi E, Panes J, Rosello‐Catafau J. The combination of ischemic preconditioning and liver Bcl‐2 overexpression is a suitable strategy to prevent liver and lung damage after hepatic ischemia‐reperfusion. Am J Pathol 160: 2111‐2122, 2002.
 431. Perianayagam MC, Liangos O, Kolyada AY, Wald R, MacKinnon RW, Li L, Rao M, Balakrishnan VS, Bonventre JV, Pereira BJ, Jaber BL. NADPH oxidase p22phox and catalase gene variants are associated with biomarkers of oxidative stress and adverse outcomes in acute renal failure. J Am Soc Nephrol 18: 255‐263, 2007.
 432. Periyasamy‐Thandavan S, Jiang M, Schoenlein P, Dong Z. Autophagy: Molecular machinery, regulation, and implications for renal pathophysiology. Am J Physiol Renal Physiol 297: F244‐F256, 2009.
 433. Periyasamy‐Thandavan S, Jiang M, Wei Q, Smith R, Yin X‐M, Dong Z. Autophagy is cytoprotective during cisplatin injury of renal proximal tubular cells. Kidney Int 74: 631‐640, 2008.
 434. Peterson O, Gabbai F, Myers R, Mizisin A, Blantz RC. A single nephron model of acute tubular injury: Role of tubularglomerular feedback. Kidney Int 36: 1037‐1044, 1989.
 435. Petrinec D, Reilly JM, Sicard GA, Lowell JA, Howard TK, Martin DR, Brennan DC, Miller SB. Insulin‐like growth factor‐I attenuates delayed graft function in a canine renal autotransplantation model. Surgery 120: 221‐226, 1996.
 436. Piechota M, Banach M, Irzmanski R, Barylski M, Piechota‐Urbanska M, Kowalski J, Pawlicki L. Plasma endothelin‐1 levels in septic patients. J Intens Care Med 22: 232‐239, 2007.
 437. Pittock ST, Norby SM, Grande JP, Croatt AJ, Bren GD, Badley AD, Caplice NM, Griffin MD, Nath KA. MCP‐1 is up‐regulated in unstressed and stressed HO‐1 knockout mice: Pathophysiologic correlates1. Kidney Int 68: 611‐622, 2005.
 438. Plotnikov EY, Kazachenko AV, Vyssokikh MY, Vasileva AK, Tcvirkun DV, Isaev NK, Kirpatovsky VI, Zorov DB. The role of mitochondria in oxidative and nitrosative stress during ischemia//reperfusion in the rat kidney. Kidney Int 72: 1493‐1502, 2007.
 439. Pritchard KA, Ackerman AW, Gross ER, Stepp DW, Shi Y, Fontana JT, Baker JE, Sessa WC. Heat shock protein 90 mediates the balance of nitric oxide and superoxide anion from endothelial nitric‐oxide synthase. J Biol Chem 276: 17621‐17624, 2001.
 440. Qian T, Nieminen A‐L, Herman B, Lemasters JJ. Mitochondrial permeability transition in pH‐dependent reperfusion injury to rat hepatocytes. Am J Physiol Cell Physiol 273: C1783‐C1792, 1997.
 441. Quigg RJ. Complement and the Kidney. J Immunol 171: 3319‐3324, 2003.
 442. Quiros Y, Vicente‐Vicente L, Morales AI, Lopez‐Novoa J, Lopez‐Hernandez FJ. An integrative overview on the mechanisms underlying the renal tubular cytotoxicity of gentamicin. Toxicol Sci 119: 245‐256, 2011.
 443. Rabb H. Immune modulation of acute kidney injury. J Am Soc Nephrol 17: 604‐606, 2006.
 444. Rabb H, Mendiola CC, Dietz J, Saba SR, Issekutz TB, Abanilla F, Bonventre JV, Ramirez G. Role of CD11a and CD11b in ischemic acute renal failure in rats. Am J Physiol 267: F1052‐F1058, 1994.
 445. Rabb H, Mendiola CC, Saba SR, Dietz JR, Smith CW, Bonventre JV, Ramirez G. Antibodies to ICAM‐1 protect kidneys in severe ischemic reperfusion injury. Biochem Biophys Res Commun 211: 67‐73, 1995.
 446. Rabb H, O'Meara YM, Maderna P, Coleman P, Brady HR. Leukocytes, cell adhesion molecules and ischemic acute renal failure. Kidney Int 51: 1463‐1468, 1997.
 447. Racusen LC. The histopathology of acute renal failure. New Horiz 3: 662‐668, 1995.
 448. Rajdev S, Hara K, Kokubo Y, Mestril R, Dillmann W, Weinstein PR, Sharp FR. Mice overexpressing rat heat shock protein 70 are protected against cerebral infarction. Ann Neurol 47: 782‐791, 2000.
 449. Rajesh KK, Torsten Toftgard N, Andrew NR. Translation of remote ischaemic preconditioning into clinical practice. Lancet 374: 1557‐1565, 2009.
 450. Ramesh G, Reeves WB. TNFR2‐mediated apoptosis and necrosis in cisplatin‐induced acute renal failure. Am J Physiol Renal Physiol 285: F610‐F618, 2003.
 451. Ramirez V, Mejia‐Vilet JM, Hernandez D, Gamba G, Bobadilla NA. Radicicol, a heat shock protein 90 inhibitor, reduces glomerular filtration rate. Am J Physiol Renal Physiol 295: F1044‐F1051, 2008.
 452. Rasbach KA, Schnellmann RG. PGC‐1[alpha] over‐expression promotes recovery from mitochondrial dysfunction and cell injury. Biochem Biophys Res Commun 355: 734‐739, 2007.
 453. Rasbach KA, Schnellmann RG. Isoflavones promote mitochondrial biogenesis. J Pharmacol Exp Ther 325: 536‐543, 2008.
 454. Ratcliffe PJ, Moonen CTW, Holloway PAH, Ledingham JGG, Radda GK. Acute renal failure in hemorrhagic hypotension: Cellular energetics and renal function. Kidney Int 30: 355‐360, 1986.
 455. Reinders MEJ, Rabelink TJ, Briscoe DM. Angiogenesis and endothelial cell repair in renal disease and allograft rejection. J Am Soc Nephrol 17: 932‐942, 2006.
 456. Rhoden E, Telöken C, Lucas M, Rhoden C, Mauri M, Zettler C, Belló‐Klein A, Barros E. Protective effect of allopurinol in the renal ischemia‐reperfusion in uninephrectomized rats. Gen Pharmacol 35: 189‐193, 2000.
 457. Rifkin IR, Leadbetter EA, Busconi L, Viglianti G, Marshak‐Rothstein A. Toll‐like receptors, endogenous ligands, and systemic autoimmune disease. Immunol Rev 204: 27‐42, 2005.
