51
|
Situmorang GR, Sheerin NS. Ischaemia reperfusion injury: mechanisms of progression to chronic graft dysfunction. Pediatr Nephrol 2019; 34:951-963. [PMID: 29603016 PMCID: PMC6477994 DOI: 10.1007/s00467-018-3940-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/18/2018] [Accepted: 03/02/2018] [Indexed: 12/18/2022]
Abstract
The increasing use of extended criteria organs to meet the demand for kidney transplantation raises an important question of how the severity of early ischaemic injury influences long-term outcomes. Significant acute ischaemic kidney injury is associated with delayed graft function, increased immune-associated events and, ultimately, earlier deterioration of graft function. A comprehensive understanding of immediate molecular events that ensue post-ischaemia and their potential long-term consequences are key to the discovery of novel therapeutic targets. Acute ischaemic injury primarily affects tubular structure and function. Depending on the severity and persistence of the insult, this may resolve completely, leading to restoration of normal function, or be sustained, resulting in persistent renal impairment and progressive functional loss. Long-term effects of acute renal ischaemia are mediated by several mechanisms including hypoxia, HIF-1 activation, endothelial dysfunction leading to vascular rarefaction, sustained pro-inflammatory stimuli involving innate and adaptive immune responses, failure of tubular cells to recover and epigenetic changes. This review describes the biological relevance and interaction of these mechanisms based on currently available evidence.
Collapse
Affiliation(s)
- Gerhard R Situmorang
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Urology Department, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, 10430, Indonesia
| | - Neil S Sheerin
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
52
|
Roles of Natural Killer T Cells and Natural Killer Cells in Kidney Injury. Int J Mol Sci 2019; 20:ijms20102487. [PMID: 31137499 PMCID: PMC6567827 DOI: 10.3390/ijms20102487] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/30/2023] Open
Abstract
Mouse natural killer T (NKT) cells and natural killer (NK) cells are innate immune cells that are highly abundant in the liver. In addition to their already-known antitumor and antimicrobial functions, their pathophysiological roles in the kidney have recently become evident. Under normal circumstances, the proportion of activated NKT cells in the kidney increases with age. Administration of a synthetic sphingoglycolipid ligand (alpha-galactosylceramide) further activates NKT cells, resulting in injury to renal vascular endothelial cells via the perforin-mediated pathway and tubular epithelial cells via the TNF-α/Fas ligand pathway, causing acute kidney injury (AKI) with hematuria. Activation of NKT cells by common bacterial DNA (CpG-ODN) also causes AKI. In addition, NKT cells together with B cells play significant roles in experimental lupus nephritis in NZB/NZW F1 mice through their Th2 immune responses. Mouse NK cells are also assumed to be involved in various renal diseases, and there may be complementary roles shared between NKT and NK cells. Human CD56+ T cells, a functional counterpart of mouse NKT cells, also damage renal cells through a mechanism similar to that of mice. A subpopulation of human CD56+ NK cells also exert strong cytotoxicity against renal cells and contribute to the progression of renal fibrosis.
Collapse
|
53
|
Turner JE, Rickassel C, Healy H, Kassianos AJ. Natural Killer Cells in Kidney Health and Disease. Front Immunol 2019; 10:587. [PMID: 30972076 PMCID: PMC6443628 DOI: 10.3389/fimmu.2019.00587] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/05/2019] [Indexed: 02/02/2023] Open
Abstract
Natural killer (NK) cells are a specialized population of innate lymphocytes that have a major effector function in local immune responses. While their immunological functions in many inflammatory diseases are well established, comparatively little is still known about their roles in kidney homeostasis and disease. Our understanding of kidney NK cells is rapidly evolving, with murine studies highlighting the functional significance of NK cells in acute and chronic forms of renal disease. Recent progress has been made in translating these murine findings to human kidneys, with indications of NK cell subset-specific roles in disease progression in both native and allograft kidneys. Clearly, a better understanding of the molecular mechanisms driving NK cell activation and importantly, their downstream interactions with intrinsic renal cells and infiltrating immune cells is necessary for the development of targeted therapeutics to halt disease progression. In this review, we discuss the properties and potential functions of kidney NK cells.
Collapse
Affiliation(s)
- Jan-Eric Turner
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Constantin Rickassel
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Helen Healy
- Conjoint Kidney Research Laboratory, Chemical Pathology-Pathology Queensland, Brisbane, QLD, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Andrew J Kassianos
- Conjoint Kidney Research Laboratory, Chemical Pathology-Pathology Queensland, Brisbane, QLD, Australia.,Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| |
Collapse
|
54
|
In the absence of natural killer cell activation donor-specific antibody mediates chronic, but not acute, kidney allograft rejection. Kidney Int 2018; 95:350-362. [PMID: 30503624 DOI: 10.1016/j.kint.2018.08.041] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 08/02/2018] [Accepted: 08/23/2018] [Indexed: 11/20/2022]
Abstract
Antibody mediated rejection (ABMR) is a major barrier to long-term kidney graft survival. Dysregulated donor-specific antibody (DSA) responses are induced in CCR5-deficient mice transplanted with complete major histocompatibility complex (MHC)-mismatched kidney allografts, and natural killer (NK) cells play a critical role in graft injury and rejection. We investigated the consequence of high DSA titers on kidney graft outcomes in the presence or absence of NK cell activation within the graft. Equivalent serum DSA titers were induced in CCR5-deficient B6 recipients of complete MHC mismatched A/J allografts and semi-allogeneic (A/J x B6) F1 kidney grafts, peaking by day 14 post-transplant. A/J allografts were rejected between days 16-28, whereas B6 isografts and semi-allogeneic grafts survived past day 65. On day 7 post-transplant, NK cell infiltration into A/J allografts was composed of distinct populations expressing high and low levels of the surface antigen NK1.1, with NK1.1low cells reflecting the highest level of activation. These NK cell populations increased with time post-transplant. In contrast, NK cell infiltration into semi-allogeneic grafts on day 7 was composed entirely of NK1.1high cells that decreased thereafter. On day 65 post-transplant the semi-allogeneic grafts had severe interstitial fibrosis, glomerulopathy, and arteriopathy, accompanied by expression of pro-fibrogenic genes. These results suggest that NK cells synergize with DSA to cause acute kidney allograft rejection, whereas high DSA titers in the absence of NK cell activation cannot provoke acute ABMR but instead induce the indolent development of interstitial fibrosis and glomerular injury that leads to late graft failure.
Collapse
|
55
|
Coelho S, Cabral G, Lopes JA, Jacinto A. Renal regeneration after acute kidney injury. Nephrology (Carlton) 2018; 23:805-814. [PMID: 29575280 DOI: 10.1111/nep.13256] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2018] [Indexed: 12/28/2022]
Abstract
Acute kidney injury is common and associated with negative renal and patient outcomes. The human kidney has a real but limited regeneration capacity. Understanding renal regeneration may allow us to manipulate this process and thus develop therapeutic weapons to improve patients' outcome. In the first part of this paper we discuss the clinical factors associated with renal recovery: baseline patient particularities, acute kidney injury characteristics and the medical approach taken in the short and long-term. In the second part, the cellular and molecular mechanisms underlying renal regeneration are explored. The immune system seems to have an important role, first promoting inflammation and then tissue healing. Other players, such as cellular senescence, mitochondrial dysfunction, renal haemodynamics and metabolic reprogramming also have a role in renal regeneration. We aim to develop a short review of renal regeneration, offering a holistic view of this process.
Collapse
Affiliation(s)
- Silvia Coelho
- Intensive Care Department, Hospital Fernando Fonseca, EPE, Amadora, Portugal.,CEDOC - Chronic Diseases Research Center, NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| | - Guadalupe Cabral
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| | - José A Lopes
- Division of Nephrology and Renal Transplantation, Department of Medicine Centro Hospitalar Lisboa Norte, EPE, Lisbon, Portugal
| | - António Jacinto
- CEDOC - Chronic Diseases Research Center, NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| |
Collapse
|
56
|
Ferhat MH, Robin A, Barbier L, Thierry A, Gombert JM, Barbarin A, Herbelin A. The Impact of Invariant NKT Cells in Sterile Inflammation: The Possible Contribution of the Alarmin/Cytokine IL-33. Front Immunol 2018; 9:2308. [PMID: 30374349 PMCID: PMC6197076 DOI: 10.3389/fimmu.2018.02308] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/17/2018] [Indexed: 01/13/2023] Open
Abstract
Although the contribution of iNKT cells to induction of sterile inflammation is now well-established, the nature of the endogenous compounds released early after cellular stress or damage that drive their activation and recruitment remains poorly understood. More precisely, iNKT cells have not been described as being reactive to endogenous non-protein damage-associated molecular-pattern molecules (DAMPs). A second subset of DAMPs, called alarmins, are tissue-derived nuclear proteins, constitutively expressed at high levels in epithelial barrier tissues and endothelial barriers. These potent immunostimulants, immediately released after tissue damage, include the alarmin IL-33. This factor has aroused interest due to its singular action as an alarmin during infectious, allergic responses and acute tissue injury, and as a cytokine, contributing to the latter resolutive/repair phase of sterile inflammation. IL-33 targets iNKT cells, inducing their recruitment in an inflammatory state, and amplifying their regulatory and effector functions. In the present review, we introduce the new concept of a biological axis of iNKT cells and IL-33, involved in alerting and controlling the immune cells in experimental models of sterile inflammation. This review will focus on acute organ injury models, especially ischemia-reperfusion injury, in the kidneys, liver and lungs, where iNKT cells and IL-33 have been presumed to mediate and/or control the injury mechanisms, and their potential relevance in human pathophysiology.
Collapse
Affiliation(s)
| | | | - Louise Barbier
- Service de Chirurgie Digestive, Oncologique, Endocrinienne et Transplantation Hépatique, CHU Trousseau, Université de Tours, Tours, France
| | - Antoine Thierry
- INSERM U1082 - IRATI Group, Poitiers, France.,Service de Néphrologie, Hémodialyse et Transplantation Rénale, CHU de Poitiers, Poitiers, France
| | - Jean-Marc Gombert
- INSERM U1082 - IRATI Group, Poitiers, France.,Service d'Immunologie et d'Inflammation, CHU de Poitiers, Poitiers, France
| | | | | |
Collapse
|
57
|
Kundert F, Platen L, Iwakura T, Zhao Z, Marschner JA, Anders HJ. Immune mechanisms in the different phases of acute tubular necrosis. Kidney Res Clin Pract 2018; 37:185-196. [PMID: 30254843 PMCID: PMC6147180 DOI: 10.23876/j.krcp.2018.37.3.185] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/25/2018] [Indexed: 12/18/2022] Open
Abstract
Acute kidney injury is a clinical syndrome that can be caused by numerous diseases including acute tubular necrosis (ATN). ATN evolves in several phases, all of which are accompanied by different immune mechanisms as an integral component of the disease process. In the early injury phase, regulated necrosis, damage-associated molecular patterns, danger sensing, and neutrophil-driven sterile inflammation enhance each other and contribute to the crescendo of necroinflammation and tissue injury. In the late injury phase, renal dysfunction becomes clinically apparent, and M1 macrophage-driven sterile inflammation contributes to ongoing necroinflammation and renal dysfunction. In the recovery phase, M2-macrophages and anti-inflammatory mediators counteract the inflammatory process, and compensatory remnant nephron and cell hypertrophy promote an early functional recovery of renal function, while some tubules are still badly injured and necrotic material is removed by phagocytes. The resolution of inflammation is required to promote the intrinsic regenerative capacity of tubules to replace at least some of the necrotic cells. Several immune mechanisms support this wound-healing-like re-epithelialization process. Similar to wound healing, this response is associated with mesenchymal healing, with a profound immune cell contribution in terms of collagen production and secretion of profibrotic mediators. These and numerous other factors determine whether, in the chronic phase, persistent loss of nephrons and hyperfunction of remnant nephrons will result in stable renal function or progress to decline of renal function such as progressive chronic kidney disease.
Collapse
Affiliation(s)
- Fedor Kundert
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Louise Platen
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Takamasa Iwakura
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Zhibo Zhao
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Julian A Marschner
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
58
|
Honarpisheh M, Foresto-Neto O, Steiger S, Kraft F, Koehler P, von Rauchhaupt E, Potempa J, Adamowicz K, Koziel J, Lech M. Aristolochic acid I determine the phenotype and activation of macrophages in acute and chronic kidney disease. Sci Rep 2018; 8:12169. [PMID: 30111809 PMCID: PMC6093867 DOI: 10.1038/s41598-018-30628-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 08/03/2018] [Indexed: 01/12/2023] Open
Abstract
Acute and chronic kidney injuries are multifactorial traits that involve various risk factors. Experimental animal models are crucial to unravel important aspects of injury and its pathophysiological mechanisms. Translating knowledge obtained from experimental approaches into clinically useful information is difficult; therefore, significant attention needs to be paid to experimental procedures that mimic human disease. Herein, we compared aristolochic acid I (AAI) acute and chronic kidney injury model with unilateral ischemic-reperfusion injury (uIRI), cisplatin (CP)- or folic acid (FA)-induced renal damage. The administration of AAI showed significant changes in serum creatinine and BUN upon CKD. The number of neutrophils and macrophages were highly increased as well as AAI-induced CKD characterized by loss of tubular epithelial cells and fibrosis. The in vitro and in vivo data indicated that macrophages play an important role in the pathogenesis of AA-induced nephropathy (AAN) associated with an excessive macrophage accumulation and an alternative activated macrophage phenotype. Taken together, we conclude that AA-induced injury represents a suitable and relatively easy model to induce acute and chronic kidney injury. Moreover, our data indicate that this model is appropriate and superior to study detailed questions associated with renal macrophage phenotypes.
Collapse
Affiliation(s)
- Mohsen Honarpisheh
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Orestes Foresto-Neto
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Stefanie Steiger
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Franziska Kraft
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Paulina Koehler
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Ekaterina von Rauchhaupt
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany
| | - Jan Potempa
- Departments of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Karina Adamowicz
- Departments of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Joanna Koziel
- Departments of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maciej Lech
- Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IV, Department of Nephrology, LMU Munich, Germany. .,Departments of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
59
|
Williams TM, Wise AF, Layton DS, Ricardo SD. Phenotype and influx kinetics of leukocytes and inflammatory cytokine production in kidney ischemia/reperfusion injury. Nephrology (Carlton) 2018; 23:75-85. [PMID: 27696567 DOI: 10.1111/nep.12941] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/26/2016] [Accepted: 09/29/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIM Kidney ischemia/reperfusion (IR) injury is characterized by tubular epithelial cell (TEC) death and an inflammatory response involving cytokine production and immune cell infiltration. In various kidney diseases, increased macrophage numbers correlate with injury severity and poor prognosis. However, macrophage plasticity enables a diverse range of functions, including wound healing, making them a key target for novel therapies. This study aimed to comprehensively characterize the changes in myeloid and epithelial cells and the production of cytokines throughout the experimental IR model of acute kidney injury to aid in the identification of targets to promote and enhance kidney regeneration and repair. METHODS Flow cytometric analysis of murine unilateral IR injury was used to assess TEC and myeloid cell subpopulations in conjunction with histological analysis and cytokine production at 6 h, 1, 3, 5 and 7 days post IR injury, spanning the initial inflammatory phase and the following reparative phase. RESULTS IR injury resulted in a rapid infiltration of Ly6Chigh monocytes and neutrophils with a steady rise in F4/80high MHCIIhigh macrophages over the injury time. The production of the inflammatory cytokines IL-6, MCP-1 and TNF coincided with an increase in IL-10 production. CONCLUSION This characterization will provide a reference point for future studies designed to manipulate immune cell phenotype and function in order to promote endogenous repair of damaged kidneys.
Collapse
Affiliation(s)
- Timothy M Williams
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Andrea F Wise
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Daniel S Layton
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia.,Biosecurity Flagship, CSIRO, Geelong, Australia
| | - Sharon D Ricardo
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| |
Collapse
|
60
|
Osteopontin Blockade Attenuates Renal Injury After Ischemia Reperfusion by Inhibiting NK Cell Infiltration. Shock 2018; 47:52-60. [PMID: 27504800 DOI: 10.1097/shk.0000000000000721] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Renal ischemia-reperfusion (RIR) injury is a common occurrence after major surgery and shock, leading to acute kidney injury (AKI). Osteopontin (OPN) is a secreted glycoprotein that acts as a proinflammatory cytokine and activator of T lymphocytes. We hypothesized that blockade of OPN reduces the severity of inflammation and injury in RIR. Renal ischemia was induced in adult C57BL/6 mice via bilateral clamping of renal pedicles for 35 min, followed by reperfusion for 24 h. Anti-OPN antibody (Ab), nonimmunized isotype immunoglobulin G, or normal saline was injected intravenously at the time of reperfusion. Blood and kidneys were collected for analysis. At 24 h after RIR, OPN mRNA and protein levels were significantly increased in renal tissue compared with sham mice. In serum, elevated levels of blood urea nitrogen and creatinine were reduced in anti-OPN Ab-treated mice compared with vehicle. Anti-OPN Ab-treated mice also had decreased mRNA levels of injury markers neutrophil gelatinase-associated lipocalin and kidney injury molecule-1 compared with the vehicle. The histologic architecture and apoptosis of renal tissue were improved in the anti-OPN Ab-treated mice. In renal tissue, inflammatory cytokines interleukin 6 and tumor necrosis factor-α protein levels were reduced in the Ab-treated mice. Natural killer (NK) cell infiltration was decreased after anti-OPN Ab treatment, as was neutrophil infiltration, shown by reduced chemokine expression and Gr1 renal immunohistochemical staining. These findings demonstrate a beneficial role of OPN blockade in RIR associated with NK cell-mediated downregulation of inflammatory cytokines and chemokines. Administration of anti-OPN Ab may therefore serve as an immunomodulatory adjunct in the treatment of RIR-induced AKI.
Collapse
|
61
|
Ferhat M, Robin A, Giraud S, Sena S, Goujon JM, Touchard G, Hauet T, Girard JP, Gombert JM, Herbelin A, Thierry A. Endogenous IL-33 Contributes to Kidney Ischemia-Reperfusion Injury as an Alarmin. J Am Soc Nephrol 2018; 29:1272-1288. [PMID: 29436517 DOI: 10.1681/asn.2017060650] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 01/03/2018] [Indexed: 12/17/2022] Open
Abstract
Inflammation is a prominent feature of ischemia-reperfusion injury (IRI), which is characterized by leukocyte infiltration and renal tubular injury. However, signals that initiate these events remain poorly understood. We examined the role of the nuclear alarmin IL-33 in tissue injury and innate immune response triggered by experimental kidney ischemia-reperfusion. In wild-type mice, we found that IL-33 was constitutively expressed throughout the kidney in peritubular and periglomerular spaces, mainly by microvascular endothelial cells, from which it was released immediately during IRI. Compared with wild-type mice, mice lacking IL-33 (IL-33Gt/Gt) exhibited reductions in early tubular cell injury and subsequent renal infiltration of IFN-γ/IL-17A-producing neutrophils, with preservation of renal functions. This protection associated with decreased renal recruitment of myeloid dendritic cells, natural killer (NK) cells, and invariant natural killer T (iNKT) cells, the latter of which were reported as deleterious in IRI. Increases in the level of circulating IL-12, a key IL-33 cofactor, and the expression of ST2, an IL-33-specific receptor, on the surface of iNKT cells preceded the IL-33- and iNKT cell-dependent phase of neutrophil infiltration. Furthermore, IL-33 directly targeted iNKT cells in vitro, inducing IFN-γ and IL-17A production. We propose that endogenous IL-33 is released as an alarmin and contributes to kidney IRI by promoting iNKT cell recruitment and cytokine production, resulting in neutrophil infiltration and activation at the injury site. Our findings show a novel molecular mediator contributing to innate immune cell recruitment induced by renal ischemia-reperfusion and may provide therapeutic insights into AKI associated with renal transplantation.
Collapse
Affiliation(s)
- Maroua Ferhat
- Unité Mixte de Recherche 1082, Institut National de la Santé et de la Recherche Médicale, Poitiers, France.,Faculté de Médecine et de Pharmacie, University of Poitiers, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Aurélie Robin
- Unité Mixte de Recherche 1082, Institut National de la Santé et de la Recherche Médicale, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Sébastien Giraud
- Unité Mixte de Recherche 1082, Institut National de la Santé et de la Recherche Médicale, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Sandra Sena
- Unité Mixte de Recherche 1082, Institut National de la Santé et de la Recherche Médicale, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Jean-Michel Goujon
- Unité Mixte de Recherche 1082, Institut National de la Santé et de la Recherche Médicale, Poitiers, France.,Faculté de Médecine et de Pharmacie, University of Poitiers, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,Department of Anatomic Pathology, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Guy Touchard
- Faculté de Médecine et de Pharmacie, University of Poitiers, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,Departments of Nephrology and Transplantation, Poitiers, France
| | - Thierry Hauet
- Unité Mixte de Recherche 1082, Institut National de la Santé et de la Recherche Médicale, Poitiers, France.,Faculté de Médecine et de Pharmacie, University of Poitiers, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,Department of Biochemistry, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Jean-Philippe Girard
- Institute of Pharmacology and Structural Biology, Toulouse, France.,Centre National de la Recherche Scientifique Unité Mixte de Recherche UMR 5089, Toulouse, France.,Faculté des Siences et Ingénierie, University of Toulouse, Toulouse, France; and
| | - Jean-Marc Gombert
- Unité Mixte de Recherche 1082, Institut National de la Santé et de la Recherche Médicale, Poitiers, France.,Faculté de Médecine et de Pharmacie, University of Poitiers, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,Laboratory of Immunology, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - André Herbelin
- Unité Mixte de Recherche 1082, Institut National de la Santé et de la Recherche Médicale, Poitiers, France; .,Faculté de Médecine et de Pharmacie, University of Poitiers, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - Antoine Thierry
- Unité Mixte de Recherche 1082, Institut National de la Santé et de la Recherche Médicale, Poitiers, France.,Faculté de Médecine et de Pharmacie, University of Poitiers, Poitiers, France.,Unité Mixte de Recherche 1082, Centre Hospitalier Universitaire de Poitiers, Poitiers, France.,Departments of Nephrology and Transplantation, Poitiers, France
| |
Collapse
|
62
|
Uehara H, Minami K, Quante M, Nian Y, Heinbokel T, Azuma H, El Khal A, Tullius SG. Recall features and allorecognition in innate immunity. Transpl Int 2018; 31:6-13. [PMID: 28926127 PMCID: PMC7781186 DOI: 10.1111/tri.13073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/20/2017] [Accepted: 09/15/2017] [Indexed: 11/28/2022]
Abstract
Alloimmunity traditionally distinguishes short-lived, rapid and nonspecific innate immune responses from adaptive immune responses that are characterized by a highly specific response initiated in a delayed fashion. Key players of innate immunity such as natural killer (NK) cells and macrophages present the first-line defence of immunity. The concept of unspecific responses in innate immunity has recently been challenged. The discovery of pattern recognition receptors (PRRs) has demonstrated that innate immune cells respond in a semi-specific fashion through the recognition of pathogen-associated molecular patterns (PAMPs) representing conserved molecular structures shared by large groups of microorganisms. Although immunological memory has generally been considered as exclusive to adaptive immunity, recent studies have demonstrated that innate immune cells have the potential to acquire memory. Here, we discuss allospecific features of innate immunity and their relevance in transplantation.
Collapse
Affiliation(s)
- Hirofumi Uehara
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Koichiro Minami
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Markus Quante
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Yeqi Nian
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Timm Heinbokel
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Haruhito Azuma
- Department of Urology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Abdala El Khal
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Stefan G. Tullius
- Division of Transplant Surgery and Transplantation Surgery Research Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
63
|
Hultström M, Becirovic-Agic M, Jönsson S. Comparison of acute kidney injury of different etiology reveals in-common mechanisms of tissue damage. Physiol Genomics 2017; 50:127-141. [PMID: 29341864 DOI: 10.1152/physiolgenomics.00037.2017] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury (AKI) is a syndrome of reduced glomerular filtration rate and urine production caused by a number of different diseases. It is associated with renal tissue damage. This tissue damage can cause tubular atrophy and interstitial fibrosis that leads to nephron loss and progression of chronic kidney disease (CKD). This review describes the in-common mechanisms behind tissue damage in AKI caused by different underlying diseases. Comparing six high-quality microarray studies of renal gene expression after AKI in disease models (gram-negative sepsis, gram-positive sepsis, ischemia-reperfusion, malignant hypertension, rhabdomyolysis, and cisplatin toxicity) identified 5,254 differentially expressed genes in at least one of the AKI models; 66% of genes were found only in one model, showing that there are unique features to AKI depending on the underlying disease. There were in-common features in the form of four genes that were differentially expressed in all six models, 49 in at least five, and 215 were found in common between at least four models. Gene ontology enrichment analysis could be broadly categorized into the injurious processes hypoxia, oxidative stress, and inflammation, as well as the cellular outcomes of cell death and tissue remodeling in the form of epithelial-to-mesenchymal transition. Pathway analysis showed that MYC is a central connection in the network of activated genes in-common to AKI, which suggests that it may be a central regulator of renal gene expression in tissue injury during AKI. The outlining of this molecular network may be useful for understanding progression from AKI to CKD.
Collapse
Affiliation(s)
- Michael Hultström
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University , Uppsala , Sweden.,Anaesthesia and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University , Uppsala , Sweden
| | - Mediha Becirovic-Agic
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University , Uppsala , Sweden
| | - Sofia Jönsson
- Integrative Physiology, Department of Medical Cell Biology, Uppsala University , Uppsala , Sweden
| |
Collapse
|
64
|
Lu J, Yi Y, Pan R, Zhang C, Han H, Chen J, Liu W. Berberine protects HK-2 cells from hypoxia/reoxygenation induced apoptosis via inhibiting SPHK1 expression. J Nat Med 2017; 72:390-398. [PMID: 29260413 DOI: 10.1007/s11418-017-1152-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 11/02/2017] [Indexed: 02/01/2023]
Abstract
Renal ischemia reperfusion injury (RIRI) refers to the irreversible damage for renal function when blood perfusion is recovered after ischemia for an extended period, which is common in clinical surgeries and has been regarded as a major risk for acute renal failures (ARF) that is accompanied with unimaginably high morbidity and mortality. Hypoxia during ischemia followed by reoxygenation via reperfusion serves as a major event contributing to cell apoptosis, which has been widely accepted as the vital pathogenesis in RIRI. Preventing apoptosis in renal tubular epithelial cell has been considered as effective method for blocking RIRI. In this paper, we established a hypoxia/reoxygenation (H/R) injury model in human proximal tubular epithelial HK-2 cells. Here, we found increased SPHK1 levels in H/R injured HK-2 cells, which could be significantly down regulated after berberine treatment. Berberine has been reported to exert a protective effect on H/R-induced apoptosis of HK-2 cells. So, in our present study, we planned to investigate whether SPHK1 participated in the anti-apoptosis process of berberine in H/R injured HK-2 cells. Our study confirmed the protective effect of berberine against H/R-induced apoptosis in HK-2 cells through promoting cells viability, inhibiting cells apoptosis, and down-regulating p-P38, caspase-3, caspase-9 as well as SPHK1, while up regulating the ratio of Bcl-2/Bax. However, SPHK1 overexpression in HK-2 cells induced severe apoptosis, which can be significantly ameliorated with additional berberine treatment. We concluded that berberine could remarkably prevent H/R-induced apoptosis in HK-2 cells through down-regulating SPHK1 expression levels, and the mechanisms included the suppression of p38 MAPK activation and mitochondrial stress pathways.
Collapse
Affiliation(s)
- Jianrao Lu
- Department of Nephrology, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| | - Yang Yi
- Department of Nephrology, Jingan District Central Hospital/Jingan Branch, Huashan Hospital affiliated to Fudan University, Shanghai, 200040, China
| | - Ronghua Pan
- Department of Nephrology, Liyang Hospital of traditional Chinese medicine, Jiangsu Province, 213300, China.
| | - Chuanfu Zhang
- Department of Nephrology, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Haiyan Han
- Department of Nephrology, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Jie Chen
- Department of Nephrology, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Wenrui Liu
- Department of Nephrology, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| |
Collapse
|
65
|
Antonangeli F, Soriani A, Cerboni C, Sciumè G, Santoni A. How Mucosal Epithelia Deal with Stress: Role of NKG2D/NKG2D Ligands during Inflammation. Front Immunol 2017; 8:1583. [PMID: 29209320 PMCID: PMC5701928 DOI: 10.3389/fimmu.2017.01583] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/03/2017] [Indexed: 01/22/2023] Open
Abstract
Mucosal epithelia encounter both physicochemical and biological stress during their life and have evolved several mechanisms to deal with them, including regulation of immune cell functions. Stressed and damaged cells need to be cleared to control local inflammation and trigger tissue healing. Engagement of the activating NKG2D receptor is one of the most direct mechanisms involved in the recognition of stressed cells by the immune system. Indeed, injured cells promptly express NKG2D ligands that in turn mediate the activation of lymphocytes of both innate and adaptive arms of the immune system. This review focuses on different conditions that are able to modulate NKG2D ligand expression on the epithelia. Special attention is given to the mechanisms of immunosurveillance mediated by natural killer cells, which are finely tuned by NKG2D. Different types of stress, including viral and bacterial infections, chronic inflammation, and cigarette smoke exposure, are discussed as paradigmatic conditions for NKG2D ligand modulation, and the implications for tissue homeostasis are discussed.
Collapse
Affiliation(s)
- Fabrizio Antonangeli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Cristina Cerboni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Giuseppe Sciumè
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,Neuromed I.R.C.C.S. - Istituto Neurologico Mediterraneo, Pozzilli, Italy
| |
Collapse
|
66
|
Innate Immune Response in Kidney Ischemia/Reperfusion Injury: Potential Target for Therapy. J Immunol Res 2017; 2017:6305439. [PMID: 28676864 PMCID: PMC5476886 DOI: 10.1155/2017/6305439] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/17/2017] [Indexed: 01/06/2023] Open
Abstract
Acute kidney injury caused by ischemia and subsequent reperfusion is associated with a high rate of mortality and morbidity. Ischemia/reperfusion injury in kidney transplantation causes delayed graft function and is associated with more frequent episodes of acute rejection and progression to chronic allograft nephropathy. Alloantigen-independent inflammation is an important process, participating in pathogenesis of injurious response, caused by ischemia and reperfusion. This innate immune response is characterized by the activity of classical cells belonging to the immune system, such as neutrophils, macrophages, dendritic cells, lymphocytes, and also tubular epithelial cells and endothelial cells. These immune cells not only participate in inflammation after ischemia exerting detrimental influence but also play a protective role in the healing response from ischemia/reperfusion injury. Delineating of complex mechanisms of their actions could be fruitful in future prevention and treatment of ischemia/reperfusion injury. Among numerous so far conducted experiments, observed immunomodulatory role of adenosine and adenosine receptor agonists in complex interactions of dendritic cells, natural killer T cells, and T regulatory cells is emphasized as promising in the treatment of kidney ischemia/reperfusion injury. Potential pharmacological approaches which decrease NF-κB activity and antagonize mechanisms downstream of activated Toll-like receptors are discussed.
Collapse
|
67
|
Law BMP, Wilkinson R, Wang X, Kildey K, Lindner M, Rist MJ, Beagley K, Healy H, Kassianos AJ. Interferon-γ production by tubulointerstitial human CD56 bright natural killer cells contributes to renal fibrosis and chronic kidney disease progression. Kidney Int 2017; 92:79-88. [PMID: 28396119 DOI: 10.1016/j.kint.2017.02.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/31/2017] [Accepted: 02/02/2017] [Indexed: 01/04/2023]
Abstract
Natural killer (NK) cells are a population of lymphoid cells that play a significant role in mediating innate immune responses. Studies in mice suggest a pathological role for NK cells in models of kidney disease. In this study, we characterized the NK cell subsets present in native kidneys of patients with tubulointerstitial fibrosis, the pathological hallmark of chronic kidney disease. Significantly higher numbers of total NK cells (CD3-CD56+) were detected in renal biopsies with tubulointerstitial fibrosis compared with diseased biopsies without fibrosis and healthy kidney tissue using multi-color flow cytometry. At a subset level, both the CD56dim NK cell subset and particularly the CD56bright NK cell subset were elevated in fibrotic kidney tissue. However, only CD56bright NK cells significantly correlated with the loss of kidney function. Expression of the tissue-retention and -activation molecule CD69 on CD56bright NK cells was significantly increased in fibrotic biopsy specimens compared with non-fibrotic kidney tissue, indicative of a pathogenic phenotype. Further flow cytometric phenotyping revealed selective co-expression of activating receptor CD335 (NKp46) and differentiation marker CD117 (c-kit) on CD56bright NK cells. Multi-color immunofluorescent staining of fibrotic kidney tissue localized the accumulation of NK cells within the tubulointerstitium, with CD56bright NK cells (NKp46+ CD117+) identified as the source of pro-inflammatory cytokine interferon-γ within the NK cell compartment. Thus, activated interferon-γ-producing CD56bright NK cells are positioned to play a key role in the fibrotic process and progression to chronic kidney disease.
Collapse
Affiliation(s)
- Becker M P Law
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ray Wilkinson
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia; University of Queensland Medical School, University of Queensland, Brisbane, Queensland, Australia
| | - Xiangju Wang
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Katrina Kildey
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Mae Lindner
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Melissa J Rist
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Kenneth Beagley
- Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Helen Healy
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Andrew J Kassianos
- Conjoint Kidney Research Laboratory, Pathology Queensland, Brisbane, Queensland, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Institute of Health and Biomedical Innovation/School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia; University of Queensland Medical School, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
68
|
Risti M, Bicalho MDG. MICA and NKG2D: Is There an Impact on Kidney Transplant Outcome? Front Immunol 2017; 8:179. [PMID: 28289413 PMCID: PMC5326783 DOI: 10.3389/fimmu.2017.00179] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/07/2017] [Indexed: 01/06/2023] Open
Abstract
This paper aims to present an overview of MICA and natural killer group 2 member D (NKG2D) genetic and functional interactions and their impact on kidney transplant outcome. Organ transplantation has gone from what can accurately be called a “clinical experiment” to a routine and reliable practice, which has proven to be clinically relevant, life-saving and cost-effective when compared with non-transplantation management strategies of both chronic and acute end-stage organ failures. The kidney is the most frequently transplanted organ in the world (transplant-observatory1). The two treatment options for end-stage renal disease (ESRD) are dialysis and/or transplantation. Compared with dialysis, transplantation is associated with significant improvements in quality of life and overall longevity. A strong relationship exists between allograft loss and human leukocyte antigens (HLA) antibodies (Abs). HLA Abs are not the only factor involved in graft loss, as multiple studies have shown that non-HLA antigens are also involved, even when a patient has a good HLA matche and receives standard immunosuppressive therapy. A deeper understanding of other biomarkers is therefore important, as it is likely to lead to better monitoring (and consequent success) of organ transplants. The objective is to fill the void left by extensive reviews that do not often dive this deep into the importance of MICA and NKG2D in allograft acceptance and their partnership in the immune response. There are few papers that explore the relationship between these two protagonists when it comes to kidney transplantation. This is especially true for the role of NKG2D in kidney transplantation. These reasons give a special importance to this review, which aims to be a helpful tool in the hands of researchers in this field.
Collapse
Affiliation(s)
- Matilde Risti
- LIGH - Immunogenetics and Histocompatibility Laboratory, Federal University of Paraná , Curitiba , Brazil
| | - Maria da Graça Bicalho
- LIGH - Immunogenetics and Histocompatibility Laboratory, Federal University of Paraná , Curitiba , Brazil
| |
Collapse
|
69
|
Wang YH, Zhang YG. Kidney and innate immunity. Immunol Lett 2017; 183:73-78. [PMID: 28143791 DOI: 10.1016/j.imlet.2017.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/25/2016] [Accepted: 01/23/2017] [Indexed: 12/13/2022]
Abstract
Innate immune system is an important modulator of the inflammatory response during infection and tissue injury/repair. The kidney as a vital organ with high energy demand plays a key role in regulating the disease related metabolic process. Increasing research interest has focused on the immune pathogenesis of many kidney diseases. However, innate immune cells such as dendritic cells, macrophages, NK cells and a few innate lymphocytes, as well as the complement system are essential for renal immune homeostasis and ensure a coordinated balance between tissue injury and regeneration. The innate immune response provides the first line of host defense initiated by several classes of pattern recognition receptors (PRRs), such as membrane-bound Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), together with inflammasomes responsible for early innate immune response. Although the innate immune system is well studied, the research on the detailed relationship between innate immunity and kidney is still very limited. In this review, we will focus on the innate immune sensing system in renal immune homeostasis, as well as the corresponding pathogenesis of many kidney diseases. The pivotal roles of innate immunity in renal injury and regeneration with special emphasis on kidney disease related immunoregulatory mechanism are also discussed.
Collapse
Affiliation(s)
- Ying-Hui Wang
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Faculty of Basic Medicine, Guilin Medical University, Guilin 541004, China
| | - Yu-Gen Zhang
- Center for Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin 541004, China; Department of Immunology, Faculty of Basic Medicine, Guilin Medical University, Guilin 541004, China.
| |
Collapse
|
70
|
Abstract
The clinical category of acute kidney injury includes a wide range of completely different disorders, many with their own pathomechanisms and treatment targets. In this review we focus on the role of inflammation in the pathogenesis of acute tubular necrosis (ATN). We approach this topic by first discussing the role of the immune system in the different phases of ATN (ie, early and late injury phase, recovery phase, and the long-term outcome phase of an ATN episode). A more detailed discussion focuses on putative therapeutic targets among the following mechanisms and mediators: oxidative stress and reactive oxygen species-related necroinflammation, regulated cell death-related necroinflammation, immunoregulatory lipid mediators, cytokines and cytokine signaling, chemokines and chemokine signaling, neutrophils and neutrophils extracellular traps (NETs) associated neutrophil cell death, called NETosis, extracellular histones, proinflammatory mononuclear phagocytes, humoral mediators such as complement, pentraxins, and natural antibodies. Any prioritization of these targets has to take into account the intrinsic differences between rodent models and human ATN, the current acute kidney injury definitions, and the timing of clinical decision making. Several conceptual problems need to be solved before anti-inflammatory drugs that are efficacious in rodent ATN may become useful therapeutics for human ATN.
Collapse
Affiliation(s)
- Shrikant R Mulay
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Alexander Holderied
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Santhosh V Kumar
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany.
| |
Collapse
|
71
|
El-Sisi AE, Sokar SS, Abu-Risha SE, Ibrahim HA. Combination of tadalafil and diltiazem attenuates renal ischemia reperfusion-induced acute renal failure in rats. Biomed Pharmacother 2016; 84:861-869. [PMID: 27736652 DOI: 10.1016/j.biopha.2016.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/27/2016] [Accepted: 10/03/2016] [Indexed: 12/24/2022] Open
|
72
|
Mehrotra P, Collett JA, McKinney SD, Stevens J, Ivancic CM, Basile DP. IL-17 mediates neutrophil infiltration and renal fibrosis following recovery from ischemia reperfusion: compensatory role of natural killer cells in athymic rats. Am J Physiol Renal Physiol 2016; 312:F385-F397. [PMID: 27852609 DOI: 10.1152/ajprenal.00462.2016] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 12/19/2022] Open
Abstract
T cells have been implicated in the pathogenesis of acute kidney injury (AKI) and its progression to chronic kidney disease (CKD). Previous studies suggest that Th17 cells participate during the AKI-to-CKD transition, and inhibition of T cell activity by mycophenolate mofetil (MMF) or losartan attenuates the development of fibrosis following AKI. We hypothesized that T cell-deficient rats may have reduced levels of IL-17 cytokine leading to decreased fibrosis following AKI. Renal ischemis-reperfusion (I/R) was performed on T cell-deficient athymic rats (Foxn1rnu-/rnu-) and control euthymic rats (Foxn1rnu-/+), and CKD progression was hastened by unilateral nephrectomy at day 33 and subsequent exposure to 4.0% sodium diet. Renal fibrosis developed in euthymic rats and was reduced by MMF treatment. Athymic rats exhibited a similar degree of fibrosis, but this was unaffected by MMF treatment. FACS analysis demonstrated that the number of IL-17+ cells was similar between postischemic athymic vs. euthymic rats. The source of IL-17 production in euthymic rats was predominately from conventional T cells (CD3+/CD161-). In the absence of conventional T cells in athymic rats, a compensatory pathway involving natural killer cells (CD3-/CD161+) was the primary source of IL-17. Blockade of IL-17 activity using IL-17Rc receptor significantly decreased fibrosis and neutrophil recruitment in both euthymic and athymic rats compared with vehicle-treated controls. Taken together, these data suggest that IL-17 secretion participates in the pathogenesis of AKI-induced fibrosis possibly via the recruitment of neutrophils and that the source of IL-17 may be from either conventional T cells or NK cells.
Collapse
Affiliation(s)
- Purvi Mehrotra
- Department of Cellular and Integrative Physiology, Indiana University of Medicine, Indianapolis, Indiana
| | - Jason A Collett
- Department of Cellular and Integrative Physiology, Indiana University of Medicine, Indianapolis, Indiana
| | - Seth D McKinney
- Department of Cellular and Integrative Physiology, Indiana University of Medicine, Indianapolis, Indiana
| | - Jackson Stevens
- Department of Cellular and Integrative Physiology, Indiana University of Medicine, Indianapolis, Indiana
| | - Carlie M Ivancic
- Department of Cellular and Integrative Physiology, Indiana University of Medicine, Indianapolis, Indiana
| | - David P Basile
- Department of Cellular and Integrative Physiology, Indiana University of Medicine, Indianapolis, Indiana
| |
Collapse
|
73
|
Zou X, Gu D, Zhang G, Zhong L, Cheng Z, Liu G, Zhu Y. NK Cell Regulatory Property is Involved in the Protective Role of MSC-Derived Extracellular Vesicles in Renal Ischemic Reperfusion Injury. Hum Gene Ther 2016; 27:926-935. [PMID: 27510907 DOI: 10.1089/hum.2016.057] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Immunomodulation has been regarded as an important therapeutic aspect of mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) in renal ischemic reperfusion injury (IRI), and the specific mechanism still unclear. Here, we attempt to test the function of human MSC-EVs on renal IRI by targeting the natural killer (NK) cells and to investigate the possible mechanism. Data indicated that EVs decreased NK cells in spleen and ischemic kidney. Both the EVs and antibody-dependent depletion of NK cells displayed a protective role in IRI rats. Moreover, the splenectomy model was established to evaluate the role of spleen in this process. It showed that the NK cell regulatory ability and renal protective effects by EVs still exist without spleen, which is unlike MSC properties published previously. Further, the down-regulation of chemokines in injured kidney and the delivery of RNAs through EVs in vitro were also observed. Through the microRNA array test, various inflammation-related microRNAs highly expressed in MSC-EVs compared with fibroblast EVs were tested. Thus, these results indicated that MSC-EVs could ameliorate renal ischemic reperfusion injury by decreasing NK cells and the spleen is not necessary in this process. The regulation of chemokines in injured kidney was the other factor, and the transfer of various microRNAs in the MSC-EVs may be involved. This provides direction for future clinical applications.
Collapse
Affiliation(s)
- Xiangyu Zou
- 1 Department of Urology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Di Gu
- 1 Department of Urology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Guangyuan Zhang
- 1 Department of Urology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China.,2 Department of Urology, Affiliated Zhongda Hospital of Southeast University , Nanjing, China
| | - Liang Zhong
- 3 Department of Urology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | | | - Guohua Liu
- 1 Department of Urology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| | - Yingjian Zhu
- 1 Department of Urology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai, China
| |
Collapse
|
74
|
Hussein AM, Sakr HF, Alenzi FQ. Possible Underlying Mechanisms of the Renoprotective Effect of Remote Limb Ischemic Preconditioning Against Renal Ischemia/Reperfusion Injury: A Role of Osteopontin, Transforming Growth Factor-Beta and Survivin. Nephron Clin Pract 2016; 134:117-129. [PMID: 27486809 DOI: 10.1159/000447953] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/16/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND It has been documented that remote limb ischemic preconditioning (rIPC) protect kidneys against renal ischemia/reperfusion (I/R). We hypothesized that osteopontin (OPN), transforming growth factor beta (TGF-β), apoptotic proteins (survivin and caspase-3) and oxidative stress play role in the renoprotective effects of rIPC. MATERIALS AND METHODS Fifty-four male Sprague-Dawley rats were randomized into 3 equal groups: sham group, I/R group (left renal 45 min ischemia) and rIPC group (as I/R group with 3 cycles of left hind limb ischemia just before renal ischemia). Each group was subdivided into 24, 48 and 72 h groups according to the time of sacrifice. We measured serum creatinine and blood urea nitrogen (BUN) at the baseline and end points. Also, left kidney was harvested at study end points for assessment of the expression of OPN, TGF-β, apoptotic proteins (survivin and caspase-3) and oxidative stress markers (malondialdehyde (MDA), glutathione (GSH) and superoxide dismutase (SOD)) in kidney tissues and histopathological examination. RESULTS Serum creatinine and BUN levels and histopathological damage score were significantly lower in rIPC group than I/R group (p < 0.005). Also, compared to I/R group, the levels of MDA and the expression of OPN, TGF-β and caspase-3 in kidney tissues were significantly lower in rIPC group, while the levels of SOD and GSH and the expression of survivin in kidney tissues were significantly higher in rIPC group at all time points (p ≤ 0.05). CONCLUSIONS rIPC exhibited protective effects against renal I/R injury which might be due to inhibition of OPN expression, inflammatory cytokine TGF-β and caspase-3 and activation of anti-apoptotic protein survivin as well as improvement of oxidative stress in kidney tissues.
Collapse
Affiliation(s)
- Abdelaziz M Hussein
- Department of Medical Physiology, Mansoura University, Faculty of Medicine, Mansoura, Egypt
| | | | | |
Collapse
|
75
|
Seyda M, Elkhal A, Quante M, Falk CS, Tullius SG. T Cells Going Innate. Trends Immunol 2016; 37:546-556. [PMID: 27402226 DOI: 10.1016/j.it.2016.06.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023]
Abstract
Natural killer (NK) cell receptors (NKRs) play a crucial role in the homeostasis of antigen-experienced T cells. Indeed, prolonged antigen stimulation may induce changes in the receptor repertoire of T cells to a profile that features NKRs. Chronic antigen exposure, at the same time, has been shown to trigger the loss of costimulatory CD28 molecules with recently reported intensified antigen thresholds of antigen-experienced CD8(+) T cells. In transplantation, NKRs have been shown to assist allograft rejection in a CD28-independent fashion. We discuss here a role for CD28-negative T cells that have acquired the competency of the NKR machinery, potentially promoting allorecognition either through T cell receptor (TCR) crossreactivity or independently from TCR recognition. Collectively, NKRs can bring about innate-like T cells by providing alternative costimulatory pathways that gain relevance in chronic inflammation, potentially leading to resistance to CD28-targeting immunosuppressants.
Collapse
Affiliation(s)
- Midas Seyda
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - Abdallah Elkhal
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Markus Quante
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christine S Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany; German Center for Infection Research (DZIF), Hannover, Germany
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
76
|
Kimura S, Ozaki KS, Ueki S, Zhang M, Yokota S, Stolz DB, Geller DA, Murase N. Contribution of alloantigens to hepatic ischemia/reperfusion injury: Roles of natural killer cells and innate immune recognition of nonself. Liver Transpl 2016; 22:80-90. [PMID: 26335784 DOI: 10.1002/lt.24330] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 08/17/2015] [Accepted: 08/27/2015] [Indexed: 12/24/2022]
Abstract
Hepatic ischemia/reperfusion injury (IRI) remains a major clinical problem and involves the innate immune system's recognition of "nonself." Considering the efficient nonself recognition by natural killer (NK) cells, we hypothesize in this study that hepatic IRI associated with liver transplantation (LT) could be augmented in allogeneic rather than in syngeneic (Syn) grafts due to alloantigen recognition by innate immune cells, especially by NK cells. Using green fluorescent protein (GFP)/Sprague-Dawley rats, we tested our hypothesis in a rat LT model with 18 hours of cold storage in University of Wisconsin solution. Hepatic IRI was significantly augmented in allografts with higher alanine transaminase levels, increased necrosis, and vigorous proinflammatory mediator up-regulation compared to Syn grafts. Injury increased in allografts associated with augmented GFP+ host leukocyte infiltration due to significantly increased host CD11b/c+ and RP-1(+) neutrophil recruitment. A large number of liver-resident (donor) mature CD11b/c+ NK cells quickly diminished from allografts, but not from Syn grafts. Depletion of mature NK cells from liver grafts with anti-asialo monosialotetrahexosylganglioside significantly improved hepatic IRI and reduced neutrophil infiltration and proinflammatory mediators. In conclusion, early innate immune responses were more significantly enhanced in allografts than in Syn grafts during hepatic IRI, in part through NK cell recognition of "missing self."
Collapse
Affiliation(s)
- Shoko Kimura
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Kikumi S Ozaki
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Shinya Ueki
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Matthew Zhang
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Shinichiro Yokota
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - David A Geller
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Noriko Murase
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| |
Collapse
|
77
|
dos Santos DC, Campos EF, Saraiva Câmara NO, David DSR, Malheiros DMAC. Compartment-specific expression of natural killer cell markers in renal transplantation: immune profile in acute rejection. Transpl Int 2015; 29:443-52. [DOI: 10.1111/tri.12726] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/08/2015] [Accepted: 11/19/2015] [Indexed: 01/18/2023]
Affiliation(s)
- Daniela Cristina dos Santos
- Department of Pathology; Botucatu Medical School; State University of São Paulo; São Paulo Brazil
- Department of Pathology; Faculty of Medicine; University of São Paulo; São Paulo Brazil
| | - Erika Fernandes Campos
- Department of Immunology; Institute of Biomedical Sciences; University of São Paulo State; São Paulo Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Immunology; Institute of Biomedical Sciences; University of São Paulo State; São Paulo Brazil
| | | | | |
Collapse
|
78
|
Rabb H, Griffin MD, McKay DB, Swaminathan S, Pickkers P, Rosner MH, Kellum JA, Ronco C. Inflammation in AKI: Current Understanding, Key Questions, and Knowledge Gaps. J Am Soc Nephrol 2015; 27:371-9. [PMID: 26561643 DOI: 10.1681/asn.2015030261] [Citation(s) in RCA: 423] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Inflammation is a complex biologic response that is essential for eliminating microbial pathogens and repairing tissue after injury. AKI associates with intrarenal and systemic inflammation; thus, improved understanding of the cellular and molecular mechanisms underlying the inflammatory response has high potential for identifying effective therapies to prevent or ameliorate AKI. In the past decade, much knowledge has been generated about the fundamental mechanisms of inflammation. Experimental work in small animal models has revealed many details of the inflammatory response that occurs within the kidney after typical causes of AKI, including insights into the molecular signals released by dying cells, the role of pattern recognition receptors, the diverse subtypes of resident and recruited immune cells, and the phased transition from destructive to reparative inflammation. Although this expansion of the basic knowledge base has increased the number of mechanistically relevant targets of intervention, progress in developing therapies that improve AKI outcomes by modulation of inflammation remains slow. In this article, we summarize the most important recent developments in understanding the inflammatory mechanisms of AKI, highlight key limitations of the commonly used animal models and clinical trial designs that may prevent successful clinical application, and suggest priority approaches for research toward clinical translation in this area.
Collapse
Affiliation(s)
- Hamid Rabb
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland;
| | - Matthew D Griffin
- Regenerative Medicine Institute, School of Medicine, College of Medicine Nursing and Health Sciences, National University of Ireland, Galway, Ireland
| | - Dianne B McKay
- Division of Nephrology and Hypertension, Department of Medicine, University of California, San Diego, La Jolla, California
| | | | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mitchell H Rosner
- Division of Nephrology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - John A Kellum
- Center for Critical Care Nephrology and Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Claudio Ronco
- Department of Nephrology Dialysis and Transplantation, Saint Bortolo Hospital and the International Renal Research Institute, Vicenza, Italy
| | | |
Collapse
|
79
|
Victorino F, Sojka DK, Brodsky KS, McNamee EN, Masterson JC, Homann D, Yokoyama WM, Eltzschig HK, Clambey ET. Tissue-Resident NK Cells Mediate Ischemic Kidney Injury and Are Not Depleted by Anti-Asialo-GM1 Antibody. THE JOURNAL OF IMMUNOLOGY 2015; 195:4973-85. [PMID: 26453755 DOI: 10.4049/jimmunol.1500651] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 09/15/2015] [Indexed: 01/01/2023]
Abstract
NK cells are innate lymphoid cells important for immune surveillance, identifying and responding to stress, infection, and/or transformation. Whereas conventional NK (cNK) cells circulate systemically, many NK cells reside in tissues where they appear to be poised to locally regulate tissue function. In the present study, we tested the contribution of tissue-resident NK (trNK) cells to tissue homeostasis by studying ischemic injury in the mouse kidney. Parabiosis experiments demonstrate that the kidney contains a significant fraction of trNK cells under homeostatic conditions. Kidney trNK cells developed independent of NFIL3 and T-bet, and they expressed a distinct cell surface phenotype as compared with cNK cells. Among these, trNK cells had reduced asialo-GM1 (AsGM1) expression relative to cNK cells, a phenotype observed in trNK cells across multiple organs and mouse strains. Strikingly, anti-AsGM1 Ab treatment, commonly used as an NK cell-depleting regimen, resulted in a robust and selective depletion of cNKs, leaving trNKs largely intact. Using this differential depletion, we tested the relative contribution of cNK and trNK cells in ischemic kidney injury. Whereas anti-NK1.1 Ab effectively depleted both trNK and cNK cells and protected against ischemic/reperfusion injury, anti-AsGM1 Ab preferentially depleted cNK cells and failed to protect against injury. These data demonstrate unanticipated specificity of anti-AsGM1 Ab depletion on NK cell subsets and reveal a new approach to study the contributions of cNK and trNK cells in vivo. In total, these data demonstrate that trNK cells play a key role in modulating local responses to ischemic tissue injury in the kidney and potentially other organs.
Collapse
Affiliation(s)
- Francisco Victorino
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045; Immunology Graduate Program, University of Colorado School of Medicine, Aurora, CO 80045
| | - Dorothy K Sojka
- Rheumatology Division, Washington University School of Medicine, St. Louis, MO 63110
| | - Kelley S Brodsky
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Eoin N McNamee
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Joanne C Masterson
- Gastrointestinal Eosinophilic Diseases Program, Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045; Digestive Health Institute, Children's Hospital Colorado, Aurora, CO 80045; and
| | - Dirk Homann
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Wayne M Yokoyama
- Rheumatology Division, Washington University School of Medicine, St. Louis, MO 63110; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110
| | - Holger K Eltzschig
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045
| | - Eric T Clambey
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045;
| |
Collapse
|
80
|
Song H, Kim Y, Park G, Kim YS, Kim S, Lee HK, Chung WY, Park SJ, Han SY, Cho D, Hur D. Transforming growth factor-β1 regulates human renal proximal tubular epithelial cell susceptibility to natural killer cells via modulation of the NKG2D ligands. Int J Mol Med 2015; 36:1180-8. [PMID: 26311146 DOI: 10.3892/ijmm.2015.2317] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 08/13/2015] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor-β (TGF-β) has a significant role in the response to injury and tissue repair, and it has been detected in various cell types. However, the mechanism by which it regulates the response to ischemia‑reperfusion injury (IRI) and manipulates natural killer (NK) cells is not well understood. In the present study, TGF‑β modulated NK cell function, thereby promoting recovery from renal IRI. Human renal proximal tubular epithelial cells (HK‑2) treated with TGF‑β exhibited increased surface and intracellular expression of the NK group 2 member D (NKG2D) ligand MICA. This increased surface expression of MICA inhibited NK cell cytotoxicity to the HK‑2 cells. In addition, an enzyme‑linked immunosorbent assay revealed that TGF‑β treatment evidently increased the amount of soluble MICA released into the culture supernatant from HK‑2 cells. Taken together, these findings suggest that TGF‑β‑induced release of soluble MICA leads to downregulation of NKG2D, thereby preventing NK cell‑mediated cytotoxicity toward renal proximal tubular epithelial cells in renal IRI, which in turn improves the survival of these cells.
Collapse
Affiliation(s)
- Hyunkeun Song
- Department of Microbiology and Immunology, Laboratory for Medical Oncology, Inje University College of Medicine, Busan 614‑735, Republic of Korea
| | - Yeonye Kim
- Department of Microbiology and Immunology, Laboratory for Medical Oncology, Inje University College of Medicine, Busan 614‑735, Republic of Korea
| | - Gabin Park
- Department of Anatomy, Inje University College of Medicine, Busan 614‑735, Republic of Korea
| | - Yeong-Seok Kim
- Department of Anatomy, Inje University College of Medicine, Busan 614‑735, Republic of Korea
| | - Seonghan Kim
- Department of Anatomy, Inje University College of Medicine, Busan 614‑735, Republic of Korea
| | - Hyun-Kyung Lee
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan 614‑735, Republic of Korea
| | - Woo Yeong Chung
- Department of Pediatrics, Inje University Busan Paik Hospital, Busan 614‑735, Republic of Korea
| | - Seok Ju Park
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan 614‑735, Republic of Korea
| | - Sang-Youb Han
- Department of Internal Medicine, Inje University Ilsan-Paik Hospital, Goyang, Gyeonggi 411‑706, Republic of Korea
| | - Daeho Cho
- Department of Life Science, Sookmyung Women's University, Yongsan-ku, Seoul 140-742, Republic of Korea
| | - Daeyoung Hur
- Department of Anatomy, Inje University College of Medicine, Busan 614‑735, Republic of Korea
| |
Collapse
|
81
|
Hébert MJ, Jevnikar AM. The Impact of Regulated Cell Death Pathways on Alloimmune Responses and Graft Injury. CURRENT TRANSPLANTATION REPORTS 2015. [DOI: 10.1007/s40472-015-0067-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
82
|
Salvadori M, Rosso G, Bertoni E. Update on ischemia-reperfusion injury in kidney transplantation: Pathogenesis and treatment. World J Transplant 2015; 5:52-67. [PMID: 26131407 PMCID: PMC4478600 DOI: 10.5500/wjt.v5.i2.52] [Citation(s) in RCA: 267] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 01/16/2015] [Accepted: 04/27/2015] [Indexed: 02/05/2023] Open
Abstract
Ischemia/reperfusion injury is an unavoidable relevant consequence after kidney transplantation and influences short term as well as long-term graft outcome. Clinically ischemia/reperfusion injury is associated with delayed graft function, graft rejection, chronic rejection and chronic graft dysfunction. Ischemia/reperfusion affects many regulatory systems at the cellular level as well as in the renal tissue that result in a distinct inflammatory reaction of the kidney graft. Underlying factors of ischemia reperfusion include energy metabolism, cellular changes of the mitochondria and cellular membranes, initiation of different forms of cell death-like apoptosis and necrosis together with a recently discovered mixed form termed necroptosis. Chemokines and cytokines together with other factors promote the inflammatory response leading to activation of the innate immune system as well as the adaptive immune system. If the inflammatory reaction continues within the graft tissue, a progressive interstitial fibrosis develops that impacts long-term graft outcome. It is of particular importance in kidney transplantation to understand the underlying mechanisms and effects of ischemia/reperfusion on the graft as this knowledge also opens strategies to prevent or treat ischemia/reperfusion injury after transplantation in order to improve graft outcome.
Collapse
|
83
|
Zhang ZX, Huang X, Jiang J, Lau A, Yin Z, Liu W, Haig A, Jevnikar AM. Natural Killer Cells Mediate Long-term Kidney Allograft Injury. Transplantation 2015; 99:916-924. [PMID: 25719259 DOI: 10.1097/tp.0000000000000665] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Chronic allograft injury remains the leading cause of late kidney graft loss despite improvements in immunosuppressive drugs and a reduction in acute T cell-mediated rejection. We have recently demonstrated that natural killer (NK) cells are cytotoxic to tubular epithelial cells and contribute to acute kidney ischemia-reperfusion injury. The role of NK cells in kidney allograft rejection has not been studied. METHODS A "parent to F1" kidney transplant model was used to study NK cell-mediated transplant rejection. RESULTS The C57BL/6 kidneys were transplanted into fully nephrectomized CB6F1 (C57BL/6 x BALB/c) mice. Serum creatinine levels increased from baseline (18.8 ± 5.0 μmol/L to 37.2 ± 5.9 μmol/L, P < 0.001) at 60 days after transplantation. B6Rag-to-CB6F1Rag (B6RagxBALB/cRag) recipients, which lack T and B cells but retain NK cells, showed similar levels of kidney dysfunction 65 days after transplantation (creatinine, 33.8 ± 7.9 μmol/L vs 17.5 ± 5.1 μmol/L in nontransplant Rag mice, P < 0.05). Importantly, depletion of NK cells in Rag1 recipients inhibited kidney injury (24.6 ± 5.5 μmol/L, P < 0.05). Osteopontin, which can activate NK cells to mediate tubular epithelial cell death in vitro, was highly expressed in 60 days kidney grafts. Osteopontin null kidney grafts had reduced injury after transplantation into CB6F1 mice (17.7 ± 3.1 μmol/L, P < 0.001). CONCLUSIONS Collectively, these data demonstrate for the first time that independent of T and B cells, NK cells have a critical role in mediating long-term transplant kidney injury. Specific therapeutic strategies that target NK cells in addition to conventional immunosuppression may be required to attenuate chronic kidney transplant injury.
Collapse
Affiliation(s)
- Zhu-Xu Zhang
- 1 Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ontario, Canada. 2 Department of Medicine, Western University, London, Ontario, Canada. 3 Department of Pathology, Western University, London, Ontario, Canada. 4 Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
84
|
Abstract
Understanding innate immune responses and their correlation to alloimmunity after solid organ transplantation is key to optimizing long term graft outcome. While Ischemia/Reperfusion injury (IRI) has been well studied, new insight into central mechanisms of innate immune activation, i.e. chemokine mediated cell trafficking and the role of Toll-like receptors have evolved recently. The mechanistic implications of Neutrophils, Macrophages/Monocytes, NK-cells, Dendritic cells in renal IRI has been proven by selective depletion of these cell types, thereby offering novel therapeutic interventions. At the same time, the multi-faceted role of different T-cell subsets in IRI has gained interest, highlighting the dichotomous effects of differentiated T-cells and suggesting more selective therapeutic approaches. Targeting innate immune cells and their activation and migration pathways, respectively, has been promising in experimental models holding translational potential. This review will summarize the effects of innate immune activation and potential strategies to interfere with the immunological cascade following renal IRI.
Collapse
|
85
|
Antigen dependently activated cluster of differentiation 8-positive T cells cause perforin-mediated neurotoxicity in experimental stroke. J Neurosci 2015; 34:16784-95. [PMID: 25505331 DOI: 10.1523/jneurosci.1867-14.2014] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Neuroinflammation plays a key role in secondary brain damage after stroke. Although deleterious effects of proinflammatory cytokines are well characterized, direct cytotoxic effects of invading immune cells on the ischemic brain and the importance of their antigen-dependent activation are essentially unknown. Here we examined the effects of adaptive and innate immune cells-cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells-that share the direct perforin-mediated cytotoxic pathway on outcome after cerebral ischemia in mice. Although CTLs and NK cells both invaded the ischemic brain, only brain-infiltrating CTLs but not NK cells were more activated than their splenic counterparts. Depletion of CTLs decreased infarct volumes and behavioral deficit in two ischemia models, whereas NK cell depletion had no effect. Correspondingly, adoptive CTL transfer from wild-type into Rag1 knock-out mice increased infarct size. Adoptive CTL transfer from perforin knock-out or interferon-γ knock-out mice into Rag1 knock-out mice revealed that CTL neurotoxicity was mediated by perforin. Accordingly, CTLs isolated from wild-type or interferon-γ knock-out but not from perforin knock-out mice induced neuronal cell death in vitro. CTLs derived from ovalbumin-specific T-cell receptor transgenic mice were not activated and infiltrated less into the ischemic brain compared with wild-type CTLs. Their transfer did not increase the infarct size of Rag1 knock-out mice, indicating antigen-dependent activation as an essential component of CTL neurotoxicity. Our findings underscore the importance of antigen-dependent, direct cytotoxic immune responses in stroke and suggest modulation of CTLs and their effector pathways as a potential new strategy for stroke therapy.
Collapse
|
86
|
Abstract
Acute kidney injury (AKI) prolongs hospital stay and increases mortality in various clinical settings. Ischaemia-reperfusion injury (IRI), nephrotoxic agents and infection leading to sepsis are among the major causes of AKI. Inflammatory responses substantially contribute to the overall renal damage in AKI. Both innate and adaptive immune systems are involved in the inflammatory process occurring in post-ischaemic AKI. Proinflammatory damage-associated molecular patterns, hypoxia-inducible factors, adhesion molecules, dysfunction of the renal vascular endothelium, chemokines, cytokines and Toll-like receptors are involved in the activation and recruitment of immune cells into injured kidneys. Immune cells of both the innate and adaptive immune systems, such as neutrophils, dendritic cells, macrophages and lymphocytes contribute to the pathogenesis of renal injury after IRI, and some of their subpopulations also participate in the repair process. These immune cells are also involved in the pathogenesis of nephrotoxic AKI. Experimental studies of immune cells in AKI have resulted in improved understanding of the immune mechanisms underlying AKI and will be the foundation for development of novel diagnostic and therapeutic targets. This Review describes what is currently known about the function of the immune system in the pathogenesis and repair of ischaemic and nephrotoxic AKI.
Collapse
Affiliation(s)
- Hye Ryoun Jang
- Nephrology Division, Department of Medicine, Samsung Medical Centre, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, 81 Irwon-Ro Gangnam-gu, Seoul 135-710, South Korea
| | - Hamid Rabb
- Nephrology Division, Department of Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
87
|
Park SJ, Lee JS, Kwon B, Cho HR. Integration of the Innate and Adaptive Immunity by CD137-CD137L Bidirectional Signals: Implications in Allograft Rejection. KOREAN JOURNAL OF TRANSPLANTATION 2014. [DOI: 10.4285/jkstn.2014.28.3.113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Sang June Park
- Department of Surgery, Ulsan University Hospital, University of Ulsan College of Medicine, School of Biological Sciences4, University of Ulsan, Ulsan, Korea
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, School of Biological Sciences4, University of Ulsan, Ulsan, Korea
| | - Jong Soo Lee
- Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, School of Biological Sciences4, University of Ulsan, Ulsan, Korea
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, School of Biological Sciences4, University of Ulsan, Ulsan, Korea
| | - Byungsuk Kwon
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, School of Biological Sciences4, University of Ulsan, Ulsan, Korea
| | - Hong Rae Cho
- Department of Surgery, Ulsan University Hospital, University of Ulsan College of Medicine, School of Biological Sciences4, University of Ulsan, Ulsan, Korea
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan College of Medicine, School of Biological Sciences4, University of Ulsan, Ulsan, Korea
| |
Collapse
|
88
|
Jang HR, Park JH, Kwon GY, Lee JE, Huh W, Jin HJ, Choi SJ, Oh W, Oh HY, Kim YG. Effect of preemptive treatment with human umbilical cord blood-derived mesenchymal stem cells on the development of renal ischemia-reperfusion injury in mice. Am J Physiol Renal Physiol 2014; 307:F1149-61. [PMID: 25143451 DOI: 10.1152/ajprenal.00555.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human umbilical cord blood-derived mesenchymal stem cells (HUCB-MSCs) have been studied in several models of immune-mediated disease because of their unique immunomodulatory properties. We hypothesized that HUCB-MSCs could suppress the inflammatory response in postischemic kidneys and attenuate early renal injury. In 8- to 10-wk-old male C57BL/6 mice, bilateral ischemia-reperfusion injury (IRI) surgery was performed, and 1 × 10(6) HUCB-MSCs were injected intraperitoneally 24 h before surgery and during reperfusion. Renal functional and histological changes, HUCB-MSC trafficking, leukocyte infiltration, and cytokine expression were analyzed. Renal functional decline and tubular injury after IRI were attenuated by HUCB-MSC treatment. PKH-26-labeled HUCB-MSCs trafficked into the postischemic kidney. Although numbers of CD45-positive leukocytes in the postischemic kidney were comparable between groups, the expression of interferon-γ in the postischemic kidney was suppressed by HUCB-MSC treatment. The rapid decrease in intrarenal VEGF after IRI was markedly mitigated by HUCB-MSC treatment. In inflammatory conditions simulated in a cell culture experiment, VEGF secretion from HUCB-MSCs was substantially enhanced. VEGF inhibitor abolished the renoprotective effect of HUCB-MSCs after IRI. Flow cytometry analysis revealed the decreased infiltration of natural killer T cells and increased number of regulatory T cells in postischemic kidneys. In addition, these effects of HUCB-MSCs on kidney infiltrating mononuclear cells after IRI were attenuated by VEGF inhibitor. HUCB-MSCs attenuated renal injury in mice in the early injury phase after IRI, mainly by humoral effects and secretion of VEGF. Our results suggest a promising role for HUCB-MSCs in the treatment of renal IRI.
Collapse
Affiliation(s)
- Hye Ryoun Jang
- Nephrology Division, Department of Medicine, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ji Hyeon Park
- Nephrology Division, Department of Medicine, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ghee Young Kwon
- Department of Pathology, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea; and
| | - Jung Eun Lee
- Nephrology Division, Department of Medicine, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Wooseong Huh
- Nephrology Division, Department of Medicine, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Jin Jin
- Biomedical Research Institute, MEDIPOST Company Limited, Seoul, Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Company Limited, Seoul, Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Company Limited, Seoul, Korea
| | - Ha Young Oh
- Nephrology Division, Department of Medicine, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoon-Goo Kim
- Nephrology Division, Department of Medicine, Samsung Medical Center, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Seoul, Korea;
| |
Collapse
|
89
|
Pavlosky A, Lau A, Su Y, Lian D, Huang X, Yin Z, Haig A, Jevnikar AM, Zhang ZX. RIPK3-mediated necroptosis regulates cardiac allograft rejection. Am J Transplant 2014; 14:1778-1790. [PMID: 24984764 DOI: 10.1111/ajt.12779] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/02/2014] [Accepted: 04/04/2014] [Indexed: 01/25/2023]
Abstract
Cell death results in tissue damage and ultimately donor graft rejection and can occur as an active molecular process through apoptotic, necrotic and newly identified receptor interacting protein 1 and 3 kinase (RIPK1/3)-mediated necroptotic pathways. Necroptosis leads to the release of inflammatory molecules which can activate host immune cells. This pathway has yet to be studied in heart transplantation. We have found that necroptosis was induced in murine cardiac microvascular endothelial cell (MVEC) under anti-apoptotic condition following tumor necrosis factor alpha treatment. Necroptotic cell death and release of the danger molecule high mobility group box 1 (HMGB1) were inhibited by the RIPK1 inhibiting molecule necrostatin-1 and by genetic deletion of RIPK3. In addition, tissue necrosis, release of HMGB1 and graft cell infiltrate were attenuated in RIPK3 null heart allografts following transplantation. Finally, a brief sirolimus treatment markedly prolonged RIPK3 null cardiac allograft survival in allogeneic BALB/c recipients as compared to WT C57BL/6 donor grafts (95 ± 5.8 vs. 24 ± 2.6 days, p < 0.05). This study has demonstrated that RIPK1/3 contributes to MVEC death and cardiac allograft survival through necroptotic death and the release of danger molecules. Our results suggest that targeting RIPK-mediated necroptosis may be an important therapeutic strategy in transplantation.
Collapse
Affiliation(s)
- A Pavlosky
- Matthew Mailing Centre for Translational Transplantation Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada; Department of Pathology, Western University, London, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Lau A, Wang S, Liu W, Haig A, Zhang ZX, Jevnikar AM. Glycyrrhizic acid ameliorates HMGB1-mediated cell death and inflammation after renal ischemia reperfusion injury. Am J Nephrol 2014; 40:84-95. [PMID: 25059568 DOI: 10.1159/000364908] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/24/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND Renal ischemia reperfusion injury (IRI) leads to acute kidney injury (AKI) and the death of tubular epithelial cells (TEC). The release of high-mobility group box-1 (HMGB1) and other damage-associated molecular pattern moieties from dying cells may promote organ dysfunction and inflammation by effects on TEC. Glycyrrhizic acid (GZA) is a functional inhibitor of HMGB1, but its ability to attenuate the HMGB1-mediated injury of TEC has not been tested. METHODS/RESULTS In vitro, hypoxia and cytokine treatment killed TEC and resulted in the progressive release of HMGB1 into the supernatant. GZA reduced the hypoxia-induced TEC death as measured by annexin-V and propidium iodide. Hypoxia increased the expression of MCP-1 and CXCL1 in TEC, which was reduced by GZA in a dose-dependent manner. Similarly, the HMGB1 activation of effector NK cells was inhibited by GZA. To test the effect of HMGB1 neutralization by GZA in vivo, mice were subjected to renal IRI. HMGB1 protein expression increased progressively in kidneys from 4 to 24 h after ischemia and was detected in tubular cells by 4 h using immunohistochemistry. GZA preserved renal function after IRI and reduced tubular necrosis and neutrophil infiltration by histological analyses and ethidium homodimer staining. CONCLUSIONS Importantly, these data demonstrate for the first time that AKI following hypoxia and renal IRI may be promoted by HMGB1 release, which can reduce the survival of TEC and augment inflammation. Inhibition of the interaction of HMGB1 with TEC through GZA may represent a therapeutic strategy for the attenuation of renal injury following IRI and transplantation.
Collapse
Affiliation(s)
- Arthur Lau
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Ont., Canada
| | | | | | | | | | | |
Collapse
|
91
|
Molitoris BA. Therapeutic translation in acute kidney injury: the epithelial/endothelial axis. J Clin Invest 2014; 124:2355-63. [PMID: 24892710 PMCID: PMC4089444 DOI: 10.1172/jci72269] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Acute kidney injury (AKI) remains a major clinical event with rising incidence, severity, and cost; it now has a morbidity and mortality exceeding acute myocardial infarction. There is also a documented conversion to and acceleration of chronic kidney disease to end-stage renal disease. The multifactorial nature of AKI etiologies and pathophysiology and the lack of diagnostic techniques have hindered translation of preclinical success. An evolving understanding of epithelial, endothelial, and inflammatory cell interactions and individualization of care will result in the eventual development of effective therapeutic strategies. This review focuses on epithelial and endothelial injury mediators, interactions, and targets for therapy.
Collapse
|
92
|
Slegtenhorst BR, Dor FJ, Rodriguez H, Voskuil FJ, Tullius SG. Ischemia/reperfusion Injury and its Consequences on Immunity and Inflammation. CURRENT TRANSPLANTATION REPORTS 2014; 1:147-154. [PMID: 25419507 DOI: 10.1007/s40472-014-0017-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Ischemia/reperfusion injury (IRI), an inherent component of transplantation, affects organ quality and transplant outcomes. Although the complexity of the pathophysiology is recognized, detailed mechanisms remain unclear, and strategies preventing the consequences of IRI have been challenging. Of critical significance appears the link between IRI, the initiation of innate immune responses, and the (potential) augmentation of adaptive immunity. An improved understanding of those complex mechanisms and interactions may pave the way for more effective treatment strategies.
Collapse
Affiliation(s)
- Bendix R Slegtenhorst
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School ; Division of Transplant Surgery, Department of Surgery, Erasmus MC-University Medical Center
| | - Frank Jmf Dor
- Division of Transplant Surgery, Department of Surgery, Erasmus MC-University Medical Center
| | - Hector Rodriguez
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School ; Department of Cardiovascular Surgery, University Hospital of Zurich
| | - Floris J Voskuil
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School ; Division of Nephrology, Renal Transplant Unit, Department of Medicine, University Medical Center Groningen
| | - Stefan G Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School
| |
Collapse
|
93
|
Gandhi R, Yi J, Ha J, Shi H, Ismail O, Nathoo S, Bonventre JV, Zhang X, Gunaratnam L. Accelerated receptor shedding inhibits kidney injury molecule-1 (KIM-1)-mediated efferocytosis. Am J Physiol Renal Physiol 2014; 307:F205-21. [PMID: 24829508 DOI: 10.1152/ajprenal.00638.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Efficient clearance of apoptotic cells (efferocytosis) prevents inflammation and permits repair following tissue injury. Kidney injury molecule-1 (KIM-1) is a receptor for phosphatidylserine, an "eat-me" signal exposed on the surface of apoptotic cells that marks them for phagocytic clearance. KIM-1 is upregulated on proximal tubule epithelial cells (PTECs) during ischemic acute kidney injury (AKI), enabling efferocytosis by surviving PTECs. KIM-1 is spontaneously cleaved at its ectodomain region to generate a soluble fragment that serves a sensitive and specific biomarker for AKI, but the biological relevance of KIM-1 shedding is unknown. Here, we sought to determine how KIM-1 shedding might regulate efferocytosis. Using cells that endogenously and exogenously express KIM-1, we found that hydrogen peroxide-mediated oxidative injury or PMA treatment accelerated KIM-1 shedding in a dose-dependent manner. KIM-1 shedding was also accelerated when apoptotic cells were added. Accelerated shedding or the presence of excess soluble KIM-1 in the extracellular milieu significantly inhibited efferocytosis. We also identified that TNF-α-converting enzyme (TACE or ADAM17) mediates both the spontaneous and PMA-accelerated shedding of KIM-1. While accelerated shedding inhibited efferocytosis, we found that spontaneous KIM-1 cleavage does not affect the phagocytic efficiency of PTECs. Our results suggest that KIM-1 shedding is accelerated by worsening cellular injury, and excess soluble KIM-1 competitively inhibits efferocytosis. These findings may be important in AKI when there is severe cellular injury.
Collapse
Affiliation(s)
- Rushi Gandhi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - James Yi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Jihyen Ha
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Hang Shi
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Centre for Human Immunology, Western University, London, Ontario, Canada; and
| | - Ola Ismail
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Sahra Nathoo
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Xizhong Zhang
- Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Lakshman Gunaratnam
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada; Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, London, Ontario, Canada; Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada; Centre for Human Immunology, Western University, London, Ontario, Canada; and
| |
Collapse
|
94
|
Anders HJ. Immune system modulation of kidney regeneration--mechanisms and implications. Nat Rev Nephrol 2014; 10:347-58. [PMID: 24776845 DOI: 10.1038/nrneph.2014.68] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The immune system is an important guardian of tissue homeostasis. In response to injury, resident and infiltrating immune cells orchestrate all phases of danger control, resolution of inflammation and tissue regeneration or scar formation. As mammalian postnatal kidneys are not capable of de novo nephrogenesis, recovery is limited to the regeneration or repair of existing nephrons. The regenerative capacity of the nephron varies between compartments; the epithelial cells of the tubule regenerate more efficiently than the structurally highly organized podocytes. Cells of the surrounding environment modulate nephron regeneration by secreting paracrine mediators. This Review discusses immune mediators and pathways that regulate the intrinsic regenerative capacity of the nephron. Eliminating injurious triggers, modulating renal inflammation and specifically enhancing the regenerative capacity of nephrons might be a promising strategy to improve long-term outcomes in patients with acute kidney injury and/or chronic kidney disease.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München-Innenstadt, Ziemssenstrasse 1, 80336 Munich, Germany
| |
Collapse
|
95
|
The regulation of inflammatory mediators in acute kidney injury via exogenous mesenchymal stem cells. Mediators Inflamm 2014; 2014:261697. [PMID: 24839354 PMCID: PMC4009277 DOI: 10.1155/2014/261697] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/07/2014] [Accepted: 03/20/2014] [Indexed: 12/31/2022] Open
Abstract
Acute kidney injury (AKI) remains to be an independent risk factor for mortality and morbidity. Inflammation is believed to play a major role in the pathophysiology of AKI. Exogenous mesenchymal stem cells (MSCs) are now under extensive investigation as a potential therapy for AKI. Various preclinical studies indicated the beneficial effects of MSCs in alleviating renal injury and accelerating tissue repair. However the mechanisms responsible for these effects are incompletely understood. In the recent years, anti-inflammatory/immunoregulatory properties of MSCs have become one of the important issues in the treatment of AKI. This review will summarize the current literature on the regulation of inflammatory mediators via exogenous MSCs contributing to the recovery from AKI.
Collapse
|
96
|
Kim HJ, Park SJ, Koo S, Cha HJ, Lee JS, Kwon B, Cho HR. Inhibition of kidney ischemia-reperfusion injury through local infusion of a TLR2 blocker. J Immunol Methods 2014; 407:146-50. [PMID: 24681240 DOI: 10.1016/j.jim.2014.03.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 03/18/2014] [Accepted: 03/18/2014] [Indexed: 10/25/2022]
Abstract
Kidney ischemia-reperfusion injury (IRI) occurs as a result of complex interactions of kidney parenchymal cells and immune cells that are initiated by hypoxic damage of parenchymal cells. In particular, tubular epithelial cells (TECs) not only are susceptible to ischemia but also have an auto-loop system to amplify renal inflammation caused by ischemia and reperfusion. Since endogenous TLR2 ligands released from TECs trigger renal inflammation leading to kidney IRI in an autocrine manner, we hypothesized that local infusion of TLR2 blockers would prevent kidney IRI. In this study, we demonstrated that injection of antagonist anti-TLR2 mAb through the renal vein after cross-clamping significantly reduced the recruitment of NK cells to the kidney after IRI, a phenomenon that is governed by TLR2 signaling in TECs. In addition, intrarenal blocking of TLR2 signaling was shown to inhibit NK cell-mediated neutrophil infiltration and subsequent renal damage. Overall, our simple experiment system will be of help in testing the efficacy of candidate blockers targeting kidney parenchymal cells in inhibition of kidney IRI.
Collapse
Affiliation(s)
- Hye J Kim
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan, Republic of Korea
| | - Sang J Park
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan, Republic of Korea; Department of Surgery, Ulsan University Hospital and School of Medicine, University of Ulsan, Republic of Korea
| | - Sumi Koo
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan, Republic of Korea
| | - Hee J Cha
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan, Republic of Korea; Department of Pathology, Ulsan University Hospital, University of Ulsan, Republic of Korea
| | - Jong S Lee
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan, Republic of Korea; Department of Internal Medicine, Ulsan University Hospital and School of Medicine, University of Ulsan, Republic of Korea
| | - Byungsuk Kwon
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan, Republic of Korea; School of Biological Sciences, University of Ulsan, Ulsan, Republic of Korea.
| | - Hong R Cho
- Biomedical Research Center, Ulsan University Hospital, University of Ulsan, Republic of Korea; Department of Surgery, Ulsan University Hospital and School of Medicine, University of Ulsan, Republic of Korea.
| |
Collapse
|
97
|
Abstract
Organ transplantation appears today to be the best alternative to replace the loss of vital organs induced by various diseases. Transplants can, however, also be rejected by the recipient. In this review, we provide an overview of the mechanisms and the cells/molecules involved in acute and chronic rejections. T cells and B cells mainly control the antigen-specific rejection and act either as effector, regulatory, or memory cells. On the other hand, nonspecific cells such as endothelial cells, NK cells, macrophages, or polymorphonuclear cells are also crucial actors of transplant rejection. Last, beyond cells, the high contribution of antibodies, chemokines, and complement molecules in graft rejection is discussed in this article. The understanding of the different components involved in graft rejection is essential as some of them are used in the clinic as biomarkers to detect and quantify the level of rejection.
Collapse
Affiliation(s)
- Aurélie Moreau
- INSERM UMR 1064, Center for Research in Transplantation and Immunology-ITUN, CHU de Nantes 44093, France
| | | | | | | |
Collapse
|
98
|
Lau A, Wang S, Jiang J, Haig A, Pavlosky A, Linkermann A, Zhang ZX, Jevnikar AM. RIPK3-mediated necroptosis promotes donor kidney inflammatory injury and reduces allograft survival. Am J Transplant 2013; 13:2805-2818. [PMID: 24103001 DOI: 10.1111/ajt.12447] [Citation(s) in RCA: 260] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 06/12/2013] [Accepted: 07/02/2013] [Indexed: 01/25/2023]
Abstract
Kidney transplant injury occurs with ischemia and alloimmunity. Members of the receptor interacting protein kinase family (RIPK1,3) are key regulators of "necroptosis," a newly recognized, regulated form of necrosis. Necroptosis and apoptosis death appear to be counterbalanced as caspase-8 inhibition can divert death from apoptosis to necrosis. Inhibition of necroptosis in donor organs to limit injury has not been studied in transplant models. In this study, necroptosis was triggered in caspase inhibited tubular epithelial cells (TEC) exposed to tumor necrosis factor alpha in vitro, while RIPK1 inhibition with necrostatin-1 or use of RIPK3(-/-) TEC, prevented necroptosis. In vivo, short hairpin RNA silencing of caspase-8 in donor B6 mouse kidneys increased necroptosis, enhanced high-mobility group box 1 release, reduced renal function and accelerated rejection when transplanted into BALB/c recipients. Using ethidium homodimer perfusion to assess necrosis in vivo, necrosis was abrogated in RIPK3(-/-) kidneys postischemia. Following transplantation, recipients receiving RIPK3(-/-) kidneys had longer survival (p = 0.002) and improved renal function (p = 0.03) when compared to controls. In summary, we show for the first time that RIPK3-mediated necroptosis in donor kidneys can promote inflammatory injury, and has a major impact on renal ischemia-reperfusion injury and transplant survival. We suggest inhibition of necroptosis in donor organs may similarly provide a major clinical benefit.
Collapse
Affiliation(s)
- A Lau
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada; Department of Pathology, Western University, London, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Kim HJ, Lee JS, Kim A, Koo S, Cha HJ, Han JA, Do Y, Kim KM, Kwon BS, Mittler RS, Cho HR, Kwon B. TLR2 signaling in tubular epithelial cells regulates NK cell recruitment in kidney ischemia-reperfusion injury. THE JOURNAL OF IMMUNOLOGY 2013; 191:2657-64. [PMID: 23904170 DOI: 10.4049/jimmunol.1300358] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Damage-associated molecular patterns released from damaged kidney cells initiate postischemic inflammation, an essential step in the progression of kidney ischemia-reperfusion injury (IRI). However, the mechanism that coordinates this highly specific process in ischemic kidneys remains to be clarified. Previously, we demonstrated that CD137 from NK cells specifically stimulates CD137 ligand (CD137L) on tubular epithelial cells (TECs) such that TECs produced the high CXCR2 chemokine levels required for neutrophil chemotaxis. We report in the present study that endogenous TLR2 ligands released from ischemic TECs induce CCR5 chemokine expression, which is critical to promoting NK cell recruitment. By implanting CD137L(-/-) TECs into the kidney capsule of TLR2(-/-) mice, we further showed that TLR2-mediated NK cell recruitment is an uncoupled event that can occur independently of CD137L signaling in TECs, which is responsible for recruiting neutrophils. Therefore, our findings identify TECs as both a target for kidney damage and also as a master regulator that actively modulates stepwise signaling, leading to the initiation and amplification of acute sterile inflammation that inflicts kidney IRI. Being clinically important, the signaling pathway of innate receptors in epithelial cells may therefore be a good target to block acute sterile inflammation resulting from tissue damage, including kidney IRI.
Collapse
Affiliation(s)
- Hye J Kim
- Biomedical Research Center, Ulsan University Hospital, School of Medicine, University of Ulsan, Ulsan 682-714, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
Chronic and acute renal diseases, irrespective of the initiating cause, have inflammation and immune system activation as a common underlying mechanism. The purpose of this review is to provide a broad overview of immune cells and inflammatory proteins that contribute to the pathogenesis of renal disease, and to discuss some of the physiological changes that occur in the kidney as a result of immune system activation. An overview of common forms of acute and chronic renal disease is provided, followed by a discussion of common therapies that have anti-inflammatory or immunosuppressive effects in the treatment of renal disease.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| | | |
Collapse
|