1
|
Grimault O, Theron M, Boete Q, Maalouf M, Quéméneur JB, Zal F, Léon K, Pichavant-Rafini K. Investigating M101's Effect in Improving Cardiac and Diaphragmatic Mechanical and Functional Properties on Wistar Rat. Artif Organs 2025. [PMID: 40326531 DOI: 10.1111/aor.15014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND The critical importance of effective oxygen delivery during organ preservation to prevent ischemia-reperfusion injury has been reported in the literature. Moreover, traditional preservation methods often fail to meet oxygenation needs, prompting research into alternatives like hemoglobin-based oxygen carriers (HBOCs) such as M101 provided by HEMARINA. This extracellular hemoglobin, derived from Arenicola marina, shows a high affinity for oxygen and delivers it through a physiological gradient under hypoxic conditions. METHODS In this context, the mechanical and functional properties of the heart and diaphragm muscles from Wistar rats were evaluated in the presence of M101. RESULTS The experiments showed that M101-treated hearts maintained higher contraction amplitudes, heart rates, and coronary flow over time compared to controls, indicating improved functional stability. Furthermore, the M101-treated diaphragm exhibited prolonged contraction and relaxation times, with significant improvements in contraction amplitude at higher stimulation frequencies. Finally, the Burke index revealed reduced fatigue in the M101-treated group compared to the controls. CONCLUSIONS These findings suggest that M101 significantly enhances the mechanical and functional properties of both tested muscles under experimental conditions. Further research is warranted to unravel these findings and explore clinical applications.
Collapse
Affiliation(s)
- Ollivier Grimault
- Université de Brest, EA 4324 ORPHY, IBSAM, UFR Sciences et Techniques, Brest, France
| | - Michael Theron
- Université de Brest, EA 4324 ORPHY, IBSAM, UFR Sciences et Techniques, Brest, France
| | - Quentin Boete
- Université de Brest, EA 4324 ORPHY, IBSAM, UFR Sciences et Techniques, Brest, France
| | - Mathieu Maalouf
- Université de Brest, EA 4324 ORPHY, IBSAM, UFR Sciences et Techniques, Brest, France
| | | | - Franck Zal
- Hemarina SA, Aeropôle Centre, Morlaix, France
| | - Karelle Léon
- Université de Brest, EA 4324 ORPHY, IBSAM, UFR Sciences et Techniques, Brest, France
| | | |
Collapse
|
2
|
Zhang P, Liang H, Wang Y. Advances and Challenges of Thrombolytic Therapy for Donation After Circulatory Death Organs. Clin Transplant 2025; 39:e70099. [PMID: 39921605 DOI: 10.1111/ctr.70099] [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: 11/18/2024] [Revised: 01/15/2025] [Accepted: 01/23/2025] [Indexed: 02/10/2025]
Abstract
The demand for organ transplantation has exceeded the global supply of available organs. Donation after circulatory death (DCD) is considered an effective method to solve the disparity between the supply and demand of organs, by expanding the donor pool. However, DCD organs experience long-term damage caused by warm ischemia (WI) and microthrombosis caused by diffuse intravascular coagulation. Unfortunately, because of concerns about post-transplantation complications, most organs considered high-risk are discarded, resulting in wasted medical resources and economic losses. However, thrombolytic therapy before transplantation may dissolve microthrombosis in DCD organs, improve organ microcirculation, and increase organ use. Herein, we review the current status and potential value of thrombolytic therapy before DCD organ transplantation, summarize the progress of thrombolytic therapy for DCD organ transplantation according to preclinical and clinical research, and emphasize the heterogeneity and limitations of studies that have caused some controversies associated with this therapy. Overall, the role of thrombolytic therapy should not be overlooked. We anticipate that thrombolytic therapy combined with machine perfusion will provide an opportunity to improve inferior-quality DCD grafts, resulting in their becoming more widely available and safer for transplantation, thus solving the urgent problem of organ shortage.
Collapse
Affiliation(s)
- Peng Zhang
- Department of General Surgery, Division of Hepatobiliary and Pancreas Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, P.R. China
| | - Han Liang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, P.R. China
| |
Collapse
|
3
|
Carré J, Kerforne T, Hauet T, Macchi L. Tissue Injury Protection: The Other Face of Anticoagulant Treatments in the Context of Ischemia and Reperfusion Injury with a Focus on Transplantation. Int J Mol Sci 2023; 24:17491. [PMID: 38139319 PMCID: PMC10743711 DOI: 10.3390/ijms242417491] [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: 11/17/2023] [Revised: 12/06/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
Organ transplantation has enhanced the length and quality of life of patients suffering from life-threatening organ failure. Donors deceased after brain death (DBDDs) have been a primary source of organs for transplantation for a long time, but the need to find new strategies to face organ shortages has led to the broadening of the criteria for selecting DBDDs and advancing utilization of donors deceased after circulatory death. These new sources of organs come with an elevated risk of procuring organs of suboptimal quality. Whatever the source of organs for transplant, one constant issue is the occurrence of ischemia-reperfusion (IR) injury. The latter results from the variation of oxygen supply during the sequence of ischemia and reperfusion, from organ procurement to the restoration of blood circulation, triggering many deleterious interdependent processes involving biochemical, immune, vascular and coagulation systems. In this review, we focus on the roles of thrombo-inflammation and coagulation as part of IR injury, and we give an overview of the state of the art and perspectives on anticoagulant therapies in the field of transplantation, discussing benefits and risks and proposing a strategic guide to their use during transplantation procedures.