 458. Robinson JWL, Mirkovitch V, Gomba Sz. Alterations in the dog renal tubular epithelium during normothermic ischemia. Kidney Int 11: 86‐92, 1977.
 459. Romanov V, Noiri E, Czerwinski G, Finsinger D, Kessler H, Goligorsky MS. Two novel probes reveal tubular and vascular Arg‐Gly‐Asp (RGD) binding sites in the ischemic rat kidney. Kidney Int 52: 93‐102, 1997.
 460. Rookmaaker MB, Verhaar MC, van Zonneveld AJ, Rabelink TJ. Progenitor cells in the kidney: Biology and therapeutic perspectives. Kidney Int 66: 518‐522, 2004.
 461. Ruchalski K, Mao H, Singh SK, Wang Y, Mosser DD, Li F, Schwartz JH, Borkan SC. HSP72 inhibits apoptosis‐inducing factor release in ATP‐depleted renal epithelial cells. Am J Physiol Cell Physiol 285: C1483‐C1493, 2003.
 462. Sabbagh R, Chawla A, Tisdale B, Kwan K, Chatterjee S, Kwiecien J, Kapoor A. Renal histological features according to various warm ischemia time in a porcine laparoscopic and open surgery model. Can Urol Assoc J 5: 40‐43, 2011.
 463. Safirstein R. Gene expression in nephrotoxic and ischemic acute renal failure. J Am Soc Nephrol 4: 1387‐1395, 1994.
 464. Safirstein R. Renal stress response and acute renal failure. Adv Renal Replace Ther 4: 38‐42, 1997.
 465. Safirstein R, Miller P, Dikman S, Lyman N, Shapiro C. Cisplatin nephrotocxicity in rats: Defect in papillary hypertonicity. Am J Phyiol 241: F175‐F185, 1981.
 466. Safirstein R, Winston J, Moel D, Dikman S, Guttenplan J. Cisplatin nephrotoxicity: Insights into mechanism. Int J Androl 10: 325‐346, 1987.
 467. Safirstein R, Zelent AZ, Price PM. Reduced renal prepro‐epidermal growth factor mRNA and decreased EGF excretion in ARF. Kidney Int 36: 810‐815, 1989.
 468. Safirstein RL. Cell cycle events during renal injury. Ren Fail 21: 427‐431, 1999.
 469. Saikumar P, Dong Z, Patel Y, Hall K, Hopfer U, Weinberg JM, Venkatachalam MA. Role of hypoxia‐induced Bax translocation and cytochrome c release in reoxygenation injury. Oncogene 17: 3401‐3415, 1998.
 470. Sakai M, Zhang M, Homma T, Garrick B, Abraham JA, McKanna JA, Harris RC. Production of heparin binding epidermal growth factor‐like growth factor in the early phase of regeneration after acute renal injury. Isolation and localization of bioactive molecules. J Clin Invest 99: 2128‐2138, 1997.
 471. Salahudeen AK, Huang H, Joshi M, Moore NA, Jenkins JK. Involvement of the mitochondrial pathway in cold storage and rewarming‐associated apoptosis of human renal proximal tubular cells. Am J Transplant 3: 273‐280, 2003.
 472. Salman IM, Ameer OZ, Sattar MA, Abdullah NA, Yam MF, Najim HS, Khan AH, Johns EJ. Role of the renal sympathetic nervous system in mediating renal ischaemic injury‐induced reductions in renal haemodynamic and excretory functions. Pathology 42: 259‐266.
 473. Salmela K, Wramner L, Ekberg H, Hauser I, Bentdal O, Lins L‐E, Isoniemi H, Bäckman L, Persson N, Neumayer H‐H, Jørgensen PF, Spieker C, Hendry B, Nicholls A, Kirste Gn, Hasche G. A randomized multicenter trial of the anti‐ICAM‐1 monoclonal antibody (enlimomab) for the prevention of acute rejection and delayed onset of graft function in cadaveric renal transplantation: A report of The European Anti‐ICAM‐1 Renal Transplant Study Group. Transplantation 67: 729‐736, 1999.
 474. Salvioli S, Ardizzoni A, Franceschi C, Cossarizza A. JC‐1, but not DiOC6(3) or rhodamine 123, is a reliable fluorescent probe to assess [Delta][Psi] changes in intact cells: Implications for studies on mitochondrial functionality during apoptosis. FEBS Lett 411: 77‐82, 1997.
 475. Sanz AB, Santamaria B, Ruiz‐Ortega M, Egido J, Ortiz A. Mechanisms of renal apoptosis in health and disease. J Am Soc Nephrol 19: 1634‐1642, 2008.
 476. Schaudies RP, Johnson JP. Increased soluble EGF after ischemia is accompanied by a decrease in membrane associated precursors. Am J Physiol 264: F523‐F531, 1993.
 477. Schiffl H. Renal recovery from acute tubular necrosis requiring renal replacement therapy: A prospective study in critically ill patients. Nephrol Dial Transplant 21: 1248‐1252, 2006.
 478. Schinzel AC, Takeuchi O, Huang Z, Fisher JK, Zhou Z, Rubens J, Hetz C, Danial NN, Moskowitz MA, Korsmeyer SJ. Cyclophilin D is a component of mitochondrial permeability transition and mediates neuronal cell death after focal cerebral ischemia. Proc Natl Acad Sci U S A 102: 12005‐12010, 2005.
 479. Schmitt R, Cantley LG. The impact of aging on kidney repair. Am J Physiol Renal Physiol 294: F1265‐F1272, 2008.
 480. Schmitt R, Coca S, Kanbay M, Tinetti ME, Cantley LG, Parikh CR. Recovery of kidney function after acute kidney injury in the elderly: A systematic review and meta‐analysis. Am J Kidney Dis 52: 262‐271, 2008.
 481. Schmitt R, Marlier A, Cantley LG. Zag Expression during aging suppresses proliferation after kidney injury. J Am Soc Nephrol 19: 2375‐2383, 2008.
 482. Schneider J, Friderichs E, Giertz H. Comparison of the protective effects by human and bovine superoxide dismutase against ischemia‐ and reperfusion‐induced impairment of kidney function in anesthetized rats. Free Radic Biol Med 3: 21‐26, 1987.
 483. Schneider R, Raff U, Vornberger N, Schmidt M, Freund R, Reber M, Schramm L, Gambaryan S, Wanner C, Schmidt HH, Galle J. L‐Arginine counteracts nitric oxide deficiency and improves the recovery phase of ischemic acute renal failure in rats. Kidney Int 64: 216‐225, 2003.
 484. Schober A, Muller E, Thurau K, Beck F. The response of heat shock proteins 25 and 72 to ischaemia in different kidney zones. Pflugers Arch 292‐299: 1997.
