1
|
Chen Q, Guo J, Han S, Wang T, Xia K, Yu B, Lu Y, Qiu T, Zhou J. Cordycepin alleviates renal ischemia-reperfusion injury by suppressing the p38/JNK signaling pathway. Int Immunopharmacol 2025; 150:114264. [PMID: 39954658 DOI: 10.1016/j.intimp.2025.114264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Renal ischemia-reperfusion injury (IRI) makes a significant contribution to delayed graft function (DGF) and reduced allograft survival time post-transplantation, thereby complicating the prognosis of renal transplant recipients. Cordycepin, an active compound purified from the traditional Chinese medicine Cordyceps sinensis, has exhibited remarkable anti-inflammatory and organ-protective effects against various diseases, including neurological, hepatic, and metabolic disorders. Therefore, the present study used a murine model of renal ischemia/reperfusion (I/R) and HK2 cell line hypoxia/reoxygenation (H/R) to determine whether cordycepin influences renal IRI. The findings indicated that cordycepin significantly mitigated renal IRI by inhibiting the p38/JNK signaling pathway in the renal tubular epithelial cells, thereby suppressing inflammation, cell apoptosis, and ferroptosis. These findings offer a novel avenue for improving the prognosis of renal transplant recipients and allograft survival.
Collapse
Affiliation(s)
- Qi Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China; National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| | - Shangting Han
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Tianyu Wang
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Kang Xia
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Bo Yu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Yifan Lu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China.
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University ,Wuhan, Hubei 430060, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei 430060, China.
| |
Collapse
|
2
|
de Boer E, Sokolova M, Jager NM, Schjalm C, Weiss MG, Liavåg OM, Maassen H, van Goor H, Thorgersen EB, Pettersen K, Christiansen D, Ludviksen JK, Jespersen B, Mollnes TE, Leuvenink HGD, Pischke SE. Normothermic Machine Perfusion Reconstitutes Porcine Kidney Tissue Metabolism But Induces an Inflammatory Response, Which Is Reduced by Complement C5 Inhibition. Transpl Int 2024; 37:13348. [PMID: 39606689 PMCID: PMC11598510 DOI: 10.3389/ti.2024.13348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024]
Abstract
Normothermic machine perfusion (NMP) is a clinical strategy to reduce renal ischemia-reperfusion injury (IRI). Optimal NMP should restore metabolism and minimize IRI induced inflammatory responses. Microdialysis was used to evaluate renal metabolism. This study aimed to assess the effect of complement inhibition on NMP induced inflammatory responses. Twenty-two pig kidneys underwent 18 h of static cold storage (SCS) followed by 4 h of NMP using a closed-circuit system. Kidneys were randomized to receive a C5-inhibitor or placebo during SCS and NMP. Perfusion resulted in rapidly stabilized renal flow, low renal resistance, and urine production. During SCS, tissue microdialysate levels of glucose and pyruvate decreased significantly, whereas glycerol increased (p < 0.001). In the first hour of NMP, glucose and pyruvate increased while glycerol decreased (p < 0.001). After 4 h, all metabolites had returned to baseline. Inflammatory markers C3a, soluble C5b-9, TNF, IL-6, IL-1β, IL-8, and IL-10 increased significantly during NMP in perfusate and kidney tissue. C5-inhibition significantly decreased perfusate and urine soluble C5b-9 (p < 0.001; p = 0.002, respectively), and tissue IL-1β (p = 0.049), but did not alter other inflammatory markers. Microdialysis can accurately monitor the effect of NMP on renal metabolism. Closed-circuit NMP induces inflammation, which appeared partly complement-mediated. Targeting additional immune inhibitors should be the next step.
Collapse
Affiliation(s)
- Eline de Boer
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marina Sokolova
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Neeltina M. Jager
- Department of Surgery, Division of Organ Donation and Transplantation, University Medical Center Groningen, Groningen, Netherlands
| | - Camilla Schjalm
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marc G. Weiss
- Department of Medicine and Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | - Olav M. Liavåg
- Section for Transplantation Surgery, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Hanno Maassen
- Department of Surgery, Division of Organ Donation and Transplantation, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Ebbe Billmann Thorgersen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Department of Gastroenterological Surgery, Oslo University Hospital the Radium Hospital, Oslo, Norway
| | | | | | | | - Bente Jespersen
- Department of Medicine and Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | - Tom E. Mollnes
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodø, Norway
- Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - Henri G. D. Leuvenink
- Department of Surgery, Division of Organ Donation and Transplantation, University Medical Center Groningen, Groningen, Netherlands
- Department of Medicine and Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | - Søren E. Pischke
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Emergencies and Critical Care, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
3
|
Idouz K, Belhaj A, Rondelet B, Dewachter L, Flamion B, Kirschvink N, Dogné S. Cascading renal injury after brain death: Unveiling glycocalyx alteration and the potential protective role of tacrolimus. Front Cell Dev Biol 2024; 12:1449209. [PMID: 39165663 PMCID: PMC11333349 DOI: 10.3389/fcell.2024.1449209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
Brain death (BD) is a complex medical state that triggers systemic disturbances and a cascade of pathophysiological processes. This condition significantly impairs both kidney function and structural integrity, thereby presenting considerable challenges to graft viability and the long-term success of transplantation endeavors. Tacrolimus (FK506), an immunosuppressive drug, was used in this study to assess its impact as a pretreatment on brain death-induced renal injury. This study aimed to investigate changes associated with brain death-induced renal injury in a 4-month-old female porcine model. The experimental groups included brain death placebo-pretreated (BD; n = 9), brain death tacrolimus-pretreated using the clinical dose of 0.25 mg/kg the day before surgery, followed by 0.05 mg/kg/day 1 hour before the procedure (BD + FK506; n = 8), and control (ctrl, n = 7) piglets, which did not undergo brain death induction. Furthermore, we aimed to assess the effect of FK506 on these renal alterations through graft preconditioning. We hypothesized that immunosuppressive properties of FK506 reduce tissue inflammation and preserve the glycocalyx. Our findings revealed a series of interconnected events triggered by BD, leading to a deterioration of renal function and increased proteinuria, increased apoptosis in the vessels, glomeruli and tubules, significant leukocyte infiltration into renal tissue, and degradation of the glycocalyx in comparison with ctrl group. Importantly, treatment with FK506 demonstrated significant efficacy in attenuating these adverse effects. FK506 helped reduce apoptosis, maintain glycocalyx integrity, regulate neutrophil infiltration, and mitigate renal injury following BD. This study offers new insights into the pathophysiology of BD-induced renal injury, emphasizing the potential of FK506 pretreatment as a promising therapeutic intervention for organ preservation, through maintaining endothelial function with the additional benefit of limiting the risk of rejection.
Collapse
Affiliation(s)
- Kaoutar Idouz
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (Unamur), Namur, Belgium
| | - Asmae Belhaj
- Department of Cardio-Vascular, Thoracic Surgery and Lung Transplantation, CHU UCL Namur, UCLouvain, Yvoir, Belgium
| | - Benoit Rondelet
- Department of Cardio-Vascular, Thoracic Surgery and Lung Transplantation, CHU UCL Namur, UCLouvain, Yvoir, Belgium
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Université Libre de Bruxelles, Brussels, Belgium
| | - Bruno Flamion
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (Unamur), Namur, Belgium
- Clinical Development, Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Nathalie Kirschvink
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (Unamur), Namur, Belgium
| | - Sophie Dogné
- Molecular Physiology Research Unit (URPhyM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (Unamur), Namur, Belgium
| |
Collapse
|
4
|
Rao JS, Pruett TL. Immunology of the transplanted cryopreserved kidney. Cryobiology 2023; 110:1-7. [PMID: 36640932 DOI: 10.1016/j.cryobiol.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/28/2022] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Transplantation has substituted dysfunctional organs with healthy organs from donors to significantly lower morbidity and mortality associated with end-stage organ disease. Since the advent of transplantation, the promise of functional replacement has attracted an exponential mismatch between organ supply and demand. Theoretical proposals to counter the increasing needs have either been to create a source through genetic engineering of porcine donors for xenotransplantation (with more potent immunosuppression protocols) or recreate one's organ in a pig using interspecies blastocyst complementation for exogenic organ transplantation (without immunosuppression). Another promising avenue has been organ banking through cryopreservation for transplantation. Although ice free preservation and acceptable early function following rewarming is critical for success in transplantation, the immunological response that predominantly defines short- and long-term graft survival has failed to captivate attention to date. It is well sorted that thermal and metabolic stress incurred at 4 °C during recovery and reperfusion of organs for clinical transplantation has varying impact on graft survival. Considering the magnitude of cellular imbalance and injury at sub-zero/ultralow temperatures in addition to the chemical toxicity of cryoprotective agents (CPA), it is essential to assess and address the immunological response associated following transplantation to maximize the success of cryopreservation.
Collapse
Affiliation(s)
- Joseph Sushil Rao
- Division of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, USA; Schulze Diabetes Institute, Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| | - Timothy L Pruett
- Division of Solid Organ Transplantation, Department of Surgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
5
|
Shepherd HM, Gauthier JM, Terada Y, Li W, Krupnick AS, Gelman AE, Kreisel D. Updated Views on Neutrophil Responses in Ischemia-Reperfusion Injury. Transplantation 2022; 106:2314-2324. [PMID: 35749228 PMCID: PMC9712152 DOI: 10.1097/tp.0000000000004221] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ischemia-reperfusion injury is an inevitable event during organ transplantation and represents a primary risk factor for the development of early graft dysfunction in lung, heart, liver, and kidney transplant recipients. Recent studies have implicated recipient neutrophils as key mediators of this process and also have found that early innate immune responses after transplantation can ultimately augment adaptive alloimmunity and affect late graft outcomes. Here, we discuss signaling pathways involved in neutrophil recruitment and activation after ischemia-mediated graft injury in solid organ transplantation with an emphasis on lung allografts, which have been the focus of recent studies. These findings suggest novel therapeutic interventions that target ischemia-reperfusion injury-mediated graft dysfunction in transplant recipients.
Collapse
Affiliation(s)
- Hailey M. Shepherd
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Jason M. Gauthier
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Yuriko Terada
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | - Wenjun Li
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | | | - Andrew E. Gelman
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
6
|
Maines LW, Green CL, Keller SN, Fitzpatrick LR, Smith CD. The Sphingosine Kinase 2 Inhibitor Opaganib Protects Against Acute Kidney Injury in Mice. Int J Nephrol Renovasc Dis 2022; 15:323-334. [PMID: 36420520 PMCID: PMC9677921 DOI: 10.2147/ijnrd.s386396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Acute kidney injury (AKI) is a common multifactorial adverse effect of surgery, circulatory obstruction, sepsis or drug/toxin exposure that often results in morbidity and mortality. Sphingolipid metabolism is a critical regulator of cell survival and pathologic inflammation processes involved in AKI. Opaganib (also known as ABC294640) is a first-in-class experimental drug targeting sphingolipid metabolism that reduces the production and activity of inflammatory cytokines and, therefore, may be effective to prevent and treat AKI. Methods Murine models of AKI were used to assess the in vivo efficacy of opaganib including ischemia-reperfusion (IR) injury induced by either transient bilateral occlusion of renal blood flow (a moderate model) or nephrectomy followed immediately by occlusion of the contralateral kidney (a severe model) and lipopolysaccharide (LPS)-induced sepsis. Biochemical and histologic assays were used to quantify the effects of oral opaganib treatment on renal damage in these models. Results Opaganib suppressed the elevations of creatinine and blood urea nitrogen (BUN), as well as granulocyte infiltration into the kidneys, of mice that experienced moderate IR from transient bilateral ligation. Opaganib also markedly decreased these parameters and completely prevented mortality in the severe renal IR model. Additionally, opaganib blunted the elevations of BUN, creatinine and inflammatory cytokines following exposure to LPS. Conclusion The data support the hypotheses that sphingolipid metabolism is a key mediator of renal inflammatory damage following IR injury and sepsis, and that this can be suppressed by opaganib. Because opaganib has already undergone clinical testing in other diseases (cancer and Covid-19), the present studies support conducting clinical trials with this drug with surgical or septic patients at risk for AKI.
Collapse
Affiliation(s)
- Lynn W Maines
- Apogee Biotechnology Corporation, Hummelstown, PA, USA
| | | | | | | | - Charles D Smith
- Apogee Biotechnology Corporation, Hummelstown, PA, USA
- Correspondence: Charles D Smith, Apogee Biotechnology Corporation, 1214 Research Blvd, Suite 2015, Hummelstown, PA, 17036, USA, Email
| |
Collapse
|
7
|
Franchon Marques Tejada N, Ziroldo Lopes JV, Duarte Gonçalves LE, Mamede Costa Andrade da Conceição I, Franco GR, Ghirotto B, Câmara NOS. AIM2 as a putative target in acute kidney graft rejection. Front Immunol 2022; 13:839359. [PMID: 36248890 PMCID: PMC9561248 DOI: 10.3389/fimmu.2022.839359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Acute rejection (AR) is a process triggered via the recognition of grafted organ-derived antigens by the immune system, which could present as a life-threatening condition. In the context of a kidney transplant, despite improvement with immunosuppressive therapies, AR maintains a significant incidence of 10%, and currently available drugs generally act in similar and canonical pathways of lymphocyte activation. This prompted the research for different approaches to identify potential novel targets that could improve therapeutic interventions. Here, we conducted a transcriptome analysis comparing groups of acute rejection (including T cell-mediated rejection and antibody-mediated rejection) to stable grafts that included differentially expressed genes, transcription factor and kinase enrichment, and Gene Set Enrichment Analysis. These analyses revealed inflammasome enhancement in rejected grafts and AIM2 as a potential component linked to acute rejection, presenting a positive correlation to T-cell activation and a negative correlation to oxidative phosphorylation metabolism. Also, the AIM2 expression showed a global accuracy in discerning acute rejection grafts (area under the curve (AUC) = 0.755 and 0.894, p < 0.0001), and meta-analysis comprising different studies indicated a considerable enhancement of AIM2 in rejection (standardized mean difference (SMD) = 1.45, [CI 95%, 1.18 to 1.71]), especially for T cell-mediated rejection (TCMR) (SMD = 2.01, [CI 95%, 1.58 to 2.45]). These findings could guide future studies of AIM2 as either an adjuvant target for immunosuppression or a potential biomarker for acute rejection and graft survival.
Collapse
Affiliation(s)
- Nathália Franchon Marques Tejada
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - João Vitor Ziroldo Lopes
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Luis Eduardo Duarte Gonçalves
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Izabela Mamede Costa Andrade da Conceição
- Laboratory of Biochemical Genetics, Department of Biochemistry and Immunology, Institute of Biomedical Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Glória Regina Franco
- Laboratory of Biochemical Genetics, Department of Biochemistry and Immunology, Institute of Biomedical Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Bruno Ghirotto
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
- Laboratory of Biochemical Genetics, Department of Biochemistry and Immunology, Institute of Biomedical Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Niels Olsen Saraiva Câmara, ;
| |
Collapse
|
8
|
Sarioglu S, Tekin E, Unlu M, Yildiz S, Heybeli, MD C. Peritubular Capillaritis in Native Kidney Biopsies. Am J Clin Pathol 2022; 158:389-394. [PMID: 35639718 DOI: 10.1093/ajcp/aqac061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To determine the frequency and predictors of peritubular capillaritis (PTCitis) among native kidney biopsies. METHODS Consecutive native kidney biopsies of 169 patients were reexamined for capturing possible PTCitis according to the Banff Classification. The relation of PTCitis with demographic and clinicopathological findings was evaluated. Logistic regression analysis was performed to determine predictors of PTCitis. RESULTS Peritubular capillaritis was captured in 90 (53.3%) patients, with scores of 1, 2, and 3 in 57 (33.7%), 31 (18.3%), and 2 (1.2%) patients, respectively. The highest frequency of PTCitis was observed in pauci-immune glomerulonephritis. In univariate analysis, male sex, the presence of interstitial inflammation, pauci-immune glomerulonephritis, and a higher serum creatinine level were associated with a higher risk of PTCitis, while severe interstitial fibrosis/tubular atrophy was associated with a lower risk. The presence of interstitial inflammation (odds ratio [OR], 5.94 [95% confidence interval (CI), 1.41-25.03]; P = .015), pauci-immune glomerulonephritis (OR, 3.08 [95% CI, 1.01-9.36]; P = .048), and a higher serum creatinine level (per 1 mg/dL) (OR, 1.56 [95% CI, 1.14-2.11]; P = .005) were independent predictors of PTCitis development in a multivariate regression model. CONCLUSIONS Peritubular capillaritis is common in native biopsies and more likely to be observed in the presence of interstitial inflammation, pauci-immune glomerulonephritis, and a higher serum creatinine level.
Collapse
Affiliation(s)
- Sulen Sarioglu
- Department of Pathology, Dokuz Eylül University Faculty of Medicine , İzmir , Turkey
| | - Emel Tekin
- Department of Pathology , Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Mehtat Unlu
- Department of Pathology, Dokuz Eylül University Faculty of Medicine , İzmir , Turkey
| | - Serkan Yildiz
- Division of Nephrology, Medicana Hospital , İzmir , Turkey
| | | |
Collapse
|
9
|
The Pathological and Clinical Diversity of Acute Vascular Rejection in Kidney Transplantation. Transplantation 2022; 106:1666-1676. [PMID: 35266923 DOI: 10.1097/tp.0000000000004071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Vascular rejection (VR) is characterized by arteritis, steroid resistance, and increased graft loss but is poorly described using modern diagnostics. METHODS We screened 3715 consecutive biopsies and retrospectively evaluated clinical and histological phenotypes of VR (n = 100) against rejection without arteritis (v0REJ, n = 540) and normal controls (n = 1108). RESULTS Biopsy sample size affected the likelihood of arterial sampling, VR diagnosis, and final Banff v scores (P < 0.001). Local v and cv scores were greatest in larger arteries (n = 258). VR comprised 15.6% of all rejection episodes, presented earlier (median 1.0 mo, interquartile range, 0.4-8 mo) with higher serum creatinine levels and inferior graft survival, versus v0REJ (P < 0.001). Early VR (≤1 mo) was common (54%) and predicted by sensitization, delayed function, and prior corticosteroid use, with associated acute dysfunction and optimal therapeutic response, independent of Banff v score. Late VR followed under-immunosuppression in 71.4% (noncompliance 38.8%, iatrogenic 32.6%), and was associated with chronic interstitial fibrosis, incomplete renal functional recovery and persistent inflammation using sequential histopathology. The etiology was "pure" antibody-mediated VR (n = 21), mixed VR (n = 36), and "pure" T cell-mediated VR (n = 43). Isolated VR (n = 34, Banff i < 1 without tubulitis) comprised 24 T cell-mediated VR and 10 antibody-mediated VR, presenting with mild renal dysfunction, minimal Banff acute scores, and better graft survival compared with inflamed VR. Interstitial inflammation influenced acute renal dysfunction and early treatment response, whereas chronic tubulointerstitial damage determined long-term graft loss. CONCLUSIONS VR is a heterogenous entity influenced by time-of-onset, pathophysiology, accompanying interstitial inflammation and fibrosis. Adequate histological sampling is essential for its accurate diagnostic classification and treatment.
Collapse
|
10
|
Piemonti L, Sordi V, Pellegrini S, Scotti GM, Scavini M, Sioli V, Gianelli Castiglione A, Cardillo M. Circulating CXCL10 and IL-6 in solid organ donors after brain death predict graft outcomes. Sci Rep 2021; 11:6624. [PMID: 33758270 PMCID: PMC7988181 DOI: 10.1038/s41598-021-86085-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 03/10/2021] [Indexed: 11/09/2022] Open
Abstract
We tested the hypothesis that circulating CXCL10 and IL-6 in donor after brain death provide independent additional predictors of graft outcome. From January 1, 2010 to June 30, 2012 all donors after brain death managed by the NITp (n = 1100) were prospectively included in this study. CXCL10 and IL-6 were measured on serum collected for the crossmatch at the beginning of the observation period. Graft outcome in recipients who received kidney (n = 1325, follow-up 4.9 years), liver (n = 815, follow-up 4.3 years) and heart (n = 272, follow-up 5 years) was evaluated. Both CXCL-10 and IL-6 showed increased concentration in donors after brain death. The intensive care unit stay, the hemodynamic instability, the cause of death, the presence of risk factors for cardiovascular disease and the presence of ongoing infection resulted as significant determinants of IL-6 and CXCL10 donor concentrations. Both cytokines resulted as independent predictors of Immediate Graft Function. Donor IL-6 or CXCL10 were associated with graft failure after liver transplant, and acted as predictors of recipient survival after kidney, liver and heart transplantation. Serum donor IL-6 and CXCL10 concentration can provide independent incremental prediction of graft outcome among recipients followed according to standard clinical practice.
Collapse
Affiliation(s)
- Lorenzo Piemonti
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy. .,Vita-Salute San Raffaele University, Milan, Italy.
| | - Valeria Sordi
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Silvia Pellegrini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giulia Maria Scotti
- Center for Translational Genomics and Bioinformatics, IRCCS San Raffaele Hospital, Milan, Italy
| | - Marina Scavini
- Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Viviana Sioli
- Transplant Coordination Unit, Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Massimo Cardillo
- Transplant Coordination Unit, Fondazione Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
11
|
Wlodek E, Kirkpatrick RB, Andrews S, Noble R, Schroyer R, Scott J, Watson CJE, Clatworthy M, Harrison EM, Wigmore SJ, Stevenson K, Kingsmore D, Sheerin NS, Bestard O, Stirnadel-Farrant HA, Abberley L, Busz M, DeWall S, Birchler M, Krull D, Thorneloe KS, Weber A, Devey L. A pilot study evaluating GSK1070806 inhibition of interleukin-18 in renal transplant delayed graft function. PLoS One 2021; 16:e0247972. [PMID: 33684160 PMCID: PMC7939287 DOI: 10.1371/journal.pone.0247972] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/11/2020] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Delayed graft function (DGF) following renal transplantation is a manifestation of acute kidney injury (AKI) leading to poor long-term outcome. Current treatments have limited effectiveness in preventing DGF. Interleukin-18 (IL18), a biomarker of AKI, induces interferon-γ expression and immune activation. GSK1070806, an anti-IL18 monoclonal antibody, neutralizes activated (mature) IL18 released from damaged cells following inflammasome activation. This phase IIa, single-arm trial assessed the effect of a single dose of GSK1070806 on DGF occurrence post donation after circulatory death (DCD) kidney transplantation. METHODS The 3 mg/kg intravenous dose was selected based on prior studies and physiologically based pharmacokinetic (PBPK) modeling, indicating the high likelihood of a rapid and high level of IL18 target engagement when administered prior to kidney allograft reperfusion. Utilization of a Bayesian sequential design with a background standard-of-care DGF rate of 50% based on literature, and confirmed via extensive registry data analyses, enabled a statistical efficacy assessment with a minimal sample size. The primary endpoint was DGF frequency, defined as dialysis requirement ≤7 days post transplantation (except for hyperkalemia). Secondary endpoints included safety, pharmacokinetics and pharmacodynamic biomarkers. RESULTS GSK1070806 administration was associated with IL18-GSK1070806 complex detection and increased total serum IL18 levels due to IL18 half-life prolongation induced by GSK1070806 binding. Interferon-γ-induced chemokine levels declined or remained unchanged in most patients. Although the study was concluded prior to the Bayesian-defined stopping point, 4/7 enrolled patients (57%) had DGF, exceeding the 50% standard-of-care rate, and an additional two patients, although not reaching the protocol-defined DGF definition, demonstrated poor graft function. Six of seven patients experienced serious adverse events (SAEs), including two treatment-related SAEs. CONCLUSION Overall, using a Bayesian design and extensive PBPK dose modeling with only a small sample size, it was deemed unlikely that GSK1070806 would be efficacious in preventing DGF in the enrolled DCD transplant population. TRIAL REGISTRATION NCT02723786.
Collapse
Affiliation(s)
- E. Wlodek
- GlaxoSmithKline, Clinical Unit Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - R. B. Kirkpatrick
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - S. Andrews
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - R. Noble
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - R. Schroyer
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - J. Scott
- JMS Statistics Ltd, Pinner, United Kingdom
| | - C. J. E. Watson
- University of Cambridge and the NIHR Cambridge Biomedical Research Centre and the NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation at the University of Cambridge, Cambridge, United Kingdom
| | - M. Clatworthy
- University of Cambridge and the NIHR Cambridge Biomedical Research Centre and the NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation at the University of Cambridge, Cambridge, United Kingdom
| | | | - S. J. Wigmore
- Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - K. Stevenson
- Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - D. Kingsmore
- Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - N. S. Sheerin
- Newcastle Biomedical Research Centre and the NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Newcastle University, Newcastle, United Kingdom
| | - O. Bestard
- L’Hospitalet de Llobregat, Bellvitge University Hospital, Kidney Transplant Unit, Barcelona, Spain
| | | | - L. Abberley
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - M. Busz
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - S. DeWall
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - M. Birchler
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - D. Krull
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - K. S. Thorneloe
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - A. Weber
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| | - L. Devey
- GlaxoSmithKline, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
12
|
Differential Impact of Delayed Graft Function in Deceased Donor Renal Transplant Recipients With and Without Donor-specific HLA-antibodies. Transplantation 2020; 103:e273-e280. [PMID: 31205266 DOI: 10.1097/tp.0000000000002802] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Delayed graft function (DGF) and pretransplant donor-specific HLA-antibodies (DSA) are both regarded as risk factors for rejection and lower graft survival. However, the combined impact of DGF and DSA has not been studied in detail. METHODS We investigated 375 deceased donor kidney transplantations, which had DSA assignment by single-antigen bead technology and which had surveillance biopsies at 3 of 6 months. Median follow-up time was 6.1 years. RESULTS DGF occurred in 137 of 375 patients (37%), and DSA were present in 85 of 375 patients (23%). The incidence of DGF was similar in DSA-positive (DSApos)-patients and DSA-negative (DSAneg)-patients (40% versus 36%; P = 0.45). In DSAneg-patients, 5-year graft survival was not different with/without DGF (81% versus 83%; P = 0.48). By contrast, in DSApos-patients, 5-year graft survival was significantly lower with DGF (64% versus 79%; P = 0.01). Moreover, DSApos-patients with DGF had a higher 1-year incidence of subclinical rejection, which were mostly antibody-mediated or mixed rejection phenotypes. Graft loss due to rejection was significantly more frequent in DSApos-patients with DGF (5/34; 15%) compared to DSApos-patients without DGF (2/51; 4%), and DSAneg-patients with/without DGF (3/103; 3% and 4/187; 2%, respectively) (P = 0.005). In a multivariate Cox model, DSA with DGF was an independent predictor for graft (hazard ratio = 2.84 [95% confidence interval, 1.54-5.06]; P = 0.001) and death-censored graft loss (hazard ratio = 4.65 [95% confidence interval, 1.83-11.51]; P = 0.002). CONCLUSIONS DGF has a much more detrimental impact in DSApos-patients than in DSAneg-patients, which is likely related to a higher incidence of antibody-mediated rejection. If possible, the combined risks of DGF and DSA should be avoided.
Collapse
|
13
|
Abstract
Acute kidney injury (AKI), a major public health problem associated with high mortality and increased risk of progression towards end-stage renal disease, is characterized by the activation of intra-renal haemostatic and inflammatory processes. Platelets, which are present in high numbers in the circulation and can rapidly release a broad spectrum of bioactive mediators, are important acute modulators of inflammation and haemostasis, as they are the first cells to arrive at sites of acute injury, where they interact with endothelial cells and leukocytes. Diminished control of platelet reactivity by endothelial cells and/or an increased release of platelet-activating mediators can lead to uncontrolled platelet activation in AKI. As increased platelet sequestration and increased expression levels of the markers P-selectin, thromboxane A2, CC-chemokine ligand 5 and platelet factor 4 on platelets have been reported in kidneys following AKI, platelet activation likely plays a part in AKI pathology. Results from animal models and some clinical studies highlight the potential of antiplatelet therapies in the preservation of renal function in the context of AKI, but as current strategies also affect other cell types and non-platelet-derived mediators, additional studies are required to further elucidate the extent of platelet contribution to the pathology of AKI and to determine the best therapeutic approach by which to specifically target related pathogenic pathways.
Collapse
Affiliation(s)
- Marcel P B Jansen
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sandrine Florquin
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.
| |
Collapse
|
14
|
Propofol can suppress renal ischemia-reperfusion injury through the activation of PI3K/AKT/mTOR signal pathway. Gene 2019; 708:14-20. [DOI: 10.1016/j.gene.2019.05.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 05/04/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022]
|
15
|
Barron M, Zhang S, Li J. A sparse differential clustering algorithm for tracing cell type changes via single-cell RNA-sequencing data. Nucleic Acids Res 2019; 46:e14. [PMID: 29140455 PMCID: PMC5815159 DOI: 10.1093/nar/gkx1113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 10/24/2017] [Indexed: 12/15/2022] Open
Abstract
Cell types in cell populations change as the condition changes: some cell types die out, new cell types may emerge and surviving cell types evolve to adapt to the new condition. Using single-cell RNA-sequencing data that measure the gene expression of cells before and after the condition change, we propose an algorithm, SparseDC, which identifies cell types, traces their changes across conditions and identifies genes which are marker genes for these changes. By solving a unified optimization problem, SparseDC completes all three tasks simultaneously. SparseDC is highly computationally efficient and demonstrates its accuracy on both simulated and real data.
Collapse
Affiliation(s)
- Martin Barron
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Siyuan Zhang
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, IN 46617, USA
| | - Jun Li
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA.,Mike and Josie Harper Cancer Research Institute, University of Notre Dame, IN 46617, USA
| |
Collapse
|
16
|
Ilmakunnas M, Turunen AJ, Lindgren L, Salmela KT, Kyllönen LE, Andersson S, Petäjä J, Pesonen EJ. Graft Neutrophil Sequestration and Concomitant Tissue Plasminogen Activator Release During Reperfusion in Clinical Kidney Transplantation. Transplant Proc 2019; 51:647-650. [PMID: 30979447 DOI: 10.1016/j.transproceed.2019.01.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 12/14/2018] [Accepted: 01/17/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND Inflammation, coagulation, and fibrinolysis are tightly linked together. Reperfusion after transient ischemia activates both neutrophils, coagulation, and fibrinolysis. Experimental data suggest that tissue plasminogen activator (tPA) regulates renal neutrophil influx in kidney ischemia and reperfusion injury. METHODS In 30 patients undergoing kidney transplantation, we measured renal neutrophil sequestration and tPA release from blood samples drawn from the supplying artery and renal vein early after reperfusion. tPA antigen levels were measured using a commercial enzyme-linked immunosorbent assay kit. For each parameter, transrenal difference (Δ) was calculated by subtracting the value of the arterial sample (ingoing blood) from the value of the venous sample (outgoing blood). RESULTS Positive transrenal gradients of tPA antigen occurred at 1 minute [Δ = 14 (3-46) ng/mL, P < .01] and 5 minutes [Δ = 5 (-3 to 27) ng/mL, P < .01] after reperfusion. At 5 minutes after reperfusion, a negative transrenal gradient of neutrophils was observed [Δ = -0.17 (-1.45 to 0.24) x 10E9 cells/L, P < .001]. At 1 minute after reperfusion, neutrophil sequestration into the kidney (ie, negative transrenal neutrophil count) correlated significantly with tPA release from the kidney (ie, positive transrenal tPA concentration), (R = -0.513 and P = .006). CONCLUSIONS The findings suggest a proinflammatory role for tPA in ischemia and reperfusion injury in human kidney transplantation.
Collapse
Affiliation(s)
- M Ilmakunnas
- Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| | - A J Turunen
- Kanta-Häme Central Hospital, Hämeenlinna, Finland
| | - L Lindgren
- Department of Anesthesia, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - K T Salmela
- Transplantation and Liver Surgery Clinic, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - L E Kyllönen
- Transplantation and Liver Surgery Clinic, Abdominal Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - S Andersson
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - J Petäjä
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - E J Pesonen
- Department of Anesthesiology, Intensive Care and Pain Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
17
|
Kortekaas KA, de Vries DK, Roest M, Reinders MEJ, van der Veer EP, Klautz RJM, de Groot PG, Schaapherder AF, Lindeman JH. No indications for platelet activation in acute clinical myocardial or renal ischemia/reperfusion injury. Am J Transl Res 2018; 10:816-826. [PMID: 29636871 PMCID: PMC5883122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 12/24/2017] [Indexed: 06/08/2023]
Abstract
The pathophysiology of ischemia/reperfusion (I/R) injury is complex and poorly understood. Animal studies imply platelet activation as an initiator of the inflammatory response upon reperfusion. However, it remains unclear whether and how these results translate to clinical I/R. This study evaluates putative platelet activation in the context of two forms of clinical I/R (heart valve surgery with aortic-cross clamping, n = 39 and kidney transplantation, n = 34). The technique of sequential selective arteriovenous (AV) measurements over the reperfused organs was applied to exclude the influence of systemic changes occurring during surgery while simultaneously maximizing sensitivity. Platelet activation and degranulation was evaluated by assessing the expression levels of established markers, i.e. RANTES (Regulated on Activation, Normal T Cell Expressed and Secreted), β-thromboglobulin (β-TG), platelet-derived growth factor (PDGF)-BB and CXCL8 (known as interleukin-8), and by employing an in-vitro assay that specifically tests for platelet excitability. Moreover, a histological analysis was performed by means of CD41 staining. Results show stable RANTES, β-TG, PDGF-BB and CXCL8 AV-concentrations within the first half hour over the reperfused organs, suggesting that myocardial and renal I/R are not associated with platelet activation. Results from the platelet excitability assay were in line with these findings and indicated reduced and stable platelet excitability following renal and myocardial reperfusion, respectively. Histological analysis yield evidence of platelet marginalization in the reperfused organs. In conclusion, results from this study do not support a role for platelet activation in early phases of clinical I/R injury.
Collapse
Affiliation(s)
- Kirsten A Kortekaas
- Department of Cardiothoracic Surgery, Leiden University Medical CenterLeiden, The Netherlands
- Department of Cardiology, OLVG OostAmsterdam, The Netherlands
| | - Dorottya K de Vries
- Department of Surgery, Leiden University Medical CenterLeiden, The Netherlands
| | - Mark Roest
- Department of Clinical Chemistry and Hematology, University Medical Center UtrechtUtrecht, The Netherlands
- Current address: Department of Biochemistry, Cardiovascular Research Institute, Maastricht UniversityMaastricht, The Netherlands
| | - Marlies EJ Reinders
- Department of Nephrology, Leiden University Medical CenterLeiden, The Netherlands
| | - Eric P van der Veer
- Department of Nephrology, Leiden University Medical CenterLeiden, The Netherlands
| | - Robert JM Klautz
- Department of Cardiothoracic Surgery, Leiden University Medical CenterLeiden, The Netherlands
| | - Philip G de Groot
- Department of Clinical Chemistry and Hematology, University Medical Center UtrechtUtrecht, The Netherlands
| | | | - Jan H Lindeman
- Department of Surgery, Leiden University Medical CenterLeiden, The Netherlands
| |
Collapse
|
18
|
Zhao H, Alam A, Soo AP, George AJT, Ma D. Ischemia-Reperfusion Injury Reduces Long Term Renal Graft Survival: Mechanism and Beyond. EBioMedicine 2018; 28:31-42. [PMID: 29398595 PMCID: PMC5835570 DOI: 10.1016/j.ebiom.2018.01.025] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 01/18/2018] [Accepted: 01/20/2018] [Indexed: 01/10/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) during renal transplantation often initiates non-specific inflammatory responses that can result in the loss of kidney graft viability. However, the long-term consequence of IRI on renal grafts survival is uncertain. Here we review clinical evidence and laboratory studies, and elucidate the association between early IRI and later graft loss. Our critical analysis of previous publications indicates that early IRI does contribute to later graft loss through reduction of renal functional mass, graft vascular injury, and chronic hypoxia, as well as subsequent fibrosis. IRI is also known to induce kidney allograft dysfunction and acute rejection, reducing graft survival. Therefore, attempts have been made to substitute traditional preserving solutions with novel agents, yielding promising results. Ischaemia reperfusion injury (IRI) potentiates delayed renal graft function and causes reduction in renal graft survival IRI causes innate immune system activation, hypoxic injury, inflammation and graft vascular disease Reducing prolonged cold ischaemic time improves graft survival Novel protective strategies include mesenchymal stem cells, machine perfusion, and ex vivo preservation solution saturated with gas. Further studies are needed to investigate the long-term effects of novel ex vivo preservation agents
Collapse
Affiliation(s)
- Hailin Zhao
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Azeem Alam
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Aurelie Pac Soo
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | | | - Daqing Ma
- Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK.
| |
Collapse
|
19
|
Pan L, Chen J, Xing W, Xing Z, Zhang J, Peng Y, Zhang Z. Magnetic resonance imaging evaluation of renal ischaemia-reperfusion injury in a rabbit model. Exp Physiol 2017; 102:1000-1006. [PMID: 28470945 DOI: 10.1113/ep086203] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/24/2017] [Indexed: 02/06/2023]
Abstract
NEW FINDINGS What is the central question of this study? Renal ischaemia-reperfusion injury occurs in various clinical settings. The clinical diagnosis of ischaemia-reperfusion injury is routinely based on biochemical and haematological tests, which cannot evaluate the function of a single kidney. New magnetic resonance imaging techniques to identify the pathophysiological changes in the renal outer medulla were evaluated. What is the main finding and its importance? This study demonstrated that susceptibility-weighted imaging is a feasible non-invasive tool for imaging and evaluating physipathological changes in the renal outer medulla after ischaemia-reperfusion injury. The aim was to evaluate the feasibility of susceptibility-weighted imaging (SWI) as a tool to identify the changes in the renal outer medulla (OM) in a rabbit model of renal ischaemia-reperfusion injury (IRI). New Zealand rabbits were used (control group n = 10; IRI group n = 40). The rabbits in the IRI group were subjected to left renal artery clamping for 60 min. T2-weighted (T2WI) and SWI examinations were performed at 1, 12, 24 or 48 h after reperfusion (each n = 10). After the examinations, the kidneys were submitted to histological evaluation. The contrast-to-noise ratio (CNR) for the left renal OM was measured using T2WI and SWI. The T2WI and SWI scores of the integrity of the renal OM were evaluated. There were significant differences between T2WI CNRs and SWI CNRs in the control group and the IRI 1, 12 and 48 h time points (all P < 0.05). No significant difference was found between T2WI and SWI CNRs at IRI 24 h (P > 0.05). The mean SWI scores of renal OM in the IRI 1 and 12 h subgroups were both significantly lower than that in the control group (all P < 0.05). The only significant difference in the mean T2WI scores of renal OM was observed between the control and IRI 1 h groups (P < 0.05). Susceptibility-weighted imaging has a significant advantage in evaluation of healthy renal OM over conventional magnetic resonance imaging, and it is a feasible non-invasive tool for imaging and evaluating changes in the renal OM after IRI.
Collapse
Affiliation(s)
- Liang Pan
- Department of Radiology, the Third Affiliated Hospital of Suzhou University, Changzhou, People's Republic of China
| | - Jie Chen
- Department of Radiology, the Third Affiliated Hospital of Suzhou University, Changzhou, People's Republic of China.,Department of Medical Imaging, Jinling Hospital, Nanjing University School of Medicine, Nanjing, People's Republic of China
| | - Wei Xing
- Department of Radiology, the Third Affiliated Hospital of Suzhou University, Changzhou, People's Republic of China
| | - Zhaoyu Xing
- Department of Urology, the Third Affiliated Hospital of Suzhou University, Changzhou, People's Republic of China
| | - Jinggang Zhang
- Department of Radiology, the Third Affiliated Hospital of Suzhou University, Changzhou, People's Republic of China
| | - Yan Peng
- Department of Pathology, the Third Affiliated Hospital of Suzhou University, Changzhou, People's Republic of China
| | - Zhuoli Zhang
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
20
|
Abstract
Zero-time kidney biopsies, obtained at time of transplantation, are performed in many transplant centers worldwide. Decisions on kidney discard, kidney allocation, and choice of peritransplant and posttransplant treatment are sometimes based on the histological information obtained from these biopsies. This comprehensive review evaluates the practical considerations of performing zero-time biopsies, the predictive performance of zero-time histology and composite histological scores, and the clinical utility of these biopsies. The predictive performance of individual histological lesions and of composite scores for posttransplant outcome is at best moderate. No single histological lesion or composite score is sufficiently robust to be included in algorithms for kidney discard. Dual kidney transplantation has been based on histological assessment of zero-time biopsies and improves outcome in individual patients, but the waitlist effects of this strategy remain obscure. Zero-time biopsies are valuable for clinical and translational research purposes, providing insight in risk factors for posttransplant events, and as baseline for comparison with posttransplant histology. The molecular phenotype of zero-time biopsies yields novel therapeutic targets for improvement of donor selection, peritransplant management and kidney preservation. It remains however highly unclear whether the molecular expression variation in zero-time biopsies could become a better predictor for posttransplant outcome than donor/recipient baseline demographic factors.
Collapse
|
21
|
Vrakas G, Tsalis K, Roidos GN, Christoforidis E, Kouzi-Koliakou K, Lazaridis C, Vaidya A. Synergistic Effect of Ischemic Preconditioning and Antithrombin in Ischemia-Reperfusion Injury. EXP CLIN TRANSPLANT 2017; 15:320-328. [PMID: 28418287 DOI: 10.6002/ect.2015.0331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Our study aimed to determine whether antithrombin plays a synergistic role in accentuating the effects of intestinal ischemic preconditioning. MATERIALS AND METHODS Fifty rats were randomly allocated to 5 groups (10 rats/group) as follows: sham treatment (group 1); ischemia-reperfusion (group 2); ischemic preconditioning followed by ischemia-reperfusion (group 3); antithrombin + ischemia-reperfusion, similar to group 2 but including antithrombin administration (group 4); and antithrombin + ischemic preconditioning, similar to group 3 but including antithrombin administration (group 5). Blood samples and liver specimens were obtained for measurement of cytokines, myeloperoxidase, and malondialdehyde. Liver biopsies were examined by electron microscopy. RESULTS Intestinal ischemia-reperfusion induced a remote hepatic inflammatory response as evidenced by the striking increase of proinflammatory cytokines, myeloperoxidase, and malondialdehyde. Tumor necrosis factor-α levels in group 5 (12.48 ± 0.7 pg/mL) were significantly lower than in group 3 (13.64 ± 0.78 pg/mL; P = .014). Mean interleukin 1β was lower in group 5 (9.52 ± 0.67pg/mL) than in group 3 (11.05 ± 1.9 pg/mL; P > .99). Mean interleukin 6 was also significantly lower in group 5 (17.13 ± 0.54 pg/mL) than in group 3 (23.82 ± 1 pg/mL; P ≤ .001). Myeloperoxidase levels were significantly higher in group 3 (20.52 ± 2.26 U/g) than in group 5 (18.59 ± 1.03 U/g; P = .025). However, malondialdehyde levels did not significantly improve in group 5 (4.55 ± 0.46 μmol) versus group 3 (5.17 ± 0.61 μmol; P = .286). Tumor necrosis factor-α, interleukin 6, and myeloperoxidase findings show that antithrombin administration further attenuated the inflammatory response caused by ischemia-reperfusion, suggesting a synergistic effect with ischemic preconditioning. These findings were confirmed by electron microscopy. CONCLUSIONS The addition of antithrombin to ischemic preconditioning may act to attenuate or prevent damage from ischemia-reperfusion injury by inhibiting the release of cytokines and neutrophil infiltration.
Collapse
Affiliation(s)
- Georgios Vrakas
- From the the Fourth Surgical Department, Aristotle University of Thessaloniki, 57010 Thessaloniki, Greece; and the Oxford Transplant Centre, Oxford OX3 7LE, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
22
|
Ginkgo biloba extract EGb761 attenuates brain death-induced renal injury by inhibiting pro-inflammatory cytokines and the SAPK and JAK-STAT signalings. Sci Rep 2017; 7:45192. [PMID: 28332628 PMCID: PMC5362910 DOI: 10.1038/srep45192] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 02/20/2017] [Indexed: 12/16/2022] Open
Abstract
This study aimed to investigate the protective effects of EGb761, a Ginkgo Biloba extract, against brain death-induced kidney injury. Sixty male Sprague Dawley rats were randomly divided into six groups: sham, brain-death (BD), BD + EGb b48h (48 hours before BD), BD + EGb 2 h (2 hours after BD), BD + EGb 1 h, and BD + EGb 0.5 h. Six hours after BD, serum sample and kidney tissues were collected for analyses. The levels of blood urea nitrogen (BUN) and serum creatinine significantly elevated in the BD group than in sham group. In all the EGb761-treated BD animals except for the BD + Gb 2 h group, the levels of BUN and serum creatinine significantly reduced (all P < 0.01). EGb761 attenuated tubular injury and lowered the histological score. In addition, the longer duration of drug treatment was, the better protective efficacy could be observed. EGb761 significantly reduced IL-1β, IL-6, TNF-α, MCP-1, IP-10 mRNA expression and macrophage infiltration in the kidney. EGb761 treatment at 48 hour before brain death significantly attenuate the levels of p-JNK-MAPK, p-p38-MAPK, and p-STAT3 proteins (all P < 0.05, compared to BD group). In summary, our data showed that EGb761 treatment protected donor kidney from BD-induced damages by blocking SAPK and JAK-STAT signalings. Early administration of EGb761 can provide better protective efficacy.
Collapse
|
23
|
Wang L, Song J, Buggs J, Wei J, Wang S, Zhang J, Zhang G, Lu Y, Yip KP, Liu R. A new mouse model of hemorrhagic shock-induced acute kidney injury. Am J Physiol Renal Physiol 2016; 312:F134-F142. [PMID: 28042109 DOI: 10.1152/ajprenal.00347.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 10/25/2016] [Accepted: 10/31/2016] [Indexed: 11/22/2022] Open
Abstract
Current animal models of hemorrhagic shock-induced acute kidney injury (HS-induced AKI) require extensive surgical procedures and constant monitoring of hemodynamic parameters. Application of these HS-induced AKI models in mice to produce consistent kidney injury is challenging. In the present study, we developed a simple and highly reproducible mouse model of HS-induced AKI by combining moderate bleeding and renal pedicle clamping, which was abbreviated as HS-AKI. HS was induced by retroorbital bleeding of 0.4 ml blood in C57BL/6 mice. Mice were left in HS stage for 30 min, followed by renal pedicle clamping for 18 min at 36.8-37.0°C. Mean arterial pressure (MAP) and heart rate were monitored with preimplanted radio transmitters throughout the experiment. The acute response in renal blood flow (RBF) triggered by HS was measured with transonic flow probe. Mice received sham operation; bleeding alone and renal pedicle clamping alone served as respective controls. MAP was reduced from 77 ± 4 to 35 ± 3 mmHg after bleeding. RBF was reduced by 65% in the HS period. Plasma creatinine and kidney injury molecule-1 levels were increased by more than 22-fold 24 h after reperfusion. GFR was declined by 78% of baseline 3 days after reperfusion. Histological examination revealed a moderate-to-severe acute tubular damage, mostly at the cortex-medulla junction area, followed by the medullar and cortex regions. HS alone did not induce significant kidney injury, but synergistically enhanced pedicle clamping-induced AKI. This is a well-controlled, simple, and reliable mouse model of HS-AKI.
Collapse
Affiliation(s)
- Lei Wang
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida;
| | - Jiangping Song
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida
| | | | - Jin Wei
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida
| | - Shaohui Wang
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida
| | - Jie Zhang
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida
| | - Gensheng Zhang
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida.,Department of Physiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan Lu
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Kay-Pong Yip
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida
| | - Ruisheng Liu
- Department of Molecular Pharmacology & Physiology, University of South Florida College of Medicine, Tampa, Florida
| |
Collapse
|
24
|
Turunen AJ, Lindgren L, Salmela KT, Kyllönen LE, Andersson S, Pesonen E. Matrix Metalloproteinase-9 and Graft Preservation Injury in Clinical Renal Transplantation. Transplant Proc 2016; 47:2831-5. [PMID: 26707297 DOI: 10.1016/j.transproceed.2015.10.056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 09/18/2015] [Accepted: 10/28/2015] [Indexed: 12/01/2022]
Abstract
BACKGROUND Deleterious effects of matrix metalloproteinase-9 (MMP-9) have been established in experimental renal ischemia-reperfusion models but not in clinical renal transplantation thus far. METHODS We studied MMP-9 and its physiological inhibitor tissue inhibitor of matrix metalloproteinases-1 (TIMP-1) in 45 consecutive patients of a larger trial in renal transplantation: perioperative anti-thymocyte globulin (group A, n = 15), perioperative basiliximab (group B, n = 16), and conventional triple therapy (group C, n = 14). In addition to systemic blood samples, local blood samples were obtained simultaneously at 1 and 5 minutes after reperfusion from iliac artery and graft vein for calculation of transrenal changes. Because anti-thymocyte globulin activates inflammation, group A was analyzed separately. Groups B and C were pooled (group BC). RESULTS Anti-thymocyte globulin infusion caused a robust rise of MMP-9 in the systemic circulation in group A. No significant transrenal difference of MMP-9 or TIMP-1 occurred in either group during graft reperfusion. In group BC, strong transrenal release of MMP-9 at 1 minute after reperfusion correlated with cold ischemia time (R = 0.66, P = .0001) and was associated with delayed graft function (P = .052). CONCLUSIONS Renal production of MMP-9 on graft reperfusion is associated with cold ischemia time and emergence of delayed graft function. MMP inhibition may offer a means to reduce reperfusion injury in renal transplantation.
Collapse
Affiliation(s)
- A J Turunen
- Department of Surgery, Kanta-Häme Central Hospital, Hämeenlinna, Finland
| | - L Lindgren
- Department of Anesthesiology, Tampere University Hospital, Tampere, Finland
| | - K T Salmela
- Department of Transplantation and Liver Surgery, Helsinki University Hospital, Helsinki, Finland
| | - L E Kyllönen
- Department of Transplantation and Liver Surgery, Helsinki University Hospital, Helsinki, Finland
| | - S Andersson
- Children's Hospital, Helsinki University Hospital, Helsinki, Finland
| | - E Pesonen
- Department of Anesthesiology and Intensive Care Medicine, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
25
|
|
26
|
Wiebe C, Gibson IW, Blydt-Hansen TD, Pochinco D, Birk PE, Ho J, Karpinski M, Goldberg A, Storsley L, Rush DN, Nickerson PW. Rates and determinants of progression to graft failure in kidney allograft recipients with de novo donor-specific antibody. Am J Transplant 2015; 15:2921-30. [PMID: 26096305 DOI: 10.1111/ajt.13347] [Citation(s) in RCA: 277] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 03/13/2015] [Accepted: 04/05/2015] [Indexed: 01/25/2023]
Abstract
Understanding rates and determinants of clinical pathologic progression for recipients with de novo donor-specific antibody (dnDSA), especially subclinical dnDSA, may identify surrogate endpoints and inform clinical trial design. A consecutive cohort of 508 renal transplant recipients (n = 64 with dnDSA) was studied. Recipients (n = 388) without dnDSA or dysfunction had an eGFR decline of -0.65 mL/min/1.73 m(2) /year. In recipients with dnDSA, the rate eGFR decline was significantly increased prior to dnDSA onset (-2.89 vs. -0.65 mL/min/1.73 m(2) /year, p < 0.0001) and accelerated post-dnDSA (-3.63 vs. -2.89 mL/min/1.73 m(2) /year, p < 0.0001), suggesting that dnDSA is both a marker and contributor to ongoing alloimmunity. Time to 50% post-dnDSA graft loss was longer in recipients with subclinical versus a clinical dnDSA phenotype (8.3 vs. 3.3 years, p < 0.0001). Analysis of 1091 allograft biopsies found that dnDSA and time independently predicted chronic glomerulopathy (cg), but not interstitial fibrosis and tubular atrophy (IFTA). Early T cell-mediated rejection, nonadherence, and time were multivariate predictors of IFTA. Independent risk factors for post-dnDSA graft survival available prior to, or at the time of, dnDSA detection were delayed graft function, nonadherence, dnDSA mean fluorescence intensity sum score, tubulitis, and cg. Ultimately, dnDSA is part of a continuum of mixed alloimmune-mediated injury, which requires solutions targeting T and B cells.
Collapse
Affiliation(s)
- C Wiebe
- Department of Medicine, University of Manitoba, Diagnostic Services of Manitoba, Winnipeg, Manitoba, Canada
| | - I W Gibson
- Department of Pathology, University of Manitoba, Diagnostic Services of Manitoba, Winnipeg, Manitoba, Canada
| | - T D Blydt-Hansen
- Department of Pediatrics and Child Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - D Pochinco
- Diagnostic Services of Manitoba, Winnipeg, Manitoba, Canada
| | - P E Birk
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - J Ho
- Department of Medicine and Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - M Karpinski
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - A Goldberg
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - L Storsley
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - D N Rush
- Department of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - P W Nickerson
- Departments of Medicine and Immunology, University of Manitoba, Diagnostic Services of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
27
|
Fonseca I, Oliveira JC, Santos J, Malheiro J, Martins LS, Almeida M, Dias L, Pedroso S, Lobato L, Henriques AC, Mendonça D. Leptin and adiponectin during the first week after kidney transplantation: biomarkers of graft dysfunction? Metabolism 2015; 64:202-7. [PMID: 25458832 DOI: 10.1016/j.metabol.2014.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 10/03/2014] [Accepted: 10/03/2014] [Indexed: 11/29/2022]
Abstract
CONTEXT AND OBJECTIVE Based on evidence that leptin and adiponectin are removed from circulation primarily by the kidney, we designed a study to examine the longitudinal changes of these adipokines during the first week after kidney transplantation (KTx) and to test the hypothesis that higher levels of leptin and/or adiponectin could be early biomarkers of delayed graft function (DGF=dialysis requirement during the first post-transplant week) and acute rejection. STUDY DESIGN Repeated-measures prospective study. MATERIAL AND METHODS Forty consecutive adult patients with end-stage renal disease who were undergoing KTx. Leptin and adiponectin were measured in blood samples that were collected before (day-0) and after KTx (days-1, 2, 4 and 7). Linear mixed-models, receiver operating characteristic and area under curve (AUC-ROC) were used. RESULTS At post-transplant day-1, leptinemia and adiponectinemia declined 43% and 47%, respectively. At all times studied after KTx, the median leptin levels were significantly higher in patients developing DGF (n=18), but not adiponectin levels. Shortly after KTx (day-1), leptin values were significantly higher in DGF recipients in contrast to patients with promptly functioning kidneys, approximately two times higher when controlling for gender and BMI. The leptin reduction rate between pre-tranplant and one-day after KTx moderately predicted DGF (AUC=0.73). On day-1, serum leptin predicted DGF (AUC-ROC=0.76) with a performance slightly better than serum creatinine (AUC-ROC=0.72), even after correcting for BMI (AUC-ROC=0.73). Separating this analysis by gender showed that the performance of leptin in predicting DGF for male gender (AUC-ROC=0.86) improved. CONCLUSIONS Kidney graft function is an independent determinant of leptin levels, but not of adiponectin. Leptin levels at day-1 slightly outperformed serum creatinine in predicting the occurrence of DGF, and more accurately in male gender. No significant association was detected with acute rejection.
Collapse
Affiliation(s)
- Isabel Fonseca
- Department of Nephrology and Kidney Transplantation, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal; Unit for Multidisciplinary Investigation in Biomedicine (UMIB), Porto, Portugal; Institute of Public Health (ISPUP), University of Porto, Porto, Portugal.
| | - José Carlos Oliveira
- Department of Clinical Chemistry, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal
| | - Josefina Santos
- Department of Nephrology and Kidney Transplantation, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal; Unit for Multidisciplinary Investigation in Biomedicine (UMIB), Porto, Portugal
| | - Jorge Malheiro
- Department of Nephrology and Kidney Transplantation, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal; Unit for Multidisciplinary Investigation in Biomedicine (UMIB), Porto, Portugal
| | - La Salete Martins
- Department of Nephrology and Kidney Transplantation, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal; Unit for Multidisciplinary Investigation in Biomedicine (UMIB), Porto, Portugal
| | - Manuela Almeida
- Department of Nephrology and Kidney Transplantation, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal; Unit for Multidisciplinary Investigation in Biomedicine (UMIB), Porto, Portugal
| | - Leonídio Dias
- Department of Nephrology and Kidney Transplantation, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal
| | - Sofia Pedroso
- Department of Nephrology and Kidney Transplantation, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal; Unit for Multidisciplinary Investigation in Biomedicine (UMIB), Porto, Portugal
| | - Luísa Lobato
- Department of Nephrology and Kidney Transplantation, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal; Unit for Multidisciplinary Investigation in Biomedicine (UMIB), Porto, Portugal
| | - António Castro Henriques
- Department of Nephrology and Kidney Transplantation, Centro Hospitalar do Porto, Hospital de Santo António, Porto, Portugal; Unit for Multidisciplinary Investigation in Biomedicine (UMIB), Porto, Portugal
| | - Denisa Mendonça
- Department of Population Studies, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Institute of Public Health (ISPUP), University of Porto, Porto, Portugal
| |
Collapse
|
28
|
Modery-Pawlowski CL, Kuo HH, Baldwin WM, Sen Gupta A. A platelet-inspired paradigm for nanomedicine targeted to multiple diseases. Nanomedicine (Lond) 2014; 8:1709-27. [PMID: 24074391 DOI: 10.2217/nnm.13.113] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Platelets are megakaryocyte-derived anucleated cells found in the blood. They are mainly responsible for rendering hemostasis or clotting to prevent bleeding complications. Decreased platelet numbers or deficiencies in platelet functions can lead to various acute or chronic bleeding conditions and hemorrhage. On the other hand, dysregulated hyperactivity of the clotting process can lead to thrombosis and vascular occlusion. There is significant evidence that beyond hemostasis and thrombosis, platelets play crucial mechanistic roles in other disease scenarios such as inflammation, immune response and cancer metastasis by mediating several cell-cell and cell-matrix interactions, as well as aiding the disease microenvironment via secretion of multiple soluble factors. Therefore, elucidating these mechanistic functions of platelets can provide unique avenues for developing platelet-inspired nanomedicine strategies targeted to these diseases. To this end, the current review provides detailed mechanistic insight into platelets' disease-relevant functions and discusses how these mechanisms can be utilized to engineer targeted nanomedicine systems.
Collapse
Affiliation(s)
- Christa L Modery-Pawlowski
- Department of Biomedical Engineering, Case Western Reserve University, 2071 Martin Luther King Jr Drive, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
29
|
Kapitsinou PP, Sano H, Michael M, Kobayashi H, Davidoff O, Bian A, Yao B, Zhang MZ, Harris RC, Duffy KJ, Erickson-Miller CL, Sutton TA, Haase VH. Endothelial HIF-2 mediates protection and recovery from ischemic kidney injury. J Clin Invest 2014; 124:2396-409. [PMID: 24789906 DOI: 10.1172/jci69073] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The hypoxia-inducible transcription factors HIF-1 and HIF-2 mediate key cellular adaptions to hypoxia and contribute to renal homeostasis and pathophysiology; however, little is known about the cell type-specific functions of HIF-1 and HIF-2 in response to ischemic kidney injury. Here, we used a genetic approach to specifically dissect the roles of endothelial HIF-1 and HIF-2 in murine models of hypoxic kidney injury induced by ischemia reperfusion or ureteral obstruction. In both models, inactivation of endothelial HIF increased injury-associated renal inflammation and fibrosis. Specifically, inactivation of endothelial HIF-2α, but not endothelial HIF-1α, resulted in increased expression of renal injury markers and inflammatory cell infiltration in the postischemic kidney, which was reversed by blockade of vascular cell adhesion molecule-1 (VCAM1) and very late antigen-4 (VLA4) using monoclonal antibodies. In contrast, pharmacologic or genetic activation of HIF via HIF prolyl-hydroxylase inhibition protected wild-type animals from ischemic kidney injury and inflammation; however, these same protective effects were not observed in HIF prolyl-hydroxylase inhibitor-treated animals lacking endothelial HIF-2. Taken together, our data indicate that endothelial HIF-2 protects from hypoxia-induced renal damage and represents a potential therapeutic target for renoprotection and prevention of fibrosis following acute ischemic injury.
Collapse
|
30
|
Mallet V, Dutheil D, Polard V, Rousselot M, Leize E, Hauet T, Goujon JM, Zal F. Dose-Ranging Study of the Performance of the Natural Oxygen Transporter HEMO2Life in Organ Preservation. Artif Organs 2014; 38:691-701. [DOI: 10.1111/aor.12307] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Vanessa Mallet
- Institut National de la Santé et de la Recherche Médicale U1082; Faculté de Médecine et Pharmacie; Université de Poitiers; Morlaix France
- Hemarina SA; Morlaix France
| | | | | | | | - Elisabeth Leize
- Département de Prothèses; Unité de Formation et de Recherche d'Odontologie; Centre Hospitalier Universitaire de Brest; Brest France
| | - Thierry Hauet
- Institut National de la Santé et de la Recherche Médicale U1082; Faculté de Médecine et Pharmacie; Université de Poitiers; Morlaix France
- Département de Biochimie; Centre Hospitalier Universitaire de Poitiers; Morlaix France
- Fédération pour l'Étude de l'Ischemie Reperfusion en Transplantation; Morlaix France
- Plate-forme IBiSA (Infrastructures en Biologie, Sante et Agronomie); Unité de Transplantation Expérimentale; Génétique Expérimentale en Productions Animales; Département de Génétique Animale; Domaine du Magneraud; Institut National de Recherche Agronomique; Surgères France
| | - Jean Michel Goujon
- Institut National de la Santé et de la Recherche Médicale U1082; Faculté de Médecine et Pharmacie; Université de Poitiers; Morlaix France
| | | |
Collapse
|
31
|
Camarço JPF, Morais LK, Moreno JD, Rassi MC, Camarço MNDCR, Guimarães PF, Quireze Júnior C. The feasibility of an experimental model of donors after cardiac death in remote ischemic preconditioning studies for renal transplantation in porcine. Acta Cir Bras 2014; 29:82-6. [PMID: 24604310 DOI: 10.1590/s0102-86502014000200002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/21/2014] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To evaluate the feasibility of an experimental model of donors after cardiac death in remote ischemic preconditioning studies. METHODS Twelve Landrace pigs were used as organ donors. They underwent cardiac arrest by coronary en block suture and interruption of ventilatory support. Haemodynamic data regarding the donor surgical protocol were evaluated. Studies variables included mean heart rate, systolic blood pressure, diastolic blood pressure, mean arterial pressure, central venous pressure and oxygen saturation and the time to death. RESULTS Haemodynamic parameter indicated that the circulatory failure occurred after nine minutes of en block coronary suture and respiratory support interruption. The circulatory collapse occurred evenly across all groups. The heart rate and central venous pressure were statistically different between groups (p=0.023 and p=0.04), respectively. The remote preconditioning resulted in delayed time of death. CONCLUSIONS The model is feasible, and was easily reproduced. The ischemic remote preconditioning tends to a slight increase in circulatory failure time.
Collapse
Affiliation(s)
- João Paulo Figueiredo Camarço
- Federal University of Goias, GoianiaGO, Brazil, Fellow Master degree, Postgraduate Program in Health Sciences, Federal University of Goias (UFG), Goiania-GO, Brazil. Intellectual and scientific contents of the study, technical procedures, manuscript writing
| | - Lucio Kenny Morais
- UFG, GoianiaGO, Brazil, Fellow Master degree, Postgraduate Program in Health Sciences, UFG, Goiania-GO, Brazil. Acquisition and interpretation of data, manuscript writing
| | - Juan Duque Moreno
- UFG, School of Veterinary and Animal Science, GoianiaGO, Brazil, PhD, Associate Professor, Anesthesiology, School of Veterinary and Animal Science (EVZ), UFG, Goiania-GO, Brazil. Intellectual and scientific content of the study, technical procedures, manuscript writing
| | - Matheus Castrillon Rassi
- UFG, GoianiaGO, Brazil, Fellow Master degree, Postgraduate Program in Health Sciences, UFG, Goiania-GO, Brazil. Acquisition and interpretation of data, manuscript writing
| | - Mylena Naves de Castro Rocha Camarço
- Pontifical Catholic University of Goias, Department of Obstetrics/Gynecology, GoianiaGO, Brazil, Assistant Professor, Department of Obstetrics/Gynecology, Pontifical Catholic University of Goias (UCG), Goiania-GO, Brazil. Critical revision
| | - Pedro Figueiredo Guimarães
- Catholic University of Brasilia, Departament of Surgery, BrasiliaDF, Graduate student, Departament of Surgery, Catholic University of Brasilia (UCB), Brasilia-DF. Acquisition of data, technical procedures
| | - Claudemiro Quireze Júnior
- UFG, GoianiaGO, Brazil, PhD, Associate Professor, Surgical Clinics, UFG, Goiania-GO, Brazil. Intellectual and scientific contents of the study, technical procedures, manuscript writing
| |
Collapse
|
32
|
Su MW, Chang SS, Chen CH, Huang CC, Chang SW, Tsai YC, Lam CF. Preconditioning renoprotective effect of isoflurane in a rat model of virtual renal transplant. J Surg Res 2014; 189:135-42. [PMID: 24674838 DOI: 10.1016/j.jss.2014.02.035] [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: 11/13/2013] [Revised: 02/18/2014] [Accepted: 02/20/2014] [Indexed: 11/25/2022]
Abstract
BACKGROUND The development of warm-cold ischemia-reperfusion (IR) injury of the kidney grafts is inevitable during renal transplantation. However, there is currently no definite renoprotective strategy available in the protection of the graft tissue. In the present study, we compared the renal protection of preconditioning isoflurane with N-acetylcysteine (NAC) in a novel rat model of warm-cold renal IR injury. MATERIALS AND METHODS Adult Sprague-Dawley rats were randomly assigned to receive inhaled isoflurane (1.5% for 2 h), NAC (1 g/kg, intra-arterial injection) or placebo before the induction of brief warm ischemia (10 min) followed by cold ischemia (45 min) periods. Plasma levels of creatinine and tissue inflammatory reaction in the kidney were analyzed 72 h after reperfusion. RESULTS Elevated plasma level of creatinine and urea indicated the development of acute renal injury secondary to IR injury. The creatinine levels were reduced in animals pretreated with inhaled isoflurane and NAC, and the level was more significantly decreased in the isoflurane-treated group. Preconditioning with volatile isoflurane also significantly suppressed the tissue myeloperoxidase activity and expression of the inducible nitric oxide synthase. Immunostaining confirmed that myeloperoxidase expression was most significantly attenuated in the glomerulus and peritubular capillaries of rats pre-exposed to isoflurane. CONCLUSIONS We present the first study demonstrating that the administration of volatile isoflurane before induction of experimental warm-cold renal IR injury provides preconditioning renoprotective effect, which is superior to the treatment with NAC. The beneficial renoprotective effect of isoflurane is most likely mediated by attenuation of proinflammatory reaction in the injured kidney.
Collapse
Affiliation(s)
- Min-Wen Su
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Shen-Shin Chang
- Department of Surgery, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Chung-Hao Chen
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Chien-Chi Huang
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Shih-Wei Chang
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Yu-Chuan Tsai
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan
| | - Chen-Fuh Lam
- Department of Anesthesiology, National Cheng Kung University College of Medicine and Hospital, Tainan, Taiwan; Department of Anesthesiology, Buddhist Tzu-Chi General Hospital and Tzu-Chi University School of Medicine, Hualien, Taiwan.
| |
Collapse
|
33
|
Effect of a novel nuclear factor-κB activation inhibitor on renal ischemia-reperfusion injury. Transplantation 2014; 96:863-70. [PMID: 23958925 DOI: 10.1097/tp.0b013e3182a3df74] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND In kidney transplantation, the relationship between prolonged warm or cold ischemic storage of kidneys and a higher incidence of delayed graft function is previously known, and delayed graft function has been known to aggravate poor long-term graft survival. We investigated the effect of a novel nuclear factor-κB activation inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ), on renal ischemia-reperfusion (I/R) injury. METHODS DHMEQ was administered to Lewis rats once just before renal artery clamping (DHMEQ pretreatment group), and the effect on I/R injury was investigated. RESULTS In the DHMEQ pretreatment group, the 24-hr urine volume on days 1 to 3 after I/R was significantly larger, and the protein concentration of the urine on days 2 to 7 was significantly smaller than in the untreated group. The serum creatinine level was significantly improved, and significantly lower levels of the inflammatory cells and inflammatory cytokines were present in the kidneys on day 1. The relative ratio of nuclear to cytoplasmic nuclear factor-κB and oxidative stress of kidney tissue on day 1 were significantly decreased. CONCLUSIONS Treatment with DHMEQ before renal artery clamping may therefore be useful for renal I/R injury and application to renal transplantation is expected.
Collapse
|
34
|
Ojeda-Cervantes M, Barrera-Chimal J, Alberú J, Pérez-Villalva R, Morales-Buenrostro LE, Bobadilla NA. Mineralocorticoid receptor blockade reduced oxidative stress in renal transplant recipients: a double-blind, randomized pilot study. Am J Nephrol 2013; 37:481-90. [PMID: 23635604 DOI: 10.1159/000350539] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 03/06/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Previous experimental studies from our laboratory have demonstrated that aldosterone plays a central role in renal ischemic processes. This study was designed to evaluate the effect of mineralocorticoid receptor blockade in renal transplant recipients from living donors. METHODS 20 adult kidney transplant recipients from living donors were included in a double-blind, randomized, placebo-controlled clinical pilot study that compared spironolactone and placebo. Placebo or spironolactone (25 mg) was administered 1 day before and 3 days posttransplantation. Renal function and urinary kidney injury molecule-1, interleukin-18, and heat shock protein 72 as well as urinary hydrogen peroxide (H2O2) levels were quantified. RESULTS No significant differences were seen between the groups studied regarding age, gender, indication for kidney transplantation, residual renal function, renal replacement therapy, or warm and cold ischemia periods. In contrast, spironolactone administration significantly reduced the oxidative stress assessed by the urinary H2O2 excretion, in spite of no differences in renal function or reduction in tubular injury biomarkers. CONCLUSIONS The findings of this exploratory study strongly suggest that aldosterone promotes oxidative stress and that the administration of spironolactone reduces the production of urinary H2O2 as a result of lesser formation of surrogate reactive oxygen species secondary to the ischemia-reperfusion phenomenon.
Collapse
Affiliation(s)
- Marcos Ojeda-Cervantes
- Nephrology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | | | | | | | | |
Collapse
|
35
|
Kohei N, Tanabe T, Horita S, Omoto K, Ishida H, Yamaguchi Y, Tanabe K. Sequential analysis of donor-specific antibodies and pathological findings in acute antibody-mediated rejection in a rat renal transplantation model. Kidney Int 2013; 84:722-32. [PMID: 23615506 DOI: 10.1038/ki.2013.117] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 01/25/2013] [Accepted: 01/31/2013] [Indexed: 11/09/2022]
Abstract
Alloantibodies contribute significantly to renal transplant rejection by activation of complement and various cytokines with a variety of effector cells, and are a major cause of allograft loss. Although there is clinical evidence of antibody- and complement-mediated injury in renal transplantation, the mechanism of antibody-mediated rejection remains largely unknown. In order to understand the sequential production of antibodies and complement components, we presensitized recipient rats by skin transplantation. Anti-donor-specific IgG levels reached a maximum 2 weeks following presensitization after which the rats underwent renal transplantation from the same donor strain. We then evaluated sequential pathological findings based on the Banff classification and several factors related to graft rejection. In this presensitized model, peritubular capillaries were already dilated and stained for C4d. Neutrophil and mononuclear cell infiltration in these capillaries was detected beginning 2 h after transplantation. Donor-specific antibody IgG levels decreased rapidly and anti-IgG antibody stained glomerular and peritubular capillaries in the grafts beginning 2 h after transplantation. Additionally, several cytokines and complement components showed marked changes in the presensitized group. Thus, in the donor-specific presensitized recipient, alloantibodies and complement were activated immediately after transplant. C4d deposition in peritubular capillaries appears to be a key factor for the diagnosis of antibody-associated rejection.
Collapse
Affiliation(s)
- Naoki Kohei
- Department of Urology, Tokyo Women's Medical University, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Hirt-Minkowski P, Amico P, Hönger G, Praehauser C, Steiger J, Koller MT, Gürke L, Mayr M, Schaub S. Delayed graft function is not associated with an increased incidence of renal allograft rejection. Clin Transplant 2012; 26:E624-33. [PMID: 23106785 DOI: 10.1111/ctr.12041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2012] [Indexed: 12/19/2022]
Abstract
Delayed graft function (DGF) is considered as a risk factor for renal allograft rejection, but this association might be confounded by diagnostic biases (e.g., higher biopsy frequency in patients with DGF, inclusion of clinically diagnosed rejection episodes, and limited details on the rejection phenotype). This retrospective study including 329 deceased donor transplantations aimed to clarify a causal relationship between DGF and rejection. DGF occurred in 93/329 recipients (28%), whereas immediate graft function (IGF) in 236/329 recipients (72%). The percentage of patients with ≥1 allograft biopsy within the first year post-transplant was similar between the DGF and IGF group (96% vs. 94%; p=0.60). The cumulative one-yr incidence of biopsy-proven clinical (35% vs. 34%; p=0.62) and combined (sub)clinical rejection (58% vs. 60%; p=0.79) was not different between the two groups. Furthermore, there were no differences regarding rejection phenotypes/severities and time frame of occurrence. By multivariable Cox regression analysis, donor-specific HLA antibodies, younger recipient age, and immunosuppressive regimens were independent predictors for clinical rejection, while DGF was not. These results in an intermediate sized, but thoroughly investigated patient population challenge the concept that DGF is a risk factor for rejection and highlights the need for additional studies in this regard.
Collapse
Affiliation(s)
- Patricia Hirt-Minkowski
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Therapeutic role of toll-like receptor modification in cardiovascular dysfunction. Vascul Pharmacol 2012; 58:231-9. [PMID: 23070056 DOI: 10.1016/j.vph.2012.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 09/28/2012] [Accepted: 10/05/2012] [Indexed: 01/04/2023]
Abstract
Toll-like receptors (TLR) are key pattern recognition receptors in the innate immune system. The TLR-mediated immune response against pathogens is usually protective however inappropriate TLR activation may lead to excessive tissue damage. It is well recognised that TLRs respond to a variety of endogenous as well as exogenous ligands. By responding to endogenous ligands that are exposed during cellular damage, TLRs have been implicated in a range of pathological conditions associated with cardiovascular dysfunction. Increasing knowledge on the mechanisms involved in TLR signalling has encouraged the exploration of therapeutic pharmacological modulation of TLR activation in conditions such as atherosclerosis, ischaemic heart disease, heart failure and ischaemic reperfusion injury. The aim of this review is to explore the translational potentials of TLR modification in cardiovascular dysfunction, where these agents have been studied.
Collapse
|
38
|
Ait-Hsiko L, Kraaij T, Wedel J, Theisinger B, Theisinger S, Yard B, Bugert P, Schedel A. N-octanoyl-dopamine is a potent inhibitor of platelet function. Platelets 2012; 24:428-34. [PMID: 22916829 DOI: 10.3109/09537104.2012.715217] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Dopamine (DA) is a co-agonist for platelet activation; yet, donor DA treatment is associated with improved transplantation outcome in renal and heart recipients. Recently, N-octanoyl-dopamine (NOD) was developed which displays superior effects compared to DA in terms of graft protecting properties. Whereas DA is a known platelet co-agonist, the effect of NOD on platelet function is unknown. This is a hypothesis generating study with the aim to assess the effects and molecular mechanisms of NOD and NOD-like compounds on platelet function. The influence of DA, NOD, and NOD-like compounds on platelet responses to classical agonists (adenosine 5'-diphosphate (ADP), U46619) was investigated in six healthy donors by applying whole blood aggregometry (Multiplate®) and flow cytometry for Pac-1, CD62P, and CD63 expression. Changes in platelet cAMP concentrations were assessed by ELISA. While DA showed synergy in platelet activation by ADP and U46619, NOD caused significant inhibition of platelet function both in whole blood aggregometry and flow cytometry. The inhibitory effect of NOD was not mediated via cAMP levels. The nonredox-active NOD-analog N-octanoyl-tyramine had no effects on platelet function. Acetylated NOD conferred to NOD by intracellular esterases showed similar inhibitory effects as NOD. In contrast to DA, NOD is a potent inhibitor of platelet function most likely through intracellular redox-active processes. This adds to the overall protective effect of NOD on pre-transplantation injury and makes NOD an attractive candidate compound for donor or organ conditioning prior to transplantation.
Collapse
Affiliation(s)
- Lamia Ait-Hsiko
- Department of Nephrology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Perioperative Minimal Induction Therapy: A Further Step toward More Effective Immunosuppression in Transplantation. J Transplant 2012; 2012:426042. [PMID: 22685630 PMCID: PMC3364007 DOI: 10.1155/2012/426042] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 02/21/2012] [Accepted: 03/12/2012] [Indexed: 01/07/2023] Open
Abstract
Dual induction with low doses of rabbit anti-human thymoglobulin (RATG) and basiliximab effectively and safely prevented allograft rejection in high-risk renal transplant recipients. To assess whether treatment timing affects efficacy and tolerability, in this single-center, matched-cohort study, we compared posttransplant outcomes in 25 patients and 50 gender-, age-, and treatment-matched reference patients induced with the same course of 7 daily RATG infusions (0.5 mg/kg/day) started before or after engraftment, respectively. All subjects received basiliximab (20 mg) before and 4 days after transplantation, withdrew steroids within 6 days after surgery, and were maintained on steroid-free immunosuppression with cyclosporine and mycophenolate mofetil or azathioprine. Over 12 months after transplant, 1 patient (4%) and 13 reference patients (26%) had acute rejection episodes. One patient and 5 reference-patients required dialysis therapy because of delayed graft function. In all patients circulating CD4+ and CD8+ T lymphocytes were fully depleted before engraftment. Both treatments were well tolerated. In kidney transplantation, perioperative RATG infusion enhances the protective effect of low-dose RATG and basiliximab induction against graft rejection and delayed function, possibly because of more effective inhibition of early interactions between circulating T cells and graft antigens.
Collapse
|
40
|
Banz Y, Hess OM, Meier P, Korchagina EY, Gordeeva EA, Robson SC, Gajanayake T, Csizmadia E, Mettler D, Haeberli A, Bovin NV, Rieben R. Evaluation of multimeric tyrosine-O-sulfate as a cytoprotectant in an in vivo model of acute myocardial infarction in pigs. Cardiology 2012; 121:59-70. [PMID: 22398380 DOI: 10.1159/000336485] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Accepted: 01/13/2012] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Intracoronary administration of glycosaminoglycan analogs, including the complement inhibitor dextran sulfate, attenuates myocardial ischemia/reperfusion injury (I/R injury). However, dextran sulfate has a distinct anticoagulatory effect, possibly limiting its use in specific situations in vivo. We therefore developed multimeric tyrosine sulfate (sTyr-PAA), a novel, minimally anticoagulatory, fully synthetic non-carbohydrate-containing polyacrylamide conjugate, for in vivo testing in an acute closed-chest porcine model of acute myocardial infarction. METHODS Following balloon occlusion of the left anterior descending artery just after the first diagonal branch (60-minute ischemia), sTyr-PAA (approx. 10 mg/kg bodyweight, fraction with strongest complement-inhibitory and minimal anticoagulatory properties, n = 11) or phosphate-buffered saline (controls, n = 9) was administered intracoronarily into ischemic myocardium prior to 120 min of reperfusion. RESULTS sTyr-PAA significantly reduced infarct size (from 61.0 ± 12.0% of the ischemic area at risk to 39.4 ± 17.0%), plasma creatine kinase, local complement deposition and tissue factor upregulation, without affecting systemic coagulation. Protection was associated with significantly reduced myocardial neutrophil extravasation and translated into a significant improvement of ejection fraction and left ventricular enddiastolic pressure. CONCLUSIONS sTyr-PAA protected significantly against myocardial I/R injury without substantially affecting systemic coagulation. Local intravascular sTyr-PAA administration may prove advantageous in situations where bleeding complications are likely or are to be avoided at all costs.
Collapse
Affiliation(s)
- Yara Banz
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Donor Desmopressin Is Associated With Superior Graft Survival After Kidney Transplantation. Transplantation 2011; 92:1252-8. [DOI: 10.1097/tp.0b013e318236cd4c] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
42
|
Abstract
Acute kidney injury occurs with kidney transplantation and too frequently progresses to the clinical diagnosis of delayed graft function (DGF). Poor kidney function in the first week of graft life is detrimental to the longevity of the allograft. Challenges to understand the root cause of DGF include several pathologic contributors derived from the donor (ischemic injury, inflammatory signaling) and recipient (reperfusion injury, the innate immune response and the adaptive immune response). Progressive demand for renal allografts has generated new organ categories that continue to carry high risk for DGF for deceased donor organ transplantation. New therapies seek to subdue the inflammatory response in organs with high likelihood to benefit from intervention. Future success in suppressing the development of DGF will require a concerted effort to anticipate and treat tissue injury throughout the arc of the transplantation process.
Collapse
Affiliation(s)
- Andrew Siedlecki
- Nephrology Division, Department of Internal Medicine, Washington University in St. Louis School of Medicine, St Louis, MO
| | - William Irish
- CTI, Clinical Trial and Consulting Services, Raleigh, NC
| | - Daniel C. Brennan
- Nephrology Division, Department of Internal Medicine, Washington University in St. Louis School of Medicine, St Louis, MO
| |
Collapse
|
43
|
Apolipoprotein A1 and C-terminal fragment of α-1 antichymotrypsin are candidate plasma biomarkers associated with acute renal allograft rejection. Transplantation 2011; 92:388-95. [PMID: 21730889 DOI: 10.1097/tp.0b013e318225db6a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Current diagnostic methods of renal allograft rejection are neither sensitive nor specific. Needle biopsies are invasive and associated with patient morbidity. Thus, it is desirable to develop noninvasive tests to predict and diagnose rejection. METHODS Using a case-control approach, surface-enhanced laser desorption/ionization time-of-flight mass spectrometry was used to identify plasma proteins associated with renal allograft rejection. From each rejection patient (n=16), two plasma samples (one near the biopsy date and the other at a time postbiopsy) were compared. Biopsy-confirmed nonrejection patients (n=48) were further analyzed as controls. Antibody-based quantitative enzyme-linked immunosorbent assay was performed to validate candidate biomarker apolipoprotein A1 (Apo A1) in a subset of the original and a second cohort of biopsy-confirmed rejection (n=40) and nonrejection (n=70) patients. RESULTS Twenty-two proteins/peptides showed significant differences between rejection and postrejection samples. Peptides 5191 Da and 4467 Da detected rejection with 100% sensitivity and 94% specificity. The 4467 Da peptide was identified as the C-terminal fragment of α-1 antichymotrypsin and a 28 kDa protein was determined as Apo A1. Both protein levels were significantly lower at rejection compared with postrejection. Protein levels of nonrejection patients were similar to the postrejection samples. Apo A1 enzyme-linked immunosorbent assay results showed significantly lower Apo A1 levels (P=0.001 for the original and P=4.14E-11 for the second cohort) at the time of rejection compared with nonrejection which coincides with the SELDI findings. CONCLUSIONS Together α-1 antichymotrypsin, Apo A1, and the unidentified 5191 Da peptide provide a plasma molecular profile, and this is associated with acute cellular renal allograft rejection.
Collapse
|
44
|
Fukuzawa N, Petro M, Baldwin WM, Gudkov AV, Fairchild RL. A TLR5 agonist inhibits acute renal ischemic failure. THE JOURNAL OF IMMUNOLOGY 2011; 187:3831-9. [PMID: 21890657 DOI: 10.4049/jimmunol.1003238] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Reperfusion of ischemic organs induces a potent inflammatory response initiated by the generation of reactive oxygen species that directly damage tissue and promote leukocyte infiltration and activation that also mediate tissue injury. We recently found that radiation-induced tissue injury, which is caused by radiation-induced reactive oxygen species, is attenuated by administration of CBLB502, a pharmacologically optimized derivative of the TLR5 agonist flagellin. Therefore, we tested the ability of CBLB502 to attenuate injury in a murine model of acute ischemic renal failure. CBLB502 given 30 min before imposition of bilateral renal pedicle occlusion provided marked protection against the renal dysfunction and inflammation that follows reperfusion of ischemic kidneys, including marked decreases in leukocyte infiltration, proinflammatory cytokine production, and tubular injury. Importantly, CBLB502 given within 30 min after ischemic kidney reperfusion reproduced the protective effects of pretreatment with the TLR5 agonist, indicating a window following reperfusion in which CBLB502 administration abrogates acute renal ischemic failure. Bone marrow-reconstituted chimeras were used to show that the protective effects of CBLB502 could be delivered by intact MyD88 signaling on renal parenchymal cells. Consistent with this, Ab staining of kidney sections indicated that cells lining the renal vasculature expressed TLR5. Overall, these results indicate the use of TLR5 agonists as mitigators and protectants of acute renal ischemic failure.
Collapse
Affiliation(s)
- Nobuyuki Fukuzawa
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | | | |
Collapse
|
45
|
Thuillier R, Dutheil D, Trieu MTN, Mallet V, Allain G, Rousselot M, Denizot M, Goujon JM, Zal F, Hauet T. Supplementation with a new therapeutic oxygen carrier reduces chronic fibrosis and organ dysfunction in kidney static preservation. Am J Transplant 2011; 11:1845-60. [PMID: 21875432 DOI: 10.1111/j.1600-6143.2011.03614.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Static preservation is currently the most widely used organ preservation strategy; however, decreased donor organ quality is impacting outcome negatively. M101 is an O₂ carrier with high-oxygen affinity and the capacity to function at low temperatures. We tested the benefits of M101 both in vitro, on cold preserved LLC-PK1, as well as in vivo, in a large white pig kidney autotransplantation model. In vitro, M101 supplementation reduced cold storage-induced cell death. In vivo, early follow-up demonstrated superiority of M101-supplemented solutions, lowering the peak of serum creatinine and increasing the speed of function recovery. On the longer term, supplementation with M101 reduced kidney inflammation levels and maintained structural integrity, particularly with University of Wisconsin (UW). At the end of the 3-month follow-up, M101 supplementation proved beneficial in terms of survival and function, as well as slowing the advance of interstitial fibrosis. We show that addition of M101 to classic organ preservation protocols with UW and Histidine-Tryptophane-Ketoglutarate, the two most widely used solutions worldwide in kidney preservation, provides significant benefits to grafts, both on early function recovery and outcome. Simple supplementation of the solution with M101 is easily translatable to the clinic and shows promises in terms of outcome.
Collapse
Affiliation(s)
- R Thuillier
- Inserm U927, Faculté de Médecine et Pharmacie, University of Poitiers, Poitiers, F86000, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dexmedetomidine provides renoprotection against ischemia-reperfusion injury in mice. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:R153. [PMID: 21702944 PMCID: PMC3219027 DOI: 10.1186/cc10283] [Citation(s) in RCA: 212] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 05/04/2011] [Accepted: 06/24/2011] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Acute kidney injury following surgery incurs significant mortality with no proven preventative therapy. We investigated whether the α2 adrenoceptor agonist dexmedetomidine (Dex) provides protection against ischemia-reperfusion induced kidney injury in vitro and in vivo. METHODS In vitro, a stabilised cell line of human kidney proximal tubular cells (HK2) was exposed to culture medium deprived of oxygen and glucose. Dex decreased HK2 cell death in a dose-dependent manner, an effect attenuated by the α2 adrenoceptor antagonist atipamezole, and likely transduced by phosphatidylinositol 3-kinase (PI3K-Akt) signaling. In vivo C57BL/6J mice received Dex (25 μg/kg, intraperitoneal (i.p.)) 30 minutes before or after either bilateral renal pedicle clamping for 25 minutes or right renal pedicle clamping for 40 minutes and left nephrectomy. RESULTS Pre- or post-treatment with Dex provided cytoprotection, improved tubular architecture and function following renal ischemia. Consistent with this cytoprotection, dexmedetomidine reduced plasma high-mobility group protein B1 (HMGB-1) elevation when given prior to or after kidney ischemia-reperfusion; pretreatment also decreased toll-like receptor 4 (TLR4) expression in tubular cells. Dex treatment provided long-term functional renoprotection, and even increased survival following nephrectomy. CONCLUSIONS Our data suggest that Dex likely activates cell survival signal pAKT via α2 adrenoceptors to reduce cell death and HMGB1 release and subsequently inhibits TLR4 signaling to provide reno-protection.
Collapse
|
47
|
Association of polymorphisms of interleukin-8, CXCR1, CXCR2, and selectin with allograft outcomes in kidney transplantation. Transplantation 2011; 91:57-64. [PMID: 21452410 DOI: 10.1097/tp.0b013e3181fd0195] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Both chemokines and adhesion molecules mediate allograft rejection by recruiting leukocytes into the allograft. We investigated the association of six single nucleotide polymorphisms (SNPs) located in interleukin (IL)-8, CXCR1, CXCR2, and selectin with kidney allograft outcomes. METHODS The promoter regions of CXCR1 and CXCR2 were sequenced directly to find SNPs. Reporter gene assay was performed to determine the transcriptional activity of CXCR2 promoter polymorphisms. The association of SNPs in IL-8, CXCR1, CXCR2, and selectin with both acute rejection and estimated glomerular filtration rate at 1-year posttransplant was analyzed in 216 donor-recipient pairs of kidney transplantation. RESULTS The donor GA/AA genotypes of CXCR1 -2668G/A (rs2671222) were associated with increased risk for acute rejection even after adjusting for covariates such as gender, diabetes, preemptive transplantation, immunosuppressive regimen, relationship with the donor, and human leukocyte antigen mismatch (adjusted odds ratio 3.56; 95% confidence interval 1.37-9.27; P=0.009). Although the transcriptional activity of the CXCR2 variant promoter was 2.6-fold higher than that of the wild-type promoter (P=0.039), no significant association was observed between CXCR2 polymorphisms and kidney allograft outcomes. SNPs of IL-8, L-selectin, and E-selectin were not associated with kidney allograft outcomes. CONCLUSION The donor CXCR1 -2668 GA/AA genotypes were an independent risk factor for acute rejection in kidney transplantation.
Collapse
|
48
|
Tubular epithelial injury and inflammation after ischemia and reperfusion in human kidney transplantation. Ann Surg 2011; 253:598-604. [PMID: 21248631 DOI: 10.1097/sla.0b013e31820d9ae9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To provide an integrated insight into the kinetics of tubular injury, inflammation, and oxidative stress after human kidney transplantation. BACKGROUND Tissue injury due to ischemia and reperfusion is an inevitable consequence of kidney transplantation. Tubular epithelial injury, inflammation, and oxidative stress play major roles in the pathophysiology of acute kidney injury in small animals, but it remains to be established whether this paradigm holds true for human kidney transplantation. METHODS Markers of tubular injury, inflammation, and oxidative stress were compared between recipients of kidneys from donors after cardiac death (DCD; N = 8) with prolonged ischemia and recipients of living donor kidneys with minimal ischemia (N = 8). RESULTS In the early postoperative period, creatinine clearance and tubular sodium reabsorption were profoundly reduced in DCD kidneys, coinciding with significantly increased urinary concentrations of tubular injury markers (neutrophil gelatinase-associated lipocalin, N-acetyl-β--glucosaminidase, and cystatin C) and an 18-fold increase in renal production of cytokeratin-18, indicating extensive necrotic cell death. Tubular injury in DCD kidneys was followed by greater systemic inflammatory activity and oxidative stress in the postoperative period (measured with 17-plex cytokine arrays and as plasma F2-isoprostanes, respectively). In contrast, no evidence of oxidative damage to either of the 2 kidney types was found in the early reperfusion period. CONCLUSIONS These findings establish the relevance of observations in animal models for human kidney transplantation and form the basis for development of novel therapies to improve early graft function and expand the use of donor kidneys with prolonged ischemia.
Collapse
|
49
|
|
50
|
Thuillier R, Renard C, Rogel-Gaillard C, Demars J, Milan D, Forestier L, Ouldmoulene A, Goujon JM, Badet L, Hauet T. Effect of polyethylene glycol-based preservation solutions on graft injury in experimental kidney transplantation. Br J Surg 2010; 98:368-78. [DOI: 10.1002/bjs.7332] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2010] [Indexed: 12/12/2022]
Abstract
Abstract
Background
New preservation solutions are emerging, of various ionic compositions and with hydroxyethyl starch replaced by polymers such as polyethylene glycols (PEGs), offering the potential for ‘immunocamouflage’. This experimental study investigated which of three clinically available preservation protocols offered the best graft protection, based on epithelial-to-mesenchymal transition (EMT) and fibrosis.
Methods
Kidneys were preserved for 24 h at 4 °C with University of Wisconsin solution (UW) as standard, compared with solutions containing either 1 g/l PEG 35 kDa (Institute Georges Lopez solution, IGL) or 30g/l PEG 20 kDa (solution de conservation des organes et des tissus, SCOT). Animals were followed for up to 3 months and development of EMT, tubular atrophy and fibrosis was evaluated in comparison with sham-operated animals.
Results
Functional recovery was better in the SCOT group compared with the other groups. Chronic fibrosis, EMT and inflammation were observed in the UW and IGL groups, but limited in the SCOT group. Levels of profibrosis markers such as transforming growth factor β1, plasminogen activator inhibitor 1 and connective tissue growth factor were increased in IGL and UW groups compared with the SCOT group. Hypoxia-inducible factor (HIF) 1α and 2α expression was increased at 3 months in grafts preserved in UW and IGL, but detected transiently on day 14 when SCOT was used. Expression of HIF-regulated genes vascular endothelial growth factor and erythropoietin was increased in UW and IGL groups.
Conclusion
The choice of colloid and ionic content is paramount in providing long-term protection against chronic graft injury after renal transplantation. Preservation solutions based on PEGs may optimize graft quality.
Collapse
Affiliation(s)
- R Thuillier
- Institut National de la Santé et de la Recherche Médicale (INSERM) U927 and Université de Poitiers, Poitiers, France
- Centre Hospitalier Universitaire de Poitiers, Poitiers, France
- Fédération pour l'Étude de l'Ischémie Reperfusion en Transplantation (FLIRT), Poitiers, France
| | - C Renard
- Laboratoire de Radiobiologie et d'Étude du Génome, Unité Mixte de Recherche (UMR) 1061, Commissariat à l'Énergie Atomique-Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France
| | - C Rogel-Gaillard
- Laboratoire de Radiobiologie et d'Étude du Génome, Unité Mixte de Recherche (UMR) 1061, Commissariat à l'Énergie Atomique-Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France
| | - J Demars
- Laboratoire de Génétique Cellulaire, UMR 444, INRA, Castanet Tolosan, Limoges, France
| | - D Milan
- Laboratoire de Génétique Cellulaire, UMR 444, INRA, Castanet Tolosan, Limoges, France
| | - L Forestier
- Laboratoire de Génétique Moléculaire Animale, UMR 1061, Limoges, France
| | - A Ouldmoulene
- Laboratoire de Génétique Moléculaire Animale, UMR 1061, Limoges, France
| | - J M Goujon
- Institut National de la Santé et de la Recherche Médicale (INSERM) U927 and Université de Poitiers, Poitiers, France
| | - L Badet
- Institut National de la Santé et de la Recherche Médicale (INSERM) U927 and Université de Poitiers, Poitiers, France
- Fédération pour l'Étude de l'Ischémie Reperfusion en Transplantation (FLIRT), Poitiers, France
- Université Claude Bernard Lyon 1, Villeurbanne, France
- CENTAURE Network, Nantes, France
| | - T Hauet
- Centre Hospitalier Universitaire de Poitiers, Poitiers, France
- Fédération pour l'Étude de l'Ischémie Reperfusion en Transplantation (FLIRT), Poitiers, France
- Plateforme IBiSA, Génétique et Expérimentation en Productions Animales, INRA, Domaine du Magneraud, Surgères, France
| |
Collapse
|