Collapse
Affiliation(s)
- Julie Carré
- Service D’Hématologie Biologique, Centre Hospitalo-Universitaire de Poitiers, 86000 Poitiers, France;
- INSERM 1313 Ischémie Reperfusion, Métabolisme, Inflammation Stérile en Transplantation (IRMETIST), Université de Poitiers, 86000 Poitiers, France; (T.K.); (T.H.)
| | - Thomas Kerforne
- INSERM 1313 Ischémie Reperfusion, Métabolisme, Inflammation Stérile en Transplantation (IRMETIST), Université de Poitiers, 86000 Poitiers, France; (T.K.); (T.H.)
- Service D’Anesthésie-Réanimation et Médecine Péri-Opératoire, Centre Hospitalo-Universitaire de Poitiers, 86000 Poitiers, France
- FHU Survival Optimization in Organ Transplantation (SUPORT), 86000 Poitiers, France
| | - Thierry Hauet
- INSERM 1313 Ischémie Reperfusion, Métabolisme, Inflammation Stérile en Transplantation (IRMETIST), Université de Poitiers, 86000 Poitiers, France; (T.K.); (T.H.)
- FHU Survival Optimization in Organ Transplantation (SUPORT), 86000 Poitiers, France
- Service de Biochimie, Centre Hospitalo-Universitaire de Poitiers, 86000 Poitiers, France
| | - Laurent Macchi
- Service D’Hématologie Biologique, Centre Hospitalo-Universitaire de Poitiers, 86000 Poitiers, France;
- INSERM 1313 Ischémie Reperfusion, Métabolisme, Inflammation Stérile en Transplantation (IRMETIST), Université de Poitiers, 86000 Poitiers, France; (T.K.); (T.H.)
- FHU Survival Optimization in Organ Transplantation (SUPORT), 86000 Poitiers, France
| |
Collapse
|
4
|
O'Neill S, Asgari E, Callaghan C, Gardiner D, Hartog H, Iype S, Manara A, Nasralla D, Oniscu GC, Watson C. The British transplantation society guidelines on organ donation from deceased donors after circulatory death. Transplant Rev (Orlando) 2023; 37:100791. [PMID: 37598591 DOI: 10.1016/j.trre.2023.100791] [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: 08/08/2023] [Revised: 08/11/2023] [Accepted: 08/11/2023] [Indexed: 08/22/2023]
Abstract
Recipient outcomes after transplantation with organs from donation after circulatory death (DCD) donors can compare favourably and even match recipient outcomes after transplantation with organs from donation after brain death donors. Success is dependent upon establishing common practices and accepted protocols that allow the safe sharing of DCD organs and maximise the use of the DCD donor pool. The British Transplantation Society 'Guideline on transplantation from deceased donors after circulatory death' has recently been updated. This manuscript summarises the relevant recommendations from chapters specifically related to organ donation.
Collapse
Affiliation(s)
- Stephen O'Neill
- Consultant Transplant Surgeon, Belfast City Hospital, Belfast, Northern Ireland.
| | - Ellie Asgari
- Consultant Nephrologist, Guy's Hospital, London, England
| | - Chris Callaghan
- Consultant Transplant Surgeon, Guy's Hospital, London, England
| | - Dale Gardiner
- Consultant Intensivist, Queen's Medical Centre, Nottingham, England
| | - Hermien Hartog
- Consultant Transplant Surgeon, Queen Elizabeth Hospital, Birmingham, England
| | - Satheesh Iype
- Consultant Transplant Surgeon, Royal Free Hospital, London, England
| | - Alex Manara
- Consultant Intensivist, Southmead Hospital, Bristol, England
| | - David Nasralla
- Consultant Transplant Surgeon, Royal Free Hospital, London, England
| | - Gabi C Oniscu
- Consultant Transplant Surgeon, Royal Infirmary, Edinburgh, Scotland
| | - Chris Watson
- Consultant Transplant Surgeon, Addenbrooke's Hospital, Cambridge, England
| |
Collapse
|
5
|
Cao M, Wang G, He H, Yue R, Zhao Y, Pan L, Huang W, Guo Y, Yin T, Ma L, Zhang D, Huang X. Hemoglobin-Based Oxygen Carriers: Potential Applications in Solid Organ Preservation. Front Pharmacol 2021; 12:760215. [PMID: 34916938 PMCID: PMC8670084 DOI: 10.3389/fphar.2021.760215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/10/2021] [Indexed: 12/30/2022] Open
Abstract
Ameliorating graft injury induced by ischemia and hypoxia, expanding the donor pool, and improving graft quality and recipient prognosis are still goals pursued by the transplant community. The preservation of organs during this process from donor to recipient is critical to the prognosis of both the graft and the recipient. At present, static cold storage, which is most widely used in clinical practice, not only reduces cell metabolism and oxygen demand through low temperature but also prevents cell edema and resists apoptosis through the application of traditional preservation solutions, but these do not improve hypoxia and increase oxygenation of the donor organ. In recent years, improving the ischemia and hypoxia of grafts during preservation and repairing the quality of marginal donor organs have been of great concern. Hemoglobin-based oxygen carriers (HBOCs) are “made of” natural hemoglobins that were originally developed as blood substitutes but have been extended to a variety of hypoxic clinical situations due to their ability to release oxygen. Compared with traditional preservation protocols, the addition of HBOCs to traditional preservation protocols provides more oxygen to organs to meet their energy metabolic needs, prolong preservation time, reduce ischemia–reperfusion injury to grafts, improve graft quality, and even increase the number of transplantable donors. The focus of the present study was to review the potential applications of HBOCs in solid organ preservation and provide new approaches to understanding the mechanism of the promising strategies for organ preservation.