 485. Schramm L, La M, Heidbreder E, Hecker M, Beckman JS, Lopau K, Zimmermann J, Rendl J, Reiners C, Winderl S, Wanner C, Schmidt HH. L‐arginine deficiency and supplementation in experimental acute renal failure and in human kidney transplantation. Kidney Int 61: 1423‐1432, 2002.
 486. Schrier RW, Arnold PE, Van Putten VJ, Burke TJ. Cellular calcium in ischemic acute renal failure: Role of calcium entry blockers. Kidney Int 32: 313‐321, 1987.
 487. Schrier RW, Burke TJ. Role of calcium‐channel blockers in preventing acute and chronic renal failure. J Cardiovasc Pharmacol 18: S38‐S43, 1991.
 488. Schulman G, Fogo A, Gung A, Badr K, Hakim R. Complement activation retards resolution of acute ischemic renal failure in the rat. Kidney Int 40: 1069‐1074, 1991.
 489. Schumer M, Colombel MC, Sawczuk IS, Gobe G, Connor J, O'Toole KM, Olsson CA, Wise GJ, Buttyan R. Morphologic, biochemical, and molecular evidence of apoptosis during the reperfusion phase after brief periods of renal ischemia. Am J Pathol 140: 831‐838, 1992.
 490. Schwartz N, Hosford M, Sandoval RM, Wagner MC, Atkinson SJ, Bamburg J, Molitoris BA. Ischemia activates actin depolymerizing factor: Role in proximal tubule microvillar actin alterations. Am J Physiol 276: F544‐F551, 1999.
 491. Shanley PF, Brezis M, Spokes K, Silva P, Epstein FH, Rosen S. Hypoxic injury in the proximal tubule of the isolated perfused rat kidney. Kidney Int 29: 1021‐1032, 1986.
 492. Sharfuddin AA, Sandoval RM, Berg DT, McDougal GE, Campos SB, Phillips CL, Jones BE, Gupta A, Grinnell BW, Molitoris BA. Soluble thrombomodulin protects ischemic kidneys. J Am Soc Nephrol 20: 524‐534, 2009.
 493. Sheikh‐Hamad D, Cacini W, Buckley A, Isaac J, Truong L, Tsao C, Kishore B. Cellular and molecular studies on cisplatin‐induced apoptotic cell death in rat kidney. Arch Toxicol 78: 147‐155, 2004.
 494. Shelden E, Borelli MJ, Pollock FM, Bonham R. Heat shock protein 27 associates with basolateral cell boudaries in heat shocked and ATP depleted epithelial cells. J Am Soc Nephrol 13: 332‐341, 2002.
 495. Sheridan AM, Bonventre JV. Cell biology and molecular mechanisms of injury in ischemic acute renal failure. Curr Opin Nephrol Hypertens 9: 427‐434, 2000.
 496. Shi H, Patschan D, Epstein T, Goligorsky MS, Winaver J. Delayed recovery of renal regional blood flow in diabetic mice subjected to acute ischemic kidney injury. Am J Physiol 293: F1512‐F1517, 2007.
 497. Shimizu A, Yamanaka N. Apoptosis and cell desquamation in repair process of ischemic tubular necrosis. Virchows Arch B Cell Pathol Incl Mol Pathol 64: 171‐180, 1993.
 498. Shimizu H, Takahashi T, Suzuki T, Yamasaki A, Fujiwara T, Odaka Y, Hirakawa M, Fujita H, Akagi R. Protective effect of heme oxygenase induction in ischemic acute renal failure. Crit Care Med 28: 809‐817, 2000.
 499. Shoag J, Arany Z. Regulation of hypoxia‐inducible genes by PGC‐1alpha. Arterioscler Thromb Vasc Biol 30: 662‐666.
 500. Siegel N, Feldman R, Lytton B, Hayslett J, Kashgarian M. Renal cortical blood flow distribution in obstructive nephropathy in rats. Circ Res 40: 379‐384, 1977.
 501. Siegel NJ, Avison MJ, Reilly HF, Alger JR, Shulman RG. Enhanced recovery of renal ATP with postischemic infusion of ATP‐MgCl2 determined by 31P‐NMR. Am J Physiol 245: F530‐F534, 1983.
 502. Siegel NJ, Devarajan P, Van Why S. Renal cell injury: Metabolic and structural alterations. Pediatr Res 36: 129‐136, 1994.
 503. Siegel NJ, Glazier WB, Chaudry IH, Gaudio KM, Lytton B, Baue AE, Kashgarian M. Enhanced recovery from acute renal failure by the postischemic infusin of adenine nucleotides and magnesium chloride in rats. Kidney Int 17: 338‐349, 1980.
 504. Siew ED, Ware LB, Ikizler TA. Biological markers of acute kidney injury. J Am Soc Nephrol 22: 810‐820, 2011.
 505. Sikorski EM, Hock T, Hill‐Kapturczak N, Agarwal A. The story so far: Molecular regulation of the heme oxygenase‐1 gene in renal injury. Am J Physiol Renal Physiol 286: F425‐F441, 2004.
 506. Simmons CF, Bogusky RT, Humes HD. Inhibitory effects of gentamicin on renal mitochondrial oxidative phosphorylation. J Pharmacol Exp Ther 214: 709‐715, 1980.
 507. Singh AB, Kaushal V, Megyesi JK, Shah SV, Kaushal GP. Cloning and expression of rat caspase‐6 and its localization in renal ischemia/reperfusion injury. Kidney Int 62: 106‐115, 2002.
 508. Singh I, Gulati S, Orak JK, Singh AK. Expression of antioxidant enzymes in rat kidney during ischemia‐reperfusion injury. Mol Cell Biochem 125: 97‐104, 1993.
 509. Smoyer WE, Ransom R, Harris RC, Welsh MJ, Lutsch G, Benndorf R. Ischemic acute renal failure induces differential expression of small heat shock proteins. J Am Soc Nephrol 11: 211‐221, 2000.
 510. Snoeijs MG, Vink H, Voesten N, Christiaans MH, Daemen J‐WH, Peppelenbosch AG, Tordoir JH, Peutz‐Kootstra CJ, Buurman WA, Schurink GWH, van Heurn LWE. Acute ischemic injury to the renal microvasculature in human kidney transplantation. Am J Physiol Renal Physiol 299: F1134‐F1140, 2010.
 511. Solez K. Acute renal failure. In: Heptinstall R, editor. Pathophysiology of the Kidney. Toronto: Little, Brown and Company, 1983, p. 1235‐1314.
 512. Solez K, Ideura T, Silvia CB, Hamilton B, Saito H. Clonidine after renal ischemia to lessen acute renal failure and microvascular damage. Kidney Int 18: 309‐322, 1980.
 513. Solez K, Kramer E, Fox J, Heptinstall R. Medullary plasma flow and intravascular leukocyte accumulation in acute renal failure. Kidney Int 6: 24‐37, 1974.
 514. Solez K, Morel‐Maroger L, Sraer JD. The morphology of “acute tubular necrosis” in man: Analysis of 57 renal biopsies and a comparison with the glycerol model. Medicine (Baltimore) 58: 362‐376, 1979.
 515. Solez K, Racusen LC, Marcussen N, Slatnik I, Keown P, Burdick JF, Olsen S. Morphology of ischemic acute renal failure, normal function, and cyclosporine toxicity in cyclosporine‐treated renal allograft recipients. Kidney Int 43: 1058‐1067, 1993.
 516. Solez K, Racusen LC, Whelton A. Glomerular epithelial cell changes in early postischemic acute renal failure in rabbits and man. Am J Pathol 103: 163‐173, 1981.
 517. Son D, Kojima I, Inagi R, Matsumoto M, Fujita T, Nangaku M. Chronic hypoxia aggravates renal injury via suppression of Cu/Zn‐SOD: A proteomic analysis. Am J Physiol Renal Physiol 294: F62‐F72, 2008.
 518. Spargias K, Adreanides E, Demerouti E, Gkouziouta A, Manginas A, Pavlides G, Voudris V, Cokkinos DV. Iloprost prevents contrast‐induced nephropathy in patients with renal dysfunction undergoing coronary angiography or intervention. Circulation 120: 1793‐1799, 2009.
 519. Spurgeon‐Pechman KR, Donohoe DL, Mattson DL, Lund H, James L, Basile DP. Recovery from acute renal failure predisposes hypertension and secondary renal disease in response to elevated sodium. Am J Physiol Renal Physiol 293: F269‐F278, 2007.
 520. Spurgeon KS, Donohoe DL, Basile DP. Transforming growth factor‐ß in acute renal failure: Receptor expression, influence in cell proliferation, cellularity and vascularization after recovery from injury. Am J Physiol Renal Physiol 288: F568‐F577, 2005.
 521. Sreedharan R, Devarajan P, Van Why S. Pathogenesis of acute renal failure. In: Avner E, Harmon W, Niaudet P, Yoshikawa N, editors. Pediatric Nephrology. Heidleberg: Springer‐Verlag, 2009, p. 1579‐1602.
 522. Star RA. Treatment of acute renal failure. Kidney Int 54: 1817‐1831, 1998.
 523. Stracke S, Ernst F, Jehle DR, Grunewald RW, Haller H, Keller F, Jehle PM. Differentiating and proliferative effects of HGF in renal proximal tubular cells are mediated via different signalling pathways. Nephrol Dial Transplant 13: 1398‐1405, 1998.
 524. Stromski ME, Cooper K, Thulin G, Gaudio KM, Siegel NJ, Shulman RG. Chemical and functional correlates of postischemic renal ATP levels. Proc Natl Acad Sci U S A 83: 6142‐6145, 1986.
 525. Sucher R, Gehwolf P, Oberhuber R, Hermann M, Margreiter C, Werner ER, Obrist P, Schneeberger S, Ollinger R, Margreiter R, Brandacher G. Tetrahydrobiopterin protects the kidney from ischemia‐reperfusion injury. Kidney Int 77: 681‐689, 2010.
 526. Sugiura H, Yoshida T, Tsuchiya K, Mitobe M, Nishimura S, Shirota S, Akiba T, Nihei H. Klotho reduces apoptosis in experimental ischaemic acute renal failure. Nephrol Dial Transplant 20: 2636‐2645, 2005.
 527. Sugiura T, Kobuchi S, Tsutsui H, Takaoka M, Fujii T, Hayashi K, Matsumura Y. Preventive mechanisms of agmatine against ischemic acute kidney injury in rats. Eur J Pharmacol 603: 108‐113, 2009.
 528. Sun D, Samuelson LC, Yang T, Huang Y, Paliege A, Saunders T, Briggs J, Schnermann J. Mediation of tubuloglomerular feedback by adenosine: Evidence from mice lacking adenosine 1 receptors. Proc Natl Acad Sci U S A 98: 9983‐9988, 2001.
 529. Supavekin S, Zhang W, Kucherlapati R, Kaskel FJ, Moore LC, Devarajan P. Differential gene expression following early renal ischemia/reperfusion. Kidney Int 63: 1714‐1724, 2003.
 530. Surendran K, Kopan R. Chromatin‐based mechanisms of renal epithelial differentiation. J Am Soc Nephrol 22: 1208‐1212, 2011.
 531. Sutton TA. Alteration of microvascular permeability in acute kidney injury. Microvasc Res 77: 4‐7, 2009.
 532. Sutton TA, Fisher CJ, Molitoris BA. Microvascular endothelial injury and dysfunction during ischemic acute renal failure. Kidney Int 62: 1539‐1549, 2002.
 533. Sutton TA, Kelly KJ, Mang H, Plotkin Z, Sandoval R, Dagher PC. Minocycline reduces microvascular leakage in a model of ischemic renal injury. Am J Physiol Renal Physiol 288: F91‐F97, 2005.
 534. Sutton TA, Mang HE, Campos SB, Sandoval RM, Yoder MC, Molitoris BA. Injury of the renal microvascular endothelium alters barrier function after ischemia. Am J Physiol Renal Physiol 285: F191‐F198, 2003.
 535. Suzuki S, Maruyama S, Sato W, Morita Y, Sato F, Miki Y, Kato S, Katsuno M, Sobue G, Yuzawa Y, Matsuo S. Geranylgeranylacetone ameliorates ischemic acute renal failure via induction of Hsp70. 67: 2210‐2220, 2005.
 536. Tadagavadi RK, Reeves WB. Renal dendritic cells ameliorate nephrotoxic acute kidney injury. J Am Soc Nephrol 21: 53‐63, 2010.
 537. Takada M, Chandraker A, Nadeau KC, Sayegh MH, Tilney NL. The role of the B7 costimulatory pathway in experimental cold ischemia/reperfusion injury. J Clin Invest 100: 1199‐1203, 1997.
 538. Takeyama N, Miki S, Hirakawa A, Tanaka T. Role of the mitochondrial permeability transition and cytochrome c release in hydrogen peroxide‐induced apoptosis. Exp Cell Res 274: 16‐24, 2002.
 539. Taman M, Liu Y, Tolbert E, Dworkin L. Increase urinary hepatocyte growth factor excretion in human acute renal failure. Clin Nephrol 48: 241‐245, 1997.
 540. Terada Y, Tanaka H, Okado T, Shimamura H, Inoshita S, Kuwahara M, Sasaki S. Expression and function of the developmental gene Wnt‐4 during experimental acute renal failure in rats. J Am Soc Nephrol 14: 1223‐1233, 2003.
 541. Terry B, Jones D, Mueller C. Experimental ischemic renal arterial necrosis with resolution. Am J Pathol 58: 69‐83, 1970.
 542. Thadhani R, Pascual M, Bonventre JV. Acute renal failure. New Eng J Med 334: 1448‐1460, 1996.
 543. Thakar CV, Quate‐Operacz M, Leonard AC, Eckman MH. Outcomes of hemodialysis patients in a long‐term care hospital setting: A Single‐Center Study. Am J Kidney Dis 55: 300‐306, 2010.
 544. Thomas SE, Anderson S, Gordon KL, Oyama TT, Shankland SJ, Johnson RJ. Tubulointerstitial disease in aging: Evidence for underlying peritubular capillary damage, a potential role for renal ischemia. J Am Soc Nephrol 9: 231‐242, 1998.
 545. Thurman JM, Lenderink AM, Royer PA, Coleman KE, Zhou J, Lambris JD, Nemenoff RA, Quigg RJ, Holers VM. C3a is required for the production of CXC chemokines by tubular epithelial cells after renal ishemia/reperfusion. J Immunol 178: 1819‐1828, 2007.
 546. Thurman JM, Ljubanovic D, Edelstein CL, Gilkeson GS, Holers VM. Lack of a functional alternative complement pathway ameliorates ischemic acute renal failure in mice. J Immunol 170: 1517‐1523, 2003.
 547. Thurman JM, Ljubanovic D, Royer PA, Kraus DM, Molina H, Barry NP, Proctor G, Levi M, Holers VM. Altered renal tubular expression of the complement inhibitor Crry permits complement activation after ischemia/reperfusion. J Clin Invest 116: 357‐368, 2006.
 548. Tobimatsu M, Euda Y, Saito S, Tsumagari T, Konomi K. Effects of a stable prostocyclin analog on experimental ischemic acute renal failure. Ann Surg 208: 65‐70, 1988.
 549. Togel F, Isaac J, Hu Z, Weiss K, Westenfelder C. Renal SDF‐1 signals mobilization and homing of CXCR4‐positive cells to the kidney after ischemic injury. Kidney Int 67: 1772‐1784, 2005.
 550. Togel F, Isaac J, Westenfelder C. Hematopoietic stem cell mobilization–associated granulocytosis severely worsens acute renal failure. J Am Soc Nephrol 15: 1261‐1267, 2004.
 551. Togel F, Weiss K, Yang Y, Hu Z, Zhang P, Westenfelder C. Vasculotropic, paracrine actions of infused mesenchymal cells are important to the recovery from acute kidney injury. Am J Physiol 292: F1626‐F1635, 2007.
 552. Togel F, Yang Y, Zhang P, Hu Z, Westenfelder C. Bioluminescence imaging to monitor the in vivo distribution of administered mesenchymal stem cells in acute kidney injury. Am J Physiol Renal Physiol 295: F315‐F321, 2008.
 553. Togel FE, Westenfelder C. Mesenchymal stem cells: A new therapeutic tool for AKI. Nat Rev Nephrol 6: 179‐183, 2010.
 554. Toronyi E, Lord R, Bowen ID, Perner F, Szende B. Renal tubular cell necrosis and apoptosis in transplanted kidneys. Cell Biol Int 25: 267‐270, 2001.
 555. Torras J, Herrero‐Fresneda I, Lloberas N, Riera M, Ma Cruzado J, Ma Grinyo J. Promising effects of ischemic preconditioning in renal transplantation. Kidney Int 61: 2218‐2227, 2002.
 556. Torras J, Seron D, Herrero I, Martinez‐Castelao A, Carrera M, Alsina J, Grino JM. Renal protective effect of liposomed superoxide dismutase in an experimental warm ischemia model. Transplant Int 7: S472‐S475, 1994.
 557. Traktuev DO, Merfeld‐Clauss S, Li J, Kolonin M, Arap W, Pasqualini R, Johnstone BH, March KL. A population of multipotent CD34‐positive adipose stromal cells share pericyte and mesenchymal surface markers, reside in a periendothelial location, and stabilize endothelial networks. Circ Res 102: 77‐85, 2008.
 558. Tsukamoto T, Nigam SK. Tight junction proteins form large complexes and associate with the cytoskeleton in an ATP depletion model for reversible junction assembly. J Biol Chem 272: 16133‐16139, 1997.
 559. Tsuruta F, Sunayama J, Mori Y, Hattori S, Shimizu S, Tsujimoto Y, Yoshioka K, Masuyama N, Gotoh Y. JNK promotes Bax translocation to mitochondria through phosphorylation of 14‐3‐3 proteins. Embo J 23: 1889‐1899, 2004.
 560. Tumlin JA, Finkel KW, Murray PT, Samuels J, Cotsonis G, Shaw AD. Fenoldopam mesylate in early acute tubular necrosis: A randomized, double‐blind, placebo‐controlled clinical trial. Am J Kidney Dis 46: 26‐34, 2005.
 561. Uchida S, Endou H. Substrate specificity to maintain cellular ATP along the mouse nephron. Am J Physiol 255: F977‐F983, 1988.
 562. Uchino S, Kellum JA, Bellomo R, Doig GS, Morimatsu H, Morgera S, Schetz M, Tan I, Bouman C, Macedo E, Gibney N, Tolwani A, Ronco C. Acute renal failure in critically ill patients: A multinational, multicenter study. JAMA 294: 813‐818, 2005.
 563. Ueda N, Kaushal GP, Shah SV. Apoptotic mechanisms in acute renal failure. Am J Med 108: 403‐415, 2000.
 564. Ueda N, Shah SV. Tubular cell damage in acute renal failure‐apoptosis, necrosis, or both. Nephrol Dial Transplant 15: 318‐323, 2000.
 565. Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, Fujita T, Fukumoto S, Yamashita T. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature 444: 770‐774, 2006.
 566. Van Arsdale R. The pharmacology of mercury. J Am Med Associat 81: 1748‐1752, 1923.
 567. Van Why S, Mann A, Ardito T, Thulin G, Ferris S, MacLeod M, Kashgarian M, Siegel NJ. Hsp27 associates with actin and limits injury in energy depleted renal epithelia. J Am Soc Nephrol 13: 98‐106, 2003.
 568. Van Why SK, Hildebrandt F, Ardito T, Mann AS, Siegel NJ, Kashgarian M. Induction and intracellular localization of HSP‐72 after renal ischemia. Am J Physiol 263: F769‐F775, 1992.
 569. Van Why SK, Mann AS, Ardito T, Siegel NJ, Kashgarian M. Expression and molecular regulation of Na(+)‐K(+)‐ATPase after renal ischemia. Am J Physiol 267: F75‐F85, 1994.
 570. Van Why SK, Siegel NJ. Heat shock proteins in renal injury and recovery. Curr Opin Nephrol Hypertens 7: 407‐412, 1998.
 571. Van Why SK, Siegel NJ. Heat shock proteins: Role in prevention and recovery from acute renal failure. In: Molitoris BA, Finn WF, editors. Acute Renal Failure: A Companion to Brenner and Rectors The Kidney.. Philadelphia: WB Sanders, 2001, p. 143‐156.
 572. Vásárhelyi B, Tóth‐Heyn P, Treszl A, Tulassay T. Genetic polymorphisms and risk for acute renal failure in preterm neonates. Pediatr Nephrol 20: 132‐135, 2005.
 573. Venkatachalam MA, Bernard DB, Donohoe JF, Levinsky N. Ischemic damage and repair in the rat proximal tubule: Differences among S1, S2 and S3 segments. Kidney Int 14: 31‐49, 1978.
 574. Venkatachalam MA, Griffin KA, Lan R, Geng H, Saikumar P, Bidani AK. Acute kidney injury: A springboard for progression in chronic kidney disease. Am J Physiol Renal Physiol 298: F1078‐F1094, 2010.
 575. Vera T, Henegar JR, Drummond HA, Rimoldi JM, Stec DE. Protective effect of carbon monoxide–releasing compounds in ischemia‐induced acute renal failure. J Am Soc Nephrol 16: 950‐958, 2005.
 576. Villanueva S, Cespedes C, Gonzalez AA, Roessler E, Vio CP. Inhibition of bFGF‐receptor type 2 increases kidney damage and suppresses nephrogenic protein expression after ischemic acute renal failure. Am J Physiol 294: R819‐R828, 2008.
 577. Vinuesa E, Hotter G, Jung M, Herrero‐Fresneda I, Torras J, Sola A. Macrophage involvement in the kidney repair phase after ischaemia/reperfusion injury. J Pathol 214: 104‐113, 2008.
 578. Vogt M, Farber JL. On the molecular pathology of ischemic renal cell death: Reversible and irreversible cellular and mitochondrial metabolic alterations. Am J Pathol 53: 1‐25, 1968.
 579. Vukicevic S, Basic V, Rogic D, Basic N, Shih MS, Shepard A, Jin D, Dattatreyamurty B, Jones W, Dorai H, Ryan S, Griffiths D, Maliakal J, Jelic M, Pastorcic M, Stavljenic A, Sampath TK. Osteogenic protein‐1 (bone morphogenetic protein‐7) reduces severity of injury after ischemic acute renal failure in rat. J Clin Invest 102: 202‐214, 1998.
 580. Wada T, Sakai N, Matsushima K, Kaneko S. Fibrocytes: A new insight into kidney fibrosis. Kidney Int 72: 269‐273, 2007.
 581. Wahlberg JAN, Karlberg L, Persson AEG. Total and regional renal blood flow during complete unilateral ureteral obstruction. Acta Physiol Scand 121: 111‐118, 1984.
 582. Waikar SS, Curhan GC, Wald R, McCarthy EP, Chertow GM. Declining mortality in patients with acute renal failure, 1988 to 2002. J Am Soc Nephrol 17: 1143‐1150, 2006.
 583. Waikar SS, Winkelmayer WC. Chronic on acute renal failure: Long‐term implications of severe acute kidney injury. JAMA 302: 1227‐1229, 2009.
 584. Wald R, Quinn RR, Luo J, Li P, Scales DC, Mamdani MM, Ray JG. Chronic dialysis and death among survivors of acute kidney injury requiring dialysis. JAMA 302: 1179‐1185, 2009.
 585. Walker PD. Alterations in renal tubular extracellular matrix components after ischemia‐reperfusion injury to the kidney. Lab Invest 70: 339‐346, 1994.
 586. Wang A, Holcslaw T, Bashore TM, Freed MI, Miller D, Rudnick MR, Szerlip H, Thames MD, Davidson CJ, Shusterman N, Schwab SJ. Exacerbation of radiocontrast nephrotoxicity by endothelin receptor antagonism. Kidney Int 57: 1675‐1680, 2000.
 587. Wang J, Wei Q, Wang C‐Y, Hill WD, Hess DC, Dong Z. Minocycline up‐regulates Bcl‐2 and protects against cell death in mitochondria. J Biol Chem 279: 19948‐19954, 2004.
 588. Wang W, Falk SA, Jittikanont S, Gengaro PE, Edelstein CL, Schrier RW. Protective effect of renal denervation on normotensive endotoxemia‐induced acute renal failure in mice. Am J Physiol Renal Physiol 283: F583‐F587, 2002.
 589. Wang W, Mitra A, Poole B, Falk S, Lucia MS, Tayal S, Schrier R. Endothelial nitric oxide synthase‐deficient mice exhibit increased susceptibility to endotoxin‐induced acute renal failure. Am J Physiol Renal Physiol 287: F1044‐F1048, 2004.
 590. Wang W, Zolty E, Falk S, Summer S, Stearman R, Geraci M, Schrier R. Prostacyclin in endotoxemia‐induced acute kidney injury: Cyclooxygenase inhibition and renal prostacyclin synthase transgenic mice. Am J Physiol Renal Physiol 293: F1131‐F1136, 2007.
 591. Wang Y, Harris DCH. Macrophages in renal disease. J Am Soc Nephrol 22: 21‐27, 2011.
 592. 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 20: 1544‐1555, 2009.
 593. Wang Y, Wang YP, Zheng G, Lee VWS, Ouyang L, Chang DHH, Mahajan D, Coombs J, Wang YM, Alexander SI, Harris DCH. Ex vivo programmed macrophages ameliorate experimental chronic inflammatory renal disease. Kidney Int 72: 290‐299, 2007.
 594. Wang Z, Chen J‐K, Wang S‐w, Moeckel G, Harris RC. Importance of functional EGF receptors in recovery from acute nephrotoxic injury. J Am Soc Nephrol 14: 3147‐3154, 2003.
 595. Wei MC, Zong W‐X, Cheng EH‐Y, Lindsten T, Panoutsakopoulou V, Ross AJ, Roth KA, MacGregor GR, Thompson CB, Korsmeyer SJ. Proapoptotic BAX and BAK: A requisite gateway to mitochondrial dysfunction and death. Science 292: 727‐730, 2001.
 596. Wei Q, Alam MM, Wang M‐H, Yu F, Dong Z. Bid activation in kidney cells following ATP depletion in vitro and ischemia in vivo. Am J Physiol Renal Physiol 286: F803‐F809, 2004.
 597. Wei Q, Dong G, Franklin J, Dong Z. The pathological role of Bax in cisplatin nephrotoxicity. Kidney Int 72: 53‐62, 2007.
 598. Wei Q, Yin X‐M, Wang M‐H, Dong Z. Bid deficiency ameliorates ischemic renal failure and delays animal death in C57BL/6 mice. Am J Physiol Renal Physiol 290: F35‐F42, 2006.
 599. Weinberg JM. The cell biology of ischemic renal injury. Kidney Int 39: 476‐500, 1991.
 600. Weinberg JM, Davis JA, Abarzua M, Rajan T. Cytoprotective effects of glycine and glutathione against hypoxic injury to renal tubules. J Clin Invest 80: 1446‐1454, 1987.
 601. Weinberg JM, Molitoris BA. Illuminating mitochondrial function and dysfunction using multiphoton technology. J Am Soc Nephrol 20: 1164‐1166, 2009.
 602. Weinberg JM, Venkatachalam MA, Roeser NF, Nissim I. Mitochondrial dysfunction during hypoxia/reoxygenation and its correction by anaerobic metabolism of citric acid cycle intermediates. Proc Natl Acad Sci U S A 97: 2826‐2831, 2000.
 603. Wesche‐Soldato DE, Chung C‐S, Lomas‐Neira J, Doughty LA, Gregory SH, Ayala A. In vivo delivery of caspase‐8 or Fas siRNA improves the survival of septic mice. Blood 106: 2295‐2301, 2005.
 604. Westhoff JH, Schildhorn C, Jacobi C, Homme M, Hartner A, Braun H, Kryzer C, Wang C, von Zglinicki T, Kranzlin B, Gretz N, Melk A. Telomere shortening reduces regenerative capacity after acute kidney injury. J Am Soc Nephrol 21: 327‐336, 2009.
 605. Wilhelm SM, Simonson MS, Robinson AV, Stowe NT, Schulak JA. Endothelin up‐regulation and localization following renal ischemia and reperfusion. Kidney Int 55: 1011‐1018, 1999.
 606. Willinger C, Schramek H, Pfaller K, Pfaller W. Tissue distribution of Neutrophils in postischemic acute renal failure. Virchows Archiv B Cell Patholy Incl Mol Pathol 62: 237‐243, 1992.
 607. Wilson PD, Schrier RW. Nephron segment and calcium as determinants of anoxic cell death in renal cultures. Kidney Int 29: 1172‐1179, 1986.
 608. Witzgall R, Brown D, Schwarz C, Bonventre JV. Localization of proliferating cell nuclear antigen, vimentin, c‐fos and clusterin in the post‐ischemic kidney. J Clin Invest 93: 2175‐2188, 1994.
 609. Wu H, Chen G, Wyburn KR, Yin J, Bertolino P, Eris JM, Alexander SI, Sharland AF, Chadban SJ. TLR4 activation mediates kidney ischemia/reperfusion injury. J Clin Invest 117: 2847‐2859, 2007.
 610. Yamamoto T, Tada T, Brodsky SV, Tanaka H, Noiri E, Kajiya F, Goligorsky MS. Intravital videomicroscopy of peritubular capillaries in renal ischemia. Am J Physiol Renal Physiol 282: F1150‐F1155, 2002.
 611. Yamanobe T, Okada F, Iuchi Y, Onuma K, Tomita Y, Fujii J. Deterioration of ischemia/reperfusion‐induced acute renal failure in SOD1‐deficient mice. Free Radic Res 41: 200‐207, 2007.
 612. Yamashita J, Kita S, Iwamoto T, Ogata M, Takaoka M, Tazawa N, Nishikawa M, Wakimoto K, Shigekawa M, Komuro I, Matsumura Y. Attenuation of ischemia/reperfusion‐induced renal injury in mice deficient in Na+/Ca2+ exchanger. J Pharmacol Exp Ther 304: 284‐293, 2003.
 613. Yamasowa H, Shimizu S, Inoue T, Takaoka M, Matsumura Y. Endothelial nitric oxide contributes to the renal protective effects of ischemic preconditioning. J Pharmacol Exp Ther 312: 153‐159, 2005.
 614. Yanagita M, Okuda T, Endo S, Tanaka M, Takahashi K, Sugiyama F, Kunita S, Takahashi S, Fukatsu A, Yanagisawa M, Kita T, Sakurai T. Uterine sensitization‐associated gene (USAG‐1), a novel BMP antagonist expressed in the kidney, accelerates tubular injury. J Clin Invest 116: 70‐79, 2006.
 615. Yang C‐C, Lin L‐C, Wu M‐S, Chien C‐T, Lai M‐K. Repetitive hypoxic preconditioning attenuates renal ischemia/reperfusion induced oxidative injury via upregulating HIF‐1[alpha]‐dependent bcl‐2 signaling. Transplantation 88: 1251‐1260, 2009.
 616. Yang CW, Li C, Jung JY, Shin SJ, Choi BS, Lim SW, Sun BK, Kim YS, Kim J, Chang YS, Bang BK. Preconditioning with erythropoietin protects against subsequent ischemia‐reperfusion injury in rat kidney. FASEB J 17: 1754‐1755, 2003.
 617. Yang L, Besschetnova TY, Brooks CR, Shah JV, Bonventre JV. Epithelial cell cycle arrest in G2/M mediates kidney fibrosis after injury. Nat Med 16: 535‐543.
 618. Yano T, Itoh Y, Kawamura E, Maeda A, Egashira N, Nishida M, Kurose H, Oishi R. Amphotericin B‐induced renal tubular cell injury is mediated by Na+ influx through ion‐permeable pores and subsequent activation of mitogen‐activated protein kinases and elevation of intracellular Ca2+ concentration. Antimicrob Agents Chemother 53: 1420‐1426, 2009.
 619. Yasuda H, Leelahavanichkul A, Tsunoda S, Dear JW, Takahashi Y, Ito S, Hu X, Zhou H, Doi K, Childs R, Klinman DM, Yuen PST, Star RA. Chloroquine and inhibition of Toll‐like receptor 9 protect from sepsis‐induced acute kidney injury. Am J Physiol Renal Physiol 294: F1050‐F1058, 2008.
 620. Yehuda Z, Iris G. Antioxidants attenuate endotoxin‐induced acute renal failure in rats. Am J Kidney Dis 25: 51‐57, 1995.
 621. Yin M, Wheeler MD, Connor HD, Zhong Z, Bunzendahl H, Dikalova A, Samulski RJ, Schoonhoven R, Mason RP, Swenberg JA, Thurman RG. Cu/Zn‐superoxide dismutase gene attenuates ischemia‐reperfusion injury in the rat kidney. J Am Soc Nephrol 12: 2691‐2700, 2001.
 622. Yin X, Apostolov EO, Shah SV, Wang X, Bogdanov KV, Buzder T, Stewart AG, Basnakian AG. Induction of renal endonuclease G by cisplatin is reduced in DNase I‐deficient mice. J Am Soc Nephrol 18: 2544‐2553, 2007.
 623. Yoder MC. Defining human endothelial progenitor cells. J Thromb Haemost 7: 49‐52, 2009.
 624. Yokota N, Daniels F, Crosson J, Rabb H. Protective effect of T cell depletion in murine renal ischemia‐reperfusion injury. Transplantation 74: 759‐763, 2002.
 625. Yoshioka T, Fogo A, Beckman JK. Reduced activity of antioxidant enzymes underlies contrast media‐induced renal injury in volume depletion. Kidney Int 41: 1008‐1015, 1992.
 626. Ysebaert DK, De Greef KE, De Beuf A, Van Rompay AR, Vercauteren S, Persy VP, De Broe ME. T cells as mediators in renal ischemia/reperfusion injury. Kidney Int 66: 491‐496, 2004.
 627. Ysebaert DK, De Greef KE, Vercauteren SR, Ghielli M, Verpooten GA, Eyskens EJ, De Broe ME. Identification and kinetics of leukocytes after severe ischaemia/reperfusion renal injury. Nephrol Dial Transplant 15: 1562‐1574, 2000a.
 628. Ysebaert DK, De Greef KE, Vercauteren SR, Ghielli M, Verpooten GA, Eyskens EJ, De Broe ME. Identification and kinetics of leukocytes after severe ischaemia/reperfusion renal injury. Nephrol Dial Transplant 15: 1562‐1574, 2000b.
 629. Yuan H‐T, Li X‐Z, Pitera JE, Long DA, Woolf AS. Peritubular capillary loss after mouse acute nephrotoxicity correlates with down‐regulation of vascular endothelial growth factor‐A and hypoxia‐inducible factor‐1alpha. Am J Pathol 163: 2289‐2301, 2003.
 630. Zager R, Mahan J, Merola A. Effects of mannitol on the postischemic kidney. Biochemica, functinal, and morphologic assessments. Lab Invest 53: 433‐442, 1985.
 631. Zager RA. Mitochondrial free radical production induces lipid peroxidation during myohemoglobinuria. Kidney Int 49: 741‐751, 1996.
 632. Zager RA, Burkhart KM, Johnson ACM, Sacks BM. Increased proximal tubular cholesterol content: Implications for cell injury and [ldquo]acquired cytoresistance[rdquo]. Kidney Int 56: 1788‐1797, 1999.
 633. Zager RA, Foerder C, Bredl C. The influence of mannitol on myoglobinuric acute renal failure: Functional, biochemical, and morphological assessments. J Am Soc Nephrol 2: 848‐855, 1991.
 634. Zager RA, Fuerstenberg SM, Baehr PH, Myerson D, Torok‐Storb B. An evaluation of antioxidant effects on recovery from postischemic acute renal failure. J Am Soc Nephrol 4: 1588‐1597, 1994.
 635. Zager RA, Johnson A. Renal cortical cholesterol accumulation is an integral component of the systemic stress response. Kidney Int 60: 2299‐2310, 2001.
 636. Zager RA, Jurkowitz MS, Merola AJ. Responses of the normal rat kidney to sequential ischemic events. Am J Physiol 249: F148‐F159, 1985.
 637. Zager RA, Shah V, Shah H, Zager P, Johnson A, Hanson S. The mevalonate pathway during acute tubular injury: Selected determinants and consequences. Am J Pathol 161: 681‐692, 2002.
 638. Zhang B, Ramesh G, Uematsu S, Akira S, Reeves WB. TLR4 signaling mediates inflammation and tissue injury in nephrotoxicity. J Am Soc Nephrol 19: 923‐932, 2008.
 639. Zhang GH, Ichimura T, Wallin A, Kan M, Stevens JL. Regulation of rat proximal tubule epithelial cell growth by fibroblast growth factors, insulin‐like growth factor‐1 and transforming growth factor‐beta, and analysis of fibroblast growth factors in rat kidney. J Cell Physiol 148: 295‐305, 1991.
 640. Zhang X, Zheng X, Sun H, Feng B, Chen G, Vladau C, Li M, Chen D, Suzuki M, Min L, Liu W, Garcia B, Zhong R, Min W‐P. Prevention of renal ischemic injury by silencing the expression of renal caspase 3 and caspase 8. Transplantation 82: 1728‐1732, 2006.
 641. Zhang Z‐X, Wang S, Huang X, Min W‐P, Sun H, Liu W, Garcia B, Jevnikar AM. NK cells induce apoptosis in tubular epithelial cells and contribute to renal ischemia‐reperfusion injury. J Immunol 181: 7489‐7498, 2008.
 642. Zhou H, Kato A, Yasuda H, Odamaki M, Itoh H, Hishida A. The induction of heat shock protein‐72 attenuates cisplatin‐induced acute renal failure in rats. Pflugers Archiv 446: 116‐124, 2003.
 643. Zhou W, Farrar CA, Abe K, Pratt JR, Marsh JE, Wang Y, Stahl GL, Sacks SH. Predominant role for C5b‐9 in renal ischemia/reperfusion injury. J Clin Invest 105: 1363‐1371, 2000.
 644. Zhou Y, Vaidya VS, Brown RP, Zhang J, Rosenzweig BA, Thompson KL, Miller TJ, Bonventre JV, Goering PL. Comparison of kidney injury molecule‐1 and other nephrotoxicity biomarkers in urine and kidney following acute exposure to gentamicin, mercury, and chromium. Toxicol Sci 101: 159‐170, 2008.
 645. Zhuang S, Kinsey GR, Rasbach K, Schnellmann RG. Heparin‐binding epidermal growth factor and Src family kinases in proliferation of renal epithelial cells. Am J Physiol Renal Physiol 294: F459‐F468, 2008.
 646. Zhuang S, Schnellmann RG. A death‐promoting role for extracellular signal‐regulated kinase. J Pharmacol Exp Ther 319: 991‐997, 2006.
 647. Zou AP, Li N, Cowley AW Jr. Production and actions of superoxide in the renal medulla. Hypertension 37: 547‐553, 2001.
 648. Zou AP, Wu F, Cowley AW Jr. Protective effect of angiotensin II‐induced increase in nitric oxide in the renal medullary circulation. Hypertension 31: 271‐276, 1998.
 649. Zuk A, Bonventre JV, Brown D, Matlin KS. Polarity, integrin, and extracellular matrix dynamics in the postischemic rat kidney. Am J Physiol 275: C711‐C731, 1998.
 650. Zuk A, Bonventre JV, Matlin KS. Expression of fibronectin splice variants in the postischemic rat kidney. Am J Physiol Renal Physiol 280: F1037‐F1053, 2001.

Related Articles:

Top cited articles of 2018

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

David P. Basile, Melissa D. Anderson, Timothy A. Sutton. Pathophysiology of Acute Kidney Injury. Compr Physiol 2012, 2: 1303-1353. doi: 10.1002/cphy.c110041