Open access

Mechanisms of T Lymphocytes in the Damage and Repair Long Term after Renal Ischemia Reperfusion Injury

Written By

Dolores Ascon and Miguel Ascon

Submitted: 14 October 2010 Published: 23 August 2011

DOI: 10.5772/16707

From the Edited Volume

Kidney Transplantation - New Perspectives

Edited by Dr.(Md) Magdalena Trzcinska

Chapter metrics overview

2,857 Chapter Downloads

View Full Metrics

1. Introduction

Acute kidney injury (AKI) is a frequent event associated with decreased allograft survival in patients with transplanted kidneys and high mortality in patients with native kidneys (1,2). AKI is a common complication in hospitalized patients, and its incidence has risen substantially over the past 15 years (1-3). As a conservative estimate, roughly 17 million admissions annually in the United States are complicated by AKI, resulting in over $10 billion in costs to the health care system (4). Kidney transplants from living unrelated donors (not well HLA matched) with minimal ischemic injury have improved allograft survival, compared with grafts from well matched cadaveric donors with significant ischemia (5, 6). This implies that renal ischemia reperfusion injury (IRI) can have important consequences on long-term graft survival and native kidneys.

IRI is a highly complex cascade of events that includes interactions between vascular endothelium, interstitial compartments, circulating cells, and numerous mediator molecules (7). Renal ischemic injury has been found to permanently damage peritubular capillaries causing hypoxia, which may be involved in the progression of chronic renal disease after AKI (7, 8). Tubulointerstitial influx of inflammatory cells is found in many forms of chronic renal diseases, including ‘nonimmune’ diseases such as diabetes and hypertension (9). T-lymphocyte infiltration has also been observed early after moderate ischemia injury (10,11) however, the dynamics of infiltrating lymphocyte populations long term after moderate or severe ischemic injury is not very clear.

It has been demonstrated that T and B lymphocytes are important mediators in the pathogenesis of renal IRI (10, 12) however, the mechanisms by which these cells induce kidney injury is largely unknown. The trafficking of pathogenic lymphocytes into kidneys after moderate and severe ischemic injury has been postulated to contribute to kidney damage (11, 13, 14) however the physiologic state and the dynamics of trafficking of these populations long term after ischemia have not been rigorously studied. Furthermore, the activation and expression of the effector-memory phenotype by infiltrated lymphocytes suggests the possibility that these lymphocytes are responding to an injury-associated antigen (15, 16). In addition, these lymphocytes are responsible to produce inflammatory mediators not only causing local kidney structure damage, but also the severe effects on the other long distance organs, including lung, hearth, intestine, brain, liver, bone medulla.

Here we describe the trafficking of T lymphocytes into the mice (male C57BL/6J) kidneys both, in normal mice, earlier (3 to 24 h), and long term (1 to 11 weeks) after the renal injury was performed as previously described (17, 18, 19). The different T cell phenotypes and cytokine/chemokines raised at different times are compared with the baseline level cells maintained in normal kidneys (17). In the long term studies, to make our observations clinically relevant for both allograft and native kidneys, we have studied these phenomena in both a moderate bilateral ischemia (a kin to ischemia in native kidneys) and a severe unilateral ischemia (a kin to IRI in an allograft). The different kidney infiltrating T cell phenotypes and its effector molecules raised at different times after ischemia injury are presented and discussed.

Advertisement

2. Overview of experimental acute kidney injury

The mechanisms involved in renal ischemia-reperfusion injury (IRI) are complex (20, 21), invoking both innate and adaptive immunity (22, 23). Following IR, the cascade of events leading to endothelial cell dysfunction, tubular epithelial cell injury and activation of tissue-resident and infiltrating leukocytes consists of the coordinated action of cytokines/chemokines, reactive oxygen intermediates and adhesion molecules (21, 23). The early phase of innate immune response to IR begins within minutes of reperfusion, whereas the late phase adaptive response requires days to manifest. For our experiments, a well-established model of renal IRI in mice was used (17, 18, and 19).

Advertisement

3. Early trafficking of T lymphocytes into kidneys after IRI

Trafficking of CD4+ and CD8+ T lymphocytes

We have examined the trafficking of CD4+ and CD8+ T cell subsets into kidneys after ischemic injury (18). After 3 h of renal IRI, the percentages of CD4+ and CD4+NK1.1+ cells increased similarly in both sham-operated and IRI mice as compared with normal mice. However, 24 h after renal IRI, while the percentage of CD4+ T cells in the IRI mice was similar to that of control groups, the percentage of CD4+NK1.1+ cells increased (3.2%) when compared with normal (1.2%) and sham-operated (1.6%) mice. The percentage of CD8+ T cells was similar in all groups 3 and 24 h after renal IRI and no expression of NK1.1 Ag was observed on these cells. However, the increased percentage of the CD4+NK1.1+ cells in the IRI group 24 h after renal IRI could be related to renal ischemic injury because at this time point serum creatinine was increasing and visible kidney structure damage was observed. Table 1 shows summarized results.

Expression of CD69 on CD4+ and CD8+ T lymphocytes

We have investigated the activation state of the intrarenal CD4+ and CD8+ T cell subsets analyzing the expression of activation markers CD69 and CD25 (18). After 3 h of renal IRI, we observed increased expression of CD69 on CD4+ T cells in sham-operated (14.7%) and IRI (14.2%) compared with normal mice (7.1%). CD69 expression on CD8+ T cells tended to increase at 3 h, but was not statistically significant. After 24 h of renal IRI, the expression of CD69 on CD4+ and CD8+ T cells declined to lower levels than normal mice. Moreover, no increased expression of CD25 Ag on CD4+ and CD8+ T cells in any of the studied groups was found. Results demonstrated that CD4+ and CD8+ T lymphocytes infiltrating kidneys of sham-operated and IRI mice display some features of activated T lymphocytes. We hypothesized that T cells might be activated after renal IRI; however, we found a similarly increased expression of CD69 on the CD4+ and CD8+ T cells in both sham-operated and IRI mice 3 h after renal IRI. Results are summarized in Table 1.

Kidney assessment and histology changes earlier after ischemia injury

We evaluated the Ischemic kidneys following the serum creatinine levels 3 and 24 h after renal IRI (18). After 3 h of renal IRI, a significant increase in serum creatinine of IRI mice (n = 8, 1.18 mg/dl) when compared with normal (n = 8, 0.50 mg/dl) and sham-operated (n = 8, 0.70 mg/dl) mice was observed. After 24 h of renal IRI, serum creatinine significantly increased in the IRI mice (n = 8, 2.83 mg/dl) as compared with control groups. In the sham-operated mice, serum creatinine was slightly increased compared with normal mice 3 h after surgery (Fig. 1). The kidney structural injury in the cortex and the medulla of IRI mice was evaluated. Compared with kidneys of normal mice (Fig. 1A) and sham-operated mice, 3 (Fig. 1B) and 24 h (Fig. 1C) after surgery, IRI mice show slightly tubular epithelial necrosis 3 h after renal IRI (Fig. 1D) and significant tubular injury with loss of tubular structure 24 h after renal IRI (Fig. 1E).

Figure 1.

Kidney injury after 30-min bilateral ischemia. Serum creatinine of IRI mice (•) compared with normal (▴) and sham-operated (○) mice 3 and 24 h after renal IRI. A, Normal mouse kidney (no IRI). B and C, Sham-operated mice kidneys showing normal histology 3 and 24 h after surgery, respectively. D, IRI mouse kidney showing same proteinaceous casts in tubules 3 h after renal IRI. E, IRI kidney showing severe damage 24 h after renal IRI. (Pictures used with permission and courtesy of the original authors [18]).

Table 1.

T lymphocytes phenotypic trafficking into mouse kidney after IRI, expressed as cells percentages

Advertisement

4. Trafficking of T lymphocytes into kidneys long term after IRI

Trafficking of CD4+ and CD8+ T cells

Analysis of infiltrating lymphocytes long term after renal IRI (19, 24) revealed increased percentages of CD4+ (29%) and CD8+ (16%) T lymphocytes in IRI kidneys compared with kidneys of sham mice (CD4+: 11% and CD8+: 6%) after 2 weeks of bilateral renal IRI. However, similar percentage of CD4+ and CD8+ T cells was observed in sham and IRI kidneys 6 weeks after bilateral renal IRI. 6 weeks after unilateral renal IRI, we observed a significantly increased percentage of CD4+ (48%) and CD8+ (21%) T lymphocytes compared with kidneys from sham mice (CD4+: 16% and CD8+: 7%) and contralateral kidneys (CD4+: 11% and CD8+: 5%). No changes in CD4+ and CD8+ T-cell populations were observed in any of the groups 11 weeks after unilateral renal IRI. Results are summarized in Table 1. The higher levels of CD4+ and CD8+ T cells 6 and 11 weeks after ischemia as well as the return to normal levels of some populations as CD69+ and CD44+ markers after 6 weeks, demonstrate the possible limit and suppression of the immune response after long-term renal IRI. Potential modulators of this immunosuppresion could be the regulatory T cells CD4+CD25+ or CD4+CD25+ FoxP3 (25, 26) as increased populations of these regulatory T cells have been observed in long-term allogenic transplants (27).

Infiltrating of CD4+ and CD8+ T lymphocytes expressing CD69

After 2 weeks of bilateral renal IRI (19), we observed an increased expression of CD69 on CD4+ (17%) and CD8+ (9%) T cells in IRI mice when compared with sham mice (CD4+: 6% and CD8+: 2%). Similarly, increased expression of CD69 on CD4+ (22%) and CD8+ (18%) T cells in IRI mice compared with sham mice (CD4+: 15% and CD8+: 11%) was observed after 6 weeks of bilateral renal IRI. Six weeks after unilateral ischemia, we observed a significantly increased expression of CD69 on CD4+ (29%) and CD8+ (15%) T cells compared with kidneys from sham mice (CD4+: 7% and CD8+: 2%) and contralateral kidneys (CD4+: 4% and CD8+: 2%). However, 11 weeks after renal IRI, only CD4+ T cells from IRI kidneys showed increased expression of CD69 (28%) when compared with sham (13%) and contralateral (12%) kidneys. Results are summarized in Table 1. The increased infiltration of the activated CD69+ marker T lymphocytes in both unilateral and bilateral IRI kidneys, is consistent with upregulation of the early activation marker CD69 antigen observed in allograft rejections and some autoimmune diseases (2831). Activated cells produce inflammatory factors which can participate in tissue damage including fibrosis, as observed in patients with systemic sclerosis and pulmonary fibrosis (32, 33).

Infiltrating of CD4+ and CD8+ T cells displaying effector-memory phenotype

Two weeks after bilateral renal IRI (19), significantly increased percentage of effector-memory CD4+CD44hiCD62L- T cells in IRI kidneys (77%) was observed when compared with kidneys from sham mice (54%). Six weeks after bilateral renal IRI, a significantly increased percentage of CD8+ CD44hiCD62L- T cells in IRI kidneys (90%) compared with sham mice (79%) was observed. Similarly, 6 weeks after unilateral renal IRI, the IRI kidneys showed significantly increased percentage of CD4+CD44hiCD62L- T cells (96%) when compared with kidneys from sham mice (71%) and contralateral kidneys (65%). A significant increase in percentage of CD4+CD44hiCD62L- T cells was observed in IRI kidneys (93%) when compared with sham (80%) and contralateral kidneys (75%) 11 weeks after renal IRI. Results are summarized in Table 1. The high levels of effector-memory CD4+CD44hiCD62L- T cells, the ‘footprints’ of an immune response to antigens, in both unilateral and bilateral IRI kidneys, are consistent with the response to self-antigens involved in the pathogenesis of skeletal and intestinal ischemia induced by hypoxic stress (34), indicating that immune response to renal IRI could be also initiated by specific antigens.

Decreasing of NKT lymphocytes

Similar percentage of NKT cells (CD4+NK1.1+) was observed after 2 weeks of bilateral renal IRI (19). However, 6 weeks after bilateral renal IRI, we found a significantly decreased percentage of NKT cells in IRI kidneys (2%) when compared with kidneys of sham mice (4%). Eleven weeks after unilateral renal IRI, decreased percentage of NKT cells was observed in IRI kidneys (0.4%) when compared to sham (1.91%) and contralateral kidneys (3.1% ). However, no changes were observed in mice that underwent bilateral renal IRI with reduced ischemia times. In Table 1 are summarized the results. The decreased number of NKT cells 6 and 11 weeks after bilateral and unilateral renal IRI, respectively, are similar to that in liver injury (35) and rheumatoid arthritis (36).

Effect of CD+ and CD8+ T-cell depletion on kidney-cell infiltration

To determine the pathophysiologic role of infiltrating CD4+ and CD8+ T cells long term after ischemia, we depleted these cells before and after unilateral ischemia during the 6-week experiments (19). Depletion started 24 h preischemia and 3 days postischemia and cell analysis by flow cytometry was performed weekly in blood and after 6 weeks in kidney samples. Blood was 98% depleted of CD4+ and CD8+ T cells during the 6 weeks after renal IRI. In kidneys, the CD4+CD69+, CD8+CD69+, CD4+CD44hiCD62L-, and CD4+NK1.1+ cells were also depleted by approximately 98%, in relationship with the cell profiles of nondepleted control mice (data are not showed).

Histology of structural damage after long term ischemia

To observe the degree of structural damage of ischemic kidneys after 6 weeks of renal IRI in depleted mice, the kidney histology of depleted and control mice were compared. The damage in the cortex (Figure 2a) was similar in control and both depleted mice, however, medullary damage (Figure 2b) was more extensive in control and post-ischemia depleted mice (Figure 2c) than in preischemia depleted mice. Therefore, the reduced damage observed in the kidney medulla of preischemia depleted mice when compared to control mice could be related to the low expression of IFN-γ (Table 2). The IFN-γ produced by CD4+ and CD8+ T lymphocytes is involved early after renal ischemia (37, 38), and has been detected in acute and chronic kidney rejections (39). However, the increased expression of IL-1β in postischemia depleted mice could be related to the increased structural damage of kidney observed and could have distant organ affects (40).

Figure 2.

Kidney tissue from IRI mice after 2 weeks of 25 min of bilateral ischemia (a, upper panel) shows some proteinaceous casts in tubules compared with normal histology of normal and sham mouse kidneys. Kidney structure 6 weeks after unilateral renal IRI (b, lower panel) shows normal histology of sham and contralateral kidneys compared with severe kidney damage, loss of structure, and cyst formation in IRI kidneys. (Pictures used with permission and courtesy of the original authors [19]).

Regulatory T (Treg) cells involved in damage inhibition and reparative phase

Treg cells are lymphocytes with immunosuppressive properties. One important subset of Treg cells express CD4 and CD25 on the cell surface and the transcription factor, FoxP3 (41). The mechanisms of suppression by Treg cells are diverse and include: production of antiinflammatory cytokines such as IL-10 or TGF-β, direct cell-cell contact or CTLA-4 mediated inhibition and production of extracellular adenosine (42). Recently, Treg cells have been identified in normal mouse kidneys (17, 43). In WT mice, treatment with an anti-CD25 monoclonal antibody (PC61) selectively decreased kidney, spleen and blood CD4+ FoxP3+ Treg cell numbers by approximately 50%, five days after PC61 treatment (44). At that time point, Treg cell deficiency potentiated kidney IRI, measured by plasma creatinine, acute tubular necrosis (ATN), neutrophil and macrophage accumulation and pro-inflammatory cytokine transcription in the kidney after 24 hr of reperfusion (43). In lymphocyte-deficient Rag-1 KO mice, adoptive transfer of WT, but not IL-10 KO, Treg cells blocked IR-induced inflammation and kidney injury (43). These findings demonstrate that Treg cells can directly suppress the early innate inflammation, induced by IR, in an IL-10 dependent manner. In a different study, PC61 was administered 1 day prior to IRI, and while BUN levels and ATN scores were no different than control antibody-treated mice at 24 hr of reperfusion, the necrosis failed to resolve by 72 hr in the PC61-treated mice (45). In other study, using a murine model of ischemic acute kidney injury it was found that the percentage of the CD25+Foxp3+ Treg subset in the total kidney-infiltrating TCRβ+CD4+ T lymphocyte compartment was increased from 1.8 to 2.6% in IR kidneys at 3 and 10 days (46). This infiltration was accompanied of an enhanced pro-inflammatory cytokine production. These results strongly support an important role of regulatory T cells during IRI and in kidney repair after IRI.

Table 2.

Cytokines and chemokines expressed after IRI in kidney.

Advertisement

5. Upregulation of cytokines and chemokines long term after IRI

Expression of cytokines

Cytokine and chemokines are known to modulate lymphocyte and kidney cell interactions to mediate kidney injury and fibrosis. We found (19) an increased intracellular cytokine production of TNF-α and IFN-γ by CD3T+ cells infiltrating kidneys after 24 hours of IRI in mice. This observation suggests that lymphocytes infiltrating into the postischemic kidneys could have a major downstream effect on later inflammation and organ dysfunction. Thus, not only the trafficking of T cells postischemia is a potential mechanism, but what those infiltrating cells are doing at the site of injury could be crucial for pathogenesis. Given that infiltrating T cells are activated and selectively expanded in kidney long term after IRI, we hypothesized that there would be a different upregulation of these molecules postischemia in depleted and nondepleted mice. Using real-time RT-PCR, a significant upregulation of IL-1β, IL-6, tumor necrosis factor (TNF)-α, IFN-γ, MIP-2, and RANTES was seen 6 weeks after 60 min of unilateral renal IRI in normal (nondepleted T cells), compared to sham and contralateral kidneys. Depletion of CD4 and CD8 T cells starting preischemia led to significant decrease in kidney IFN-γ levels. In contrast, depletion starting 3 days after ischemia led to significant increase in IL-1β. However, the IRI kidneys of both depleted and nondepleted groups had prominent expression levels of TNF-α and RANTES. As demonstrated in both depleted and nondepleted mice 6 weeks after unilateral ischemia, the cytokines and chemokines including IL-1β, IL-6, TNF-α, MIP-2, and RANTES were significantly upregulated. The results are summarized in the Table 2. It has been reported that in moderate ischemia a modest upregulation of TNF-α and RANTES and strong upregulation of IL-1β, IL-6, IFN-γ, and MIP-2 exist (47), whereas after severe ischemia strong upregulation of TNF-α and RANTES and to a lesser extent IL-1β, IL-6, IFN-γ, and MIP-2 occur (48, 49). Similarly, in patients with acute rejection and chronic allograft nephropathy significant expression of TNF-α and RANTES were reported (49).

Expression of CXC and CC chemokines

Chemokines are mainly known for their ability to attract inflammatory cells to sites of injury. Recently, the highest levels of chemokine expression at the stage of active repair (i.e. 7 days after ischemic injury) was observed, and temporal chemokines expression pattern in more detail was examinated (50). The expression of the CC and CXC chemokines at additional reperfusion periods after ischemic injury was evaluated to determine if there is a biphasic expression coinciding with the inflammatory and reparative response after ischemic injury. Some chemokine results are summarized in the Table 2. The four CC chemokines were expressed in a monophasic fashion with a clear peak 7 days after ischemic injury. In contrast, the CXC chemokines had a biphasic expression after ischemic injury with the first peak in the early (i.e. inflammatory) phase and the second peak during the reparative phase. The CXC chemokines Cxcl1/KC, Cxcl2/MIP-2a and Cxcl10/IP-10 had the highest expression during the inflammatory phase.

Advertisement

6. Effect of renal ischemic injury on distant organs

Acute kidney injury (AKI) in native kidneys is a major clinical problem with high mortality and morbidity in the intensive care unit. This problem remains unchanged for the past 50 years in part because AKI is associated with extra-renal complications (51, 52, 53). Much of the increased risk of death associated with AKI is usually related to multi-organ dysfunction including brain, heart, lungs, liver and small intestine. After kidney IRI, inflammatory cytokines and chemokines in plasma IL-1β, IL-6, KC (IL-8), TNF-α, TNF-β, INF-γ, IL-17A, C5a, and MCP-1 increased significantly which eventually could lead to develop multi-organ failure. (54, 55, 56). In particular, AKI caused by IRI increased pulmonary vascular permeability with capillary leak (57) and change of fluid absorption in alveolar epithelial cells (58). Inflammation and apoptosis could be important mechanisms connecting the effect of AKI on lung and distant organs as show in changes of inflammatory transcriptome identified in lung after kidney IRI (59). Studies using gene microarrays analysis found marked changes in immune, inflammatory, and apoptotic processes (60). Caspase-dependent pulmonary apoptosis concurrent with activated T cell trafficking was also demonstrated in kidney after IRI (61). Altered gene expression associated with inflammation, apoptosis, and cytoskeletal structure in pulmonary endothelial cells after kidney IRI suggested possible mechanisms underlying the increased pulmonary microvascular permeability (62). Increase of IL-1β, IL-6, TNFα, MCP-1, KC (IL-8) and ICAM-1 may act as mediators in the crosstalk between kidney and lung (55, 60, 63). AKI following

Figure 3.

AKI induce distant organ effects. AKI leads to changes in distant organs, including brain, lungs, heart, liver, and small intestine, involving multiple inflammatory pathways, including increased expression of soluble pro-inflammatory mediators, innate and adaptive immunity, cellular apoptosis, microvascular inflammation and dysregulation of transport activity, oxidative stress, transcriptional responses, etc.

IRI has been reported to increase apoptosis and production of IL-1, TNF-α, and ICAM-1 in cardiac tissue (56). Changes in the microvasculature after kidney IRI were also demonstrated in brain and conferred susceptibility to stroke (64). In brain has been found increased expression of KC (IL-8), granulocyte colony-stimulating factor (G-CSF), and glial fibrillary acidic protein, an inflammatory marker (65). More recently, hepatic and small intestine dysfunction has been observed in patients suffering from AKI. Liver injury after ischemic shows peri-portal hepatocyte vacuolization, necrosis and apoptosis with inflammatory changes. Small intestinal injury after ischemic was characterized by villous lacteal capillary endothelial apoptosis, epithelial necrosis and increased leukocyte (neutrophils, macrophages and lymphocytes) infiltration. Vascular permeability was severely impaired in both liver and small intestine. After ischemic insult TNF-α, IL-17A and IL-6 levels in plasma, liver and small intestine increased significantly. Furthermore, up-regulation of KC (IL-8), MCP-1, MIP-2, ICAM-1 has been found in liver and small intestine (54). The Figure 3, shows a summarized picture of the cross talking between AKI and several long distance organs.

References

  1. 1. TilneyN. L.GuttmannR. D.1997Effects of initial ischemia/reperfusion injury on the transplanted kidney. Transplantation 64945947
  2. 2. BonventreJ. V.ZukA.2004Ischemic acute renal failure: An inflammatory disease? Kidney Int 66: 480-485,
  3. 3. TerasakiP. I.CeckaJ. M.GjertsonD. W.et al.1995survival rates of kidney transplants from spousal and living unrelated donors. N Engl J Med ; 333333336
  4. 4. ChertowG. M.BurdickE.HonourM.BonventreJ. V.BatesD. W.2005Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. JAmSoc Nephrol. 161133653370
  5. 5. SanfilippoF.VaughnW. K.SpeesE. K.et al.1984The detrimental effects of delayed graft function in cadaver donor renal transplantation. Transplantation 38643648
  6. 6. HalloranP. F.AprileM. A.FarewellV.et al.1988Early function as the principal correlate of graft survival. A multivariate analysis of 200 cadaveric renal transplants treated with a protocol incorporating antilymphocyte globulin and cyclosporine. Transplantation ; 46223228
  7. 7. BasileD. P.DonohoeD.RoetheK.et al.2001Renal ischemia injury results in permanent damage to peritubular capillaries and influences long-term function. Am J Physiol Renal Physiol ; 281: F887F899.
  8. 8. BasileD. P.DonohoeD. L.RoetheK.et al.2003Chronic renal hypoxia after acute ischemic injury: effects of L-arginine on hypoxia and secondary damage. Am J Physiol Renal Physiol ; 284: F338F348.
  9. 9. RemuzziG.BertaniT.1998Pathophysiology of progressive nephropathies. N Engl J Med ; 33914481456
  10. 10. BurneM. J.DanielsF.El GhandourA.et al.2001Identification of the CD4(+) T cell as a major pathogenic factor in ischemic acute renal failure. J Clin Invest ; 10812831290
  11. 11. PinheiroH. S.CamaraN. O.NoronhaI. L.MaugeriI. L.FrancoM. F.MedinaJ. O.Pacheco-SilvaA.2007Contribution of CD4+ T cells to the early mechanisms of ischemia- reperfusion injury in a mouse model of acute renal failure. Braz J Med Biol Res. 4055768
  12. 12. Burne-TaneyM. J.AsconD. B.DanielsF.et al.2003B cell deficiency confers protection from renal ischemia reperfusion injury. J Immunol ; 17132103215
  13. 13. Burne-TaneyM.YokotaN.RabbH.2005Persistent renal and extra renal changes long term after severe renal ischemia reperfusion injury. Kidney Int ; 6710021009
  14. 14. IbrahimS.JacobsF.ZukinY.et al.1996Immunohistochemical manifestations of unilateral kidney ischemia. Clin Transplant ; 10646652
  15. 15. BriscoeD. M.SayeghM. H.2002A rendezvous before rejection: where do T cells meet transplant antigens? Nat Med ; 8220222
  16. 16. ZhangM.AustenW. G. J.ChiuI.et al.2004Identification of a specific self-reactive IgM antibody that initiates intestinal ischemia/reperfusion injury. Proc Natl Acad Sci USA ; 10138863891
  17. 17. AsconD. B.AsconM.SatputeS.Lopez-BrionesS.RacusenL.ColvinR. B.SoloskiM. J.RabbH.2008Normal mouse kidneys contain activated and CD3+CD4- CD8- double-negative T lymphocytes with a distinct TCR repertoire. J Leukoc Biol. ; 8414001409
  18. 18. AsconD. B.Lopez-BrionesS.LiuM.et al.2006Phenotypic and functional characterization of kidney-infiltrating lymphocytes in renal ischemia reperfusion injury. J Immunol 17733803387
  19. 19. AsconM.AsconD. B.LiuM.CheadleC.SarkarC.RacusenL.HassounH. T.RabbH.2009Renal ischemia-reperfusion leads to long term infiltration of activated and effector-memory T lymphocytes. Kidney Int. ;75526535
  20. 20. SchrierR. W.WangW.PooleB.MitraA.2004Acute renal failure: definitions, diagnosis, pathogenesis, and therapy. J Clin Invest ;114514
  21. 21. BonventreJ.V. WeinbergJ.M. 2003., Recent advances in the pathophysiology of ischemic acute renal failure. J Am Soc Nephrol ;142199210
  22. 22. RabbH. 2002. ;The T cell as a bridge between innate and adaptive immune systems: implications for the kidney. Kidney Int, 61193546
  23. 23. LiL.OkusaM. D.2006Blocking the Immune respone in ischemic acute kidney injury: the role of adenosine 2A agonists. Nat Clin Pract Nephrology ;243244
  24. 24. IbrahimS.JacobsF.ZukinY.et al.1996Immunohistochemical manifestations of unilateral kidney ischemia. Clin Transplant ; 10646652
  25. 25. SarweenN.ChodosA.RaykundaliaC.et al.2004CD4+CD25+ cells controlling a pathogenic CD4 response inhibit cytokine differentiation, CXCR-3 expression, and tissue invasion. J Immunol ; 17329422951
  26. 26. MurphyT. J.NiN.ChoileainY.Zanget.al2005CD4+CD25+ regulatory T cells control innate immune reactivity after injury. J Immunol ; 17429572963
  27. 27. BraudeauC.RacapeM.GiralM.et al.2007Variation in numbers of CD4+CD25highFOXP3+ T cells with normal immuno-regulatory properties in long-term graft outcome. Transpl Int ; 20845855
  28. 28. AfeltraA. M.GaleazziG. D.SebastianiG. M.et al.1997Coexpression of CD69 and HLADR activation markers on synovial fluid T lymphocytes of patients affected by rheumatoid arthritis: a three-colour cytometric analysis. Int J Exp Pathol ; 78331336
  29. 29. SantamariaM.MarubayashiM.ArizonJ. M.et al.1992The activation antigen CD69 is selectively expressed on CD8+ endomyocardium infiltrating T lymphocytes in human rejecting heart allografts. Hum Immunol ; 3314
  30. 30. CrispinJ. C.MartinezA.de PabloP.et al.1998Participation of the CD69 antigen in the T-cell activation process of patients with systemic lupus erythematosus. Scand J Immunol 48196200
  31. 31. PosseltA. M.VincentiF.BedolliM.et al.2003CD69 expression on peripheral CD8 T cells correlates with acute rejection in renal transplant recipients. Transplantation ; 76190195
  32. 32. BresserP.JansenH. M.WellerF. R.et al.2001T-cell activation in the lungs of patients with systemic sclerosis and its relation with pulmonary fibrosis. Chest ; 120: 66S EOF68S EOF
  33. 33. LuzinaI.G. AtamasS.P. WiseR. et al. 2003., Occurrence of an activated, profibrotic pattern of gene expression in lung CD8+ T cells from scleroderma patients. Arthritis Rheum ; 4822622274
  34. 34. ZhangM.AlicotE. M.ChiuI.et al.2006Identification of the target self-antigens in reperfusion injury. J Exp Med ; 203141152
  35. 35. ShimamuraK.KawamuraH.NaguraT.et al.2005Association of NKT cells and granulocytes with liver injury after reperfusion of the portal vein. Cell Immunol ; 2343138
  36. 36. YanagiharaY.ShiozawaK.TakaiM.et al.1999Natural killer (NK) T cells are significantly decreased in the peripheral blood of patients with rheumatoid arthritis (RA). Clin Exp Immunol ; 118131136
  37. 37. LiL.HuangL.SungS. S.et al.2007NKT cell activation mediates neutrophil IFN-gamma production and renal ischemia-reperfusion injury. J Immunol 17858995911
  38. 38. DayY. J.HuangL.YeH.et al.2006Renal ischemia-reperfusion injury and adenosine 2A receptor-mediated tissue protection: the role of CD4+ T cells and IFN-gamma. J Immunol ; 17631083114
  39. 39. ObataF.YoshidaK.OhkuboM.et al.2005Contribution of CD4+ and CD8+ T cells and interferon-gamma to the progress of chronic rejection of kidney allografts: the Th1 response mediates both acute and chronic rejection. Transpl Immunol ; 142125
  40. 40. KellyK. J.2003Distant effects of experimental renal ischemia/reperfusion injury. J Am Soc Nephrol ; 1415491558
  41. 41. FontenotJ. D.GavinM. A.RudenskyA. Y.2003Foxp3 programs the development and function of CD4 +CD25+ regulatory T cells. Nat Immunol ;43306
  42. 42. ShevachE. M.2009Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity 3063645
  43. 43. KinseyG. R.SharmaR.HuangL.LiL.VergisA. L.YeH.et al.2009Regulatory T Cells Suppress Innate Immunity in Kidney Ischemia-Reperfusion Injury. J Am Soc Nephrol 20174453
  44. 44. KinseyG. R.HuangL.VergisA. L.LiL.OkusaM. D.2010Regulatory T cells contribute to the protective effect of ischemic preconditioning in the kidney. Kidney Int. 7777180
  45. 45. MonteiroR. M.CamaraN. O.RodriguesM. M.TzelepisF.DamiaoM. J.CenedezeM. A.et al.2009A role for regulatory T cells in renal acute kidney injury. Transpl Immunol 215055
  46. 46. GandolfoM. T.JangH. R.BagnascoS. M.KoG. J.AgredaP.SatputeS. R.CrowM. T.KingL. S.RabbH.2009Foxp3+ regulatory T cells participate in repair of ischemic acute kidney injury. Kidney Int. 76717729
  47. 47. HribovaP.KotschK.BrabcovaI.et al.2005Cytokines and chemokine gene expression in human kidney transplantation. Transplant Proc 37760763
  48. 48. AtamasS. P.2002Complex cytokine regulation of tissue fibrosis. Life Sci 72: 631 EOF43 EOF
  49. 49. LemayS.RabbH.PostlerG.et al.2000Prominent and sustained up-regulation of gp130-signaling cytokines and the chemokine MIP-2 in murine renal ischemia-reperfusion injury. Transplantation ; 69959963
  50. 50. StrooI.StokmanG.TeskeG. J.RavenA.ButterL. M.FlorquinS.LeemansJ. C.2010Chemokine expression in renal ischemia/reperfusion injury is most profound during the reparative phase. Int Immunol. ; 2243342
  51. 51. ChertowG. M.LevyE. M.HammermeisterK. E.GroverF.DaleyJ.1998Independent association between acute renal failure and mortality following cardiac surgery. Am J Med. ;1043438
  52. 52. PalevskyP. M.ZhangJ. H.O’ConnorT. Z.ChertowG. M.CrowleyS. T.ChoudhuryD.et al.2008Intensity of renal support in critically ill patients with acute kidney injury. N Engl J Med. ;359720
  53. 53. JonesD. R.LeeH. T.2008Perioperative renal protection. Best Pract Res Clin Anaesthesiol ;22193208
  54. 54. ParkS. W.ChenS. W. C.KimM.BrownK. M.KollsJ. K.D’AgatiV. D.LeeH. T.2011Cytokines induce small intestine and liver injury after renal ischemia or nephrectomy. Laboratory Investigation 916384
  55. 55. CampanholleG. LandgrafR.G. GonçalvesG.M. PaivaV.N. MartinsJ.O. WangP.H.MonteiroR.M. SilvaR.C. CenedezeM.A. TeixeiraV.P. ReisM.A. Pacheco-SilvaA. JancarS. CamaraN.O. 2010., Lung inflammation is induced by renal ischemia and reperfusion injury as part of the systemic inflammatory syndrome. Inflamm Res , 10598619
  56. 56. KellyK. J.2003Distant effects of experimental renal ischemia/reperfusion injury. J Am Soc Nephrol. 14154958
  57. 57. KramerA.A. PostlerG. SalhabK.F. MendezC.CareyL.C. RabbH.1999., Renal ischemia/reperfusion leads to macrophage-mediated increase in pulmonary vascular permeability. Kidney Int. ;55:2362-7
  58. 58. RabbH.WangZ.NemotoT.HotchkissJ.YokotaN.SoleimaniM.2003Acute renal failure leads to dysregulation of lung salt and water channels. Kidney Int. ;636006
  59. 59. HassounH. T.GrigoryevD. N.LieM. L.LiuM.CheadleC.TuderR. M.et al.2007Ischemic acute kidney injury induces a distant organ functional and genomic response distinguishable from bilateral nephrectomy. Am J Physiol Renal Physiol. ;293:F3040
  60. 60. GrigoryevD. N.LiuM.HassounH. T.CheadleC.BarnesK. C.RabbH.2008The local and systemic inflammatory transcriptome after acute kidney injury. J Am Soc Nephrol. ;1954758
  61. 61. HassounH. T.LieM. L.GrigoryevD. N.LiuM.TuderR. M.RabbH.2009Kidney ischemia-reperfusion injury induces caspase-dependent pulmonary apoptosis. Am J Physiol Renal Physiol. ;297:F12537
  62. 62. FeltesC.RabbH. 2009. : Acute kidney injury leads to pulmonary endothelial cell transcriptional, cytoskeletal and apoptotic changes. ASN renal week 2009, San Diego
  63. 63. HokeT. S.DouglasI. S.KleinC. L.HeZ.FangW.ThurmanJ. M.TaoY.et al.2007Acute renal failure after bilateral nephrectomy is associated with cytokine-mediated pulmonary injury. J Am Soc Nephrol 18: 155-164,
  64. 64. LiuM.StinsM. SaleemS. DoreS.RabbH. 2009. : Acute kidney injury disrupts blood brain barrier and increases susceptibility to stroke. ASN renal week, San Diego; 2009
  65. 65. LiuM.LiangY.ChigurupatiS.LathiaJ. D.PletnikovM.SunZ.et al.2008Acute kidney injury leads to inflammation and functional changes in the brain. J Am Soc Nephrol. ;19136070

Written By

Dolores Ascon and Miguel Ascon

Submitted: 14 October 2010 Published: 23 August 2011