Collapse
Affiliation(s)
- Min Cao
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Guoqing Wang
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Hongli He
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruiming Yue
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yong Zhao
- Anesthesiology, Southwest Medicine University, Luzhou, China
| | - Lingai Pan
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Weiwei Huang
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Guo
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Tao Yin
- Surgical Department, Chengdu Second People's Hospital, Chengdu, China
| | - Lina Ma
- Health Inspection and Quarantine, Chengdu Medical College, Chengdu, China
| | - Dingding Zhang
- Sichuan Provincial Key Laboratory for Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaobo Huang
- Department of Critical Care Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
6
|
Legeai C, Durand L, Savoye E, Macher MA, Bastien O. Effect of preservation solutions for static cold storage on kidney transplantation outcomes: A National Registry Study. Am J Transplant 2020; 20:3426-3442. [PMID: 32400921 DOI: 10.1111/ajt.15995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/25/2020] [Accepted: 04/22/2020] [Indexed: 01/25/2023]
Abstract
This study aimed to evaluate how 5 preservation solutions for static cold storage affected kidney transplant outcomes. It included all first single kidney transplants during 2010-2014 from donations after brain death in the French national transplant registry, excluding preemptive transplants and transplants of kidneys preserved with a hypothermic perfusion machine. The effects of each preservation solution on delayed graft function (DGF) and 1-year transplant failure were evaluated with hierarchical multivariable logistic regression models. The study finally included 7640 transplanted kidneys: 3473 (45.5%) preserved with Institut Georges Lopez-1 solution (IGL-1), 773 (10.1%) with University of Wisconsin solution, 731 (9.6%) with Solution de Conservation des Organes et Tissus (SCOT, organ and tissue preservation solution), 2215 (29.0%) with Celsior, and 448 (5.9%) with histidine-tryptophan-ketoglutarate. Primary nonfunction rates did not differ by solution. After adjustment for donor, recipient, and transplant characteristics, the DGF risk was significantly lower with IGL-1 than with all other solutions (odds ratio [OR] 0.55, 95% confidence interval [CI] 0.48-0.64). Conversely, SCOT was associated with a DGF risk significantly higher than the other solutions (OR 2.69, 95% CI 2.21-3.27) and triple that of IGL-1 (OR 3.37, 95% CI 2.72-4.16). One year after transplantation, the transplant failure rate did not differ significantly by preservation solution. The difference between the groups for 1-year mean creatinine clearance was not clinically relevant.
Collapse
Affiliation(s)
- Camille Legeai
- Organ and Tissue Procurement and Transplantation Department, Agence de la Biomédecine, Saint Denis La Plaine, France
| | - Louise Durand
- Organ and Tissue Procurement and Transplantation Department, Agence de la Biomédecine, Saint Denis La Plaine, France
| | - Emilie Savoye
- Organ and Tissue Procurement and Transplantation Department, Agence de la Biomédecine, Saint Denis La Plaine, France
| | - Marie-Alice Macher
- Organ and Tissue Procurement and Transplantation Department, Agence de la Biomédecine, Saint Denis La Plaine, France
| | - Olivier Bastien
- Organ and Tissue Procurement and Transplantation Department, Agence de la Biomédecine, Saint Denis La Plaine, France
| |
Collapse
|
7
|
Xia M, Liu D, Peng L, Li Y, Liu H, Wu L, Chen G, Liu Y, Liu H. Coagulation parameters are associated with the prognosis of immunoglobulin a nephropathy: a retrospective study. BMC Nephrol 2020; 21:447. [PMID: 33109129 PMCID: PMC7590710 DOI: 10.1186/s12882-020-02111-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/18/2020] [Indexed: 01/15/2023] Open
Abstract
Background Interstitial fibrosis/tubular atrophy (T) score is a known determinant of the progression of immunoglobulin A nephropathy (IgAN). Strong evidence indicates that the components of the coagulation system closely linked with fibrotic events have been highlighted in the kidney. However, whether the coagulation system can affect the renal outcome of IgAN remains unclear. Herein, we investigated the association of coagulation parameters and pathological phenotype of IgAN and their combined effects on the deterioration of renal function. Methods This retrospective study included N = 291 patients with biopsy-proven IgAN from May 2009 to April 2013 in the Second Xiangya Hospital. Clinical data, pathological features were collected, and the associations of coagulation parameters at biopsy, T score, and renal outcome were evaluated. T score indicated the degree of tubular atrophy or interstitial fibrosis. The renal outcome was defined as an end-stage renal disease (ESRD) or an irreversible 50% estimated glomerular filtration rate (eGFR) reduction. Results Shorter prothrombin time (PT) and the activated partial thromboplastin time (APTT) were significantly associated with T (both p < 0.001). PT (< 11.15 s) or APTT (< 29.65 s) had worse cumulative survival rate (p = 0.008, p = 0.027 respectively) and were significantly but not independently associated with a higher risk of renal outcome (p = 0.012, p = 0.032 respectively). In the combined analyses of PT, APTT, and T lesions, the odd ratios for the outcome were significantly higher in the presence of T with PT (< 11.15 s) or APTT (< 29.65 s). Conclusion Shorter PT and APTT are associated with an increased incidence of the T lesion and are additional factors that portend a poorer prognosis in IgAN. Monitoring coagulation function might be important when assessing the risk of progression. Additional studies exploring the molecular mechanism between coagulation and IgAN pathology are needed.
Collapse
Affiliation(s)
- Ming Xia
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Di Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Liang Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Yan Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Haiyang Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Lingzhi Wu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Guochun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, 410011, Hunan, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China. .,Hunan Key Laboratory of Kidney Disease and Blood Purification, 139 Renmin Road, Changsha, 410011, Hunan, China.
| |
Collapse
|
8
|
Chen Y, Shi J, Xia TC, Xu R, He X, Xia Y. Preservation Solutions for Kidney Transplantation: History, Advances and Mechanisms. Cell Transplant 2019; 28:1472-1489. [PMID: 31450971 PMCID: PMC6923544 DOI: 10.1177/0963689719872699] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Solid organ transplantation was one of the greatest medical advances during the past few
decades. Organ preservation solutions have been applied to diminish ischemic/hypoxic
injury during cold storage and improve graft survival. In this article, we provide a
general review of the history and advances of preservation solutions for kidney
transplantation. Key components of commonly used solutions are listed, and effective
supplementations for current available preservation solutions are discussed. At cellular
and molecular levels, further insights were provided into the pathophysiological
mechanisms of effective ingredients against ischemic/hypoxic renal injury during cold
storage. We pay special attention to the cellular and molecular events during
transplantation, including ATP depletion, acidosis, mitochondrial dysfunction, oxidative
stress, inflammation, and other intracellular mechanisms.
Collapse
Affiliation(s)
- Yimeng Chen
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jian Shi
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Terry C Xia
- The University of Connecticut, Storrs, CT, USA
| | - Renfang Xu
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Xiaozhou He
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ying Xia
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Kaminski J, Delpech PO, Kaaki-Hosni S, Promeyrat X, Hauet T, Hannaert P. Oxygen Consumption by Warm Ischemia-Injured Porcine Kidneys in Hypothermic Static and Machine Preservation. J Surg Res 2019; 242:78-86. [PMID: 31071608 DOI: 10.1016/j.jss.2019.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/26/2019] [Accepted: 04/04/2019] [Indexed: 01/20/2023]
Abstract
Static cold storage (SCS) and hypothermic machine perfusion (HMP) are currently standard methods for renal grafts clinical preservation. Both methods are predominantly implemented without the active delivery of oxygen, even for donation after circulatory death-like kidneys. However, even under severe hypothermia (4°C-6°C), kidneys can consume oxygen and produce ATP. What is not established, though, is to what extent and how SCS and HMP compare in terms of oxygen. Using a porcine preclinical model of renal warm ischemia (WI) to compare SCS and HMP methods, we continuously monitored and quantified oxygen level and consumption along preservation; we also determined prepreservation and postpreservation cortical ATP level; values were given as median and [min; max] range. One-hour WI reduced ATP by ∼90% (from 3.3 [1.7; 4.5] mmol/L tissue in Controls). Oxygen consumption (QO2, μmol/min per 100 g) was determined from initial solution PO2 decrease (SCS and HMP) and from arterio-venous difference (HMP). In SCS and HMP, PO2 decreased rapidly (t1/2 ∼1 h) from atmospheric levels to 52.9 [38.0; 65.9] and 8.2 [3.0, 16.0] mmHg, respectively. In HMP, QO2 was 2.7 [0.4; 3.9] versus 0.5 [0.0; 1.3] in SCS (P < 0.05); postpreservation ATP amounted to 5.8 [3.2; 6.5] in HMP versus 0.1 [0.0; 0.2] in SCS. Despite hypothermic conditions in SCS or HMP, donation after circulatory death-like renal grafts require oxygen. Increased oxygen consumption, restored ATP level, and improved histological profile in HMP might explain the established HMP superiority over SCS. These results establish a rational basis for the use of oxygen in hypothermic preservation. Optimal levels required for preservation and graft-type variants remain to be determined.
Collapse
Affiliation(s)
| | - Pierre-Olivier Delpech
- INSERM U1082-IRTOMIT, CHU de Poitiers, Poitiers, France; Service d'Urologie, CHU de Poitiers, Poitiers, France
| | | | - Xavier Promeyrat
- Service d'Urologie et de Chirurgie de la Transplantation, Hôpital Édouard-Herriot, Université Claude-Bernard Lyon 1, Lyon, France
| | - Thierry Hauet
- INSERM U1082-IRTOMIT, CHU de Poitiers, Poitiers, France; Service de Biochimie, CHU de Poitiers, Poitiers, France
| | | |
Collapse
|
10
|
Kaminski J, Hannaert P, Kasil A, Thuillier R, Leize E, Delpy E, Steichen C, Goujon JM, Zal F, Hauet T. Efficacy of the natural oxygen transporter HEMO 2 life ® in cold preservation in a preclinical porcine model of donation after cardiac death. Transpl Int 2019; 32:985-996. [PMID: 30924562 DOI: 10.1111/tri.13434] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/03/2019] [Accepted: 03/25/2019] [Indexed: 12/11/2022]
Abstract
The growing use of marginal organs for transplantation pushes current preservation methods toward their limits, and the need for improvement is pressing. We previously demonstrated the benefits of M101, a natural extracellular oxygen carrier compatible with hypothermia, for the preservation of healthy renal grafts in a porcine model of autotransplantation. Herein, we use a variant of this preclinical model to evaluate M101 potential benefits both in static cold storage (CS) and in machine perfusion (MP) preservation in the transplantation outcomes for marginal kidneys. In the CS arm, despite the absence of obvious benefits within the first 2 weeks of follow-up, M101 dose-dependently improved long-term function, normalizing creatininemia after 1 and 3 months. In the MP arm, M101 improved short- and long-term functional outcomes as well as tissue integrity. Importantly, we provide evidence for the additivity of MP and M101 functional effects, showing that the addition of the compound further improves organ preservation, by reducing short-term function loss, with no loss of function or tissue integrity recorded throughout the follow-up. Extending previous observations with healthy kidneys, the present results point at the M101 oxygen carrier as a viable strategy to improve current organ preservation methods in marginal organ transplantation.
Collapse
Affiliation(s)
- Jacques Kaminski
- Inserm U1082, Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France
| | - Patrick Hannaert
- Inserm U1082, Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France
| | - Abdelsalam Kasil
- Inserm U1082, Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France
| | - Raphael Thuillier
- Inserm U1082, Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Service de Biochimie, CHU de Poitiers, Poitiers, France
| | | | - Eric Delpy
- Aéropôle Centre, HEMARINA, Morlaix, France
| | - Clara Steichen
- Inserm U1082, Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France
| | - Jean Michel Goujon
- Inserm U1082, Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France
| | - Franck Zal
- Aéropôle Centre, HEMARINA, Morlaix, France
| | - Thierry Hauet
- Inserm U1082, Faculté de Médecine et Pharmacie, Université de Poitiers, Poitiers, France.,Service de Biochimie, CHU de Poitiers, Poitiers, France.,Fédération Hospitalo-Universitaire SUPORT, Poitiers, France.,Département de Génétique Animale, INRA, Plate-forme IBiSA, GENESI, Domaine du Magneraud, Surgères, France
| |
Collapse
|
11
|
Nifedipine Modulates Renal Lipogenesis via the AMPK-SREBP Transcriptional Pathway. Int J Mol Sci 2019; 20:ijms20071570. [PMID: 30934807 PMCID: PMC6480582 DOI: 10.3390/ijms20071570] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/16/2019] [Accepted: 03/27/2019] [Indexed: 12/13/2022] Open
Abstract
Lipid accumulation in renal cells has been implicated in the pathogenesis of obesity-related kidney disease, and lipotoxicity in the kidney can be a surrogate marker for renal failure or renal fibrosis. Fatty acid oxidation provides energy to renal tubular cells. Ca2+ is required for mitochondrial ATP production and to decrease reactive oxygen species (ROS). However, how nifedipine (a calcium channel blocker) affects lipogenesis is unknown. We utilized rat NRK52E cells pre-treated with varying concentrations of nifedipine to examine the activity of lipogenesis enzymes and lipotoxicity. A positive control exposed to oleic acid was used for comparison. Nifedipine was found to activate acetyl Coenzyme A (CoA) synthetase, acetyl CoA carboxylase, long chain fatty acyl CoA elongase, ATP-citrate lyase, and 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG CoA) reductase, suggesting elevated production of cholesterol and phospholipids. Nifedipine exposure induced a vast accumulation of cytosolic free fatty acids (FFA) and stimulated the production of reactive oxygen species, upregulated CD36 and KIM-1 (kidney injury molecule-1) expression, inhibited p-AMPK activity, and triggered the expression of SREBP-1/2 and lipin-1, underscoring the potential of nifedipine to induce lipotoxicity with renal damage. To our knowledge, this is the first report demonstrating nifedipine-induced lipid accumulation in the kidney.
Collapse
|
12
|
Kerforne T, Favreau F, Khalifeh T, Maiga S, Allain G, Thierry A, Dierick M, Baulier E, Steichen C, Hauet T. Hypercholesterolemia-induced increase in plasma oxidized LDL abrogated pro angiogenic response in kidney grafts. J Transl Med 2019; 17:26. [PMID: 30642356 PMCID: PMC6332834 DOI: 10.1186/s12967-018-1764-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/31/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Renal transplantation is increasingly associated with the presence of comorbidity factors such as dyslipidemia which could influence the graft outcome. We hypothesized that hypercholesterolemia could affect vascular repair processes and promote post-transplant renal vascular remodeling through the over-expression of the anti-angiogenic thrombospondin-1 interacting with vascular endothelial growth factor-A levels. METHODS We tested this hypothesis in vitro, in vivo and in a human cohort using (1) endothelial cells; (2) kidney auto-transplanted pig subjected (n = 5) or not (n = 6) to a diet enriched in cholesterol and (3) a renal transplanted patient cohort (16 patients). RESULTS Cells exposed to oxidized LDL showed reduced proliferation and an increased expression of thrombospondin-1. In pigs, 3 months after transplantation of kidney grafts, we observed a deregulation of the hypoxia inducible factor 1a-vascular endothelial growth factor-A axis induced in cholesterol-enriched diet animals concomitant with an overexpression of thrombospondin-1 and a decrease in cortical microvessel density promoting vascular remodeling. In patients, hypercholesterolemia was associated with decreased vascular endothelial growth factor-A plasma levels during early follow up after renal transplantation and increased chronic graft dysfunction. CONCLUSIONS These results support a potential mechanism through which a high fat-diet impedes vascular repair in kidney graft and suggest the value of controlling cholesterolemia in recipient even at the early stage of renal transplantation.
Collapse
Affiliation(s)
- Thomas Kerforne
- INSERM U1082 IRTOMIT, 2 rue de la Milétrie, CS90577, 86000 Poitiers, France
- Service d’Anesthésie-Réanimation, CHU de Poitiers, 86000 Poitiers, France
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86000 Poitiers, France
| | - Frédéric Favreau
- INSERM U1082 IRTOMIT, 2 rue de la Milétrie, CS90577, 86000 Poitiers, France
- Faculté de Médecine, EA 6309 “Maintenance Myélinique et Neuropathies Périphériques», Université de Limoges, 87000 Limoges, France
- Laboratoire de Biochimie et Génétique Moléculaire, CHU de Limoges, 87000 Limoges, France
| | - Tackwa Khalifeh
- INSERM U1082 IRTOMIT, 2 rue de la Milétrie, CS90577, 86000 Poitiers, France
- Service Medico-Chirurgical de Pediatrie, CHU de Poitiers, 86000 Poitiers, France
| | - Souleymane Maiga
- INSERM U1082 IRTOMIT, 2 rue de la Milétrie, CS90577, 86000 Poitiers, France
| | - Geraldine Allain
- INSERM U1082 IRTOMIT, 2 rue de la Milétrie, CS90577, 86000 Poitiers, France
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86000 Poitiers, France
- Service de Chirurgie Cardio-Thoracique, CHU de Poitiers, 86000 Poitiers, France
| | - Antoine Thierry
- INSERM U1082 IRTOMIT, 2 rue de la Milétrie, CS90577, 86000 Poitiers, France
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86000 Poitiers, France
- Service de Néphrologie et Transplantation, CHU de Poitiers, 86000 Poitiers, France
| | - Manuel Dierick
- UGCT-Department of Physics and Astronomy, Faculty of Sciences, Ghent University, Proeftuinstraat 86, 9000 Ghent, Belgium
| | - Edouard Baulier
- INSERM U1082 IRTOMIT, 2 rue de la Milétrie, CS90577, 86000 Poitiers, France
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86000 Poitiers, France
- Service de Biochimie, CHU de Poitiers, Poitiers, 86000 France
| | - Clara Steichen
- INSERM U1082 IRTOMIT, 2 rue de la Milétrie, CS90577, 86000 Poitiers, France
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86000 Poitiers, France
| | - Thierry Hauet
- INSERM U1082 IRTOMIT, 2 rue de la Milétrie, CS90577, 86000 Poitiers, France
- Faculté de Médecine et de Pharmacie, Université de Poitiers, 86000 Poitiers, France
- Service de Biochimie, CHU de Poitiers, Poitiers, 86000 France
- IBiSA ‘Plate-Forme MOdélisation Préclinique-Innovations Chirurgicale et Technologique (MOPICT)’, Domaine Expérimental du Magneraud, 17700 Surgères, France
- FHU SUPORT ‘SUrvival oPtimization in ORgan Transplantation’, 86000 Poitiers, France
| |
Collapse
|
13
|
Cury MFR, Olivares EQ, Garcias RC, Toledo GQ, Anselmo NA, Paskakulis LC, Botelho FFR, Carvalho NZ, Silva AAD, Agren C, Carlos CP. Inflammation and kidney injury attenuated by prior intake of Brazil nuts in the process of ischemia and reperfusion. ACTA ACUST UNITED AC 2018; 40:312-318. [PMID: 30118536 PMCID: PMC6533994 DOI: 10.1590/2175-8239-jbn-2018-0016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022]
Abstract
Introduction: Ischemia and reperfusion (IR) is a process inherent to the procedures
involved in the transplantation of organs that causes inflammation, cell
death and cell injury, and may lead to rejection of the graft. It is
possible that the anti-inflammatory properties of the Brazil nuts (BN) can
mitigate the renal injury caused by IR. Objective: To investigate whether the previous intake of BN reduces the expression of
markers of inflammation, injury, and cell death after renal IR. Methods: Male Wistar rats were distributed into six groups (N = 6/group): SHAM
(control), SHAM treated with 75 or 150 mg of BN, IR, and IR treated with 75
or 150 mg of BN. The IR procedure consisted of right nephrectomy and
occlusion of the left renal artery with a non-traumatic vascular clamp for
30 min. BN was given daily from day 1 to 7 before surgery (SHAM or IR), and
maintained until sacrifice (48 h after surgery). Inflammation was evaluated
by renal expression of COX-2 and TGF-β, injury by the expression of
vimentin, and cell death by apoptosis through caspase-3 expression
(immunohistochemistry). Results: Pretreatment with 75 mg of BN reduced renal expression of the COX-2, TGF-β,
vimentin, and caspase-3. The dose of 150 mg caused increased expression of
COX-2. Conclusion: In experimental IR, the damage can be minimized with a prior low-dose intake
of BN, improving inflammation, injury, and cell death.
Collapse
Affiliation(s)
| | - Estéfany Queiroz Olivares
- Laboratório de Pesquisa Experimental, FACERES Faculdade de Medicina, São José do Rio Preto, SP, Brasil
| | - Renata Correia Garcias
- Laboratório de Pesquisa Experimental, FACERES Faculdade de Medicina, São José do Rio Preto, SP, Brasil
| | - Giovana Queda Toledo
- Laboratório de Pesquisa Experimental, FACERES Faculdade de Medicina, São José do Rio Preto, SP, Brasil
| | - Natassia Alberici Anselmo
- Laboratório de Pesquisa Experimental, FACERES Faculdade de Medicina, São José do Rio Preto, SP, Brasil
| | | | | | - Natiele Zanardo Carvalho
- Laboratório de Pesquisa Experimental, FACERES Faculdade de Medicina, São José do Rio Preto, SP, Brasil
| | - Analice Andreoli da Silva
- Laboratório de Pesquisa Experimental, FACERES Faculdade de Medicina, São José do Rio Preto, SP, Brasil
| | - Camila Agren
- Laboratório de Pesquisa Experimental, FACERES Faculdade de Medicina, São José do Rio Preto, SP, Brasil
| | - Carla Patrícia Carlos
- Laboratório de Pesquisa Experimental, FACERES Faculdade de Medicina, São José do Rio Preto, SP, Brasil
| |
Collapse
|
14
|
Zhou Q, Lv D, Xia Y, Zhao Z, Zou H. Decreased expression of sirtuin 3 protein correlates with early stage chronic renal allograft dysfunction in a rat kidney model. Exp Ther Med 2018; 15:3725-3732. [PMID: 29581733 PMCID: PMC5863580 DOI: 10.3892/etm.2018.5909] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 12/04/2017] [Indexed: 02/06/2023] Open
Abstract
Chronic renal allograft dysfunction (CRAD) is the primary factor affecting the long-term survival of patients who have undergone renal transplantation. Oxidative stress and inflammation serve an important role in the pathological damage caused by CRAD in the early post-transplantation phase. Previous studies have demonstrated that sirtuin 3 (sirt3) protects cells from oxidative stress and inflammation. A model of renal orthotopic transplantation was established in the current study and kidney samples were harvested from the rats 12 weeks following surgery. Compared with the control groups, there were significantly increased levels of serum creatinine, blood urea nitrogen and 24 h urinary protein in the allograft group (P<0.05). Pathological examinations indicated mononuclear cell infiltration and intimal proliferation in the allograft group, which had a higher Banff score compared with the control groups. There were increased levels of malondialdehyde, decreased sirt3 protein expression and decreased superoxide dismutase enzyme activity in the allograft group compared with the control groups (P<0.05). In addition, there was a negative correlation between the expression of sirt3 and 24 h urinary protein excretion, serum creatinine levels, tubulointerstitial mononuclear cell infiltration, smooth muscle cell migration in the vascular wall and Banff scores. Thus, sirt3 may serve an important protective role in the early stage of CRAD.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Daoyuan Lv
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Yue Xia
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhihong Zhao
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Hequn Zou
- Department of Nephrology, Institute of Nephrology and Urology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
15
|
The Optimal PEG for Kidney Preservation: A Preclinical Porcine Study. Int J Mol Sci 2018; 19:ijms19020454. [PMID: 29401654 PMCID: PMC5855676 DOI: 10.3390/ijms19020454] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/26/2017] [Accepted: 01/29/2018] [Indexed: 01/12/2023] Open
Abstract
University of Wisconsin (UW) solution is not optimal for preservation of marginal organs. Polyethylene glycol (PEG) could improve protection. Similarly formulated solutions containing either 15 or 20 g/L PEG 20 kDa or 5, 15 and 30 g/L PEG 35 kDa were tested in vitro on kidney endothelial cells, ex vivo on preserved kidneys, and in vivo in a pig kidney autograft model. In vitro, all PEGs provided superior preservation than UW in terms of cell survival, adenosine triphosphate (ATP) production, and activation of survival pathways. Ex vivo, tissue injury was lower with PEG 20 kDa compared to UW or PEG 35 kDa. In vivo, function recovery was identical between UW and PEG 35 kDa groups, while PEG 20 kDa displayed swifter recovery. At three months, PEG 35 kDa 15 and 30 g/L animals had worse outcomes than UW, while 5 g/L PEG 35 kDa was similar. PEG 20 kDa was superior to both UW and PEG 35 kDa in terms of function and fibrosis development, with low activation of damage pathways. PEG 20 kDa at 15 g/L was superior to 20 g/L. While in vitro models did not discriminate between PEGs, in large animal models of transplantation we showed that PEG 20 kDa offers a higher level of protection than UW and that longer chains such as PEG 35 kDa must be used at low doses, such as found in Institut George Lopez (IGL1, 1g/L).
Collapse
|
16
|
Moore JK, Chen J, Pan H, Gaut JP, Jain S, Wickline SA. Quantification of vascular damage in acute kidney injury with fluorine magnetic resonance imaging and spectroscopy. Magn Reson Med 2017; 79:3144-3153. [PMID: 29148253 DOI: 10.1002/mrm.26985] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 12/28/2022]
Abstract
PURPOSE To design a fluorine MRI/MR spectroscopy approach to quantify renal vascular damage after ischemia-reperfusion injury, and the therapeutic response to antithrombin nanoparticles (NPs) to protect kidney function. METHODS A total of 53 rats underwent 45 min of bilateral renal artery occlusion and were treated at reperfusion with either plain perfluorocarbon NPs or NPs functionalized with a direct thrombin inhibitor (PPACK:phenyalanine-proline-arginine-chloromethylketone). Three hours after reperfusion, kidneys underwent ex vivo fluorine MRI/MR spectroscopy at 4.7 T to quantify the extent and volume of trapped NPs, as an index of vascular damage and ischemia-reperfusion injury. Microscopic evaluation of structural damage and NP trapping in non-reperfused renal segments was performed. Serum creatinine was quantified serially over 7 days. RESULTS The damaged renal cortico-medullary junction trapped a significant volume of NPs (P = 0.04), which correlated linearly (r = 0.64) with the severity of kidney injury 3 h after reperfusion. Despite global large vessel reperfusion, non-reperfusion in medullary peritubular capillaries was confirmed by MRI and microscopy, indicative of continuing hypoxia due to vascular compromise. Treatment of animals with PPACK NPs after acute kidney injury did not accelerate kidney functional recovery. CONCLUSIONS Quantification of ischemia-reperfusion injury after acute kidney injury with fluorine MRI/MR spectroscopy of perfluorocarbon NPs objectively depicts the extent and severity of vascular injury and its linear relationship to renal dysfunction. The lack of kidney function improvement after early posttreatment thrombin inhibition confirms the rapid onset of ischemia-reperfusion injury as a consequence of vascular damage and non-reperfusion. The prolongation of medullary ischemia renders cortico-medullary tubular structures susceptible to continued necrosis despite restoration of large vessel flow, which suggests limitations to acute interventions after acute kidney injury, designed to interdict renal tubular damage. Magn Reson Med 79:3144-3153, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Jeremy K Moore
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Junjie Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Hua Pan
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Joseph P Gaut
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sanjay Jain
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Samuel A Wickline
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA.,Department of Cardiovascular Science, University of South Florida, Tampa, Florida, USA.,Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
17
|
Glorion M, Polard V, Favereau F, Hauet T, Zal F, Fadel E, Sage E. Prevention of ischemia-reperfusion lung injury during static cold preservation by supplementation of standard preservation solution with HEMO 2life ® in pig lung transplantation model. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1773-1780. [PMID: 29069926 DOI: 10.1080/21691401.2017.1392315] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We describe the results of adding a new biological agent HEMO2life® to a standard preservation solution for hypothermic static lung preservation aiming to improve early functional parameters after lung transplantation. HEMO2life® is a natural oxygen carrier extracted from Arenicola marina with high oxygen affinity developed as an additive to standard organ preservation solutions. Standard preservation solution (Perfadex®) was compared with Perfadex® associated with HEMO2life® and with sham animals after 24 h of hypothermic preservation followed by lung transplantation. During five hours of lung reperfusion, functional parameters and biomarkers expression in serum and in bronchoalveolar lavage fluid (BALF) were measured. After five hours of reperfusion, HEMO2life® group led to significant improvement in functional parameters: reduction of graft vascular resistance (p < .05) and increase in graft oxygenation ratio (p < .05). Several ischemia-reperfusion related biomarkers showed positive trends in the HEMO2life® group: expression of HMG B1 in serum tended to be lower in comparison (2.1 ± 0.8 vs. 4.6 ± 1.5) with Perfadex® group, TNF-α and IL-8 in BALF were significantly higher in the two experimental groups compared to control (p < .05). During cold ischemia, expression of HIF1α and histology remained unchanged and similar to control. Supplementation of the Perfadex® solution by an innovative oxygen carrier HEMO2life® during hypothermic static preservation improves early graft function after prolonged cold ischemia in lung transplantation.
Collapse
Affiliation(s)
- M Glorion
- a Laboratoire de Chirurgie Expérimentale , Université PARIS XI, Hôpital Marie Lannelongue , Le Plessis Robinson , France.,b Department of Thoracic Surgery and Lung Transplantation , Foch Hospital , Suresnes , France
| | - V Polard
- c HEMARINA S.A. , Morlaix , France
| | - F Favereau
- d Faculté de Médecine, Université de Poitiers, INSERM U927 , Poitiers , France
| | - T Hauet
- d Faculté de Médecine, Université de Poitiers, INSERM U927 , Poitiers , France
| | - F Zal
- c HEMARINA S.A. , Morlaix , France
| | - E Fadel
- a Laboratoire de Chirurgie Expérimentale , Université PARIS XI, Hôpital Marie Lannelongue , Le Plessis Robinson , France
| | - E Sage
- a Laboratoire de Chirurgie Expérimentale , Université PARIS XI, Hôpital Marie Lannelongue , Le Plessis Robinson , France.,b Department of Thoracic Surgery and Lung Transplantation , Foch Hospital , Suresnes , France
| |
Collapse
|
18
|
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
|
19
|
Abstract
Despite advances in interventional and pharmacologic therapy, survivors of myocardial infarction remain at an increased risk of subsequent cardiovascular events. Initial pharmacological management includes both platelet inhibition and parenteral anticoagulation, whereas long-term pharmacological therapy relies on antiplatelet therapy for prevention of thrombotic complications. Biomarkers showing ongoing thrombin generation after acute coronary syndromes suggest that anticoagulants may provide additional benefit in reducing cardiovascular events. We review the pharmacokinetics of novel anticoagulants, clinical trial results, the role of monitoring, and future directions for the use of novel oral anticoagulants in the treatment of coronary artery disease. Clinical trials have shown that long-term use of oral anticoagulants decreases the risk of cardiovascular events, but they do so at a cost of an increased risk of bleeding. Future studies will need to identify optimal treatment combinations for selected patients and conditions that address both the appropriate combination of therapy and the appropriate dosage of each agent when used in combination.
Collapse
|
20
|
Melis N, Rubera I, Cougnon M, Giraud S, Mograbi B, Belaid A, Pisani DF, Huber SM, Lacas-Gervais S, Fragaki K, Blondeau N, Vigne P, Frelin C, Hauet T, Duranton C, Tauc M. Targeting eIF5A Hypusination Prevents Anoxic Cell Death through Mitochondrial Silencing and Improves Kidney Transplant Outcome. J Am Soc Nephrol 2017; 28:811-822. [PMID: 27612998 PMCID: PMC5328152 DOI: 10.1681/asn.2016010012] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/20/2016] [Indexed: 11/03/2022] Open
Abstract
The eukaryotic initiation factor 5A (eIF5A), which is highly conserved throughout evolution, has the unique characteristic of post-translational activation through hypusination. This modification is catalyzed by two enzymatic steps involving deoxyhypusine synthase (DHPS) and deoxyhypusine hydroxylase (DOHH). Notably, eIF5A may be involved in regulating the lifespan of Drosophila during long-term hypoxia. Therefore, we investigated the possibility of a link between eIF5A hypusination and cellular resistance to hypoxia/anoxia. Pharmacologic targeting of DHPS by N1-guanyl-1,7-diaminoheptane (GC7) or RNA interference-mediated inhibition of DHPS or DOHH induced tolerance to anoxia in immortalized mouse renal proximal cells. Furthermore, GC7 treatment of cells reversibly induced a metabolic shift toward glycolysis as well as mitochondrial remodeling and led to downregulated expression and activity of respiratory chain complexes, features characteristic of mitochondrial silencing. GC7 treatment also attenuated anoxia-induced generation of reactive oxygen species in these cells and in normoxic conditions, decreased the mitochondrial oxygen consumption rate of cultured cells and mice. In rats, intraperitoneal injection of GC7 substantially reduced renal levels of hypusinated eIF5A and protected against ischemia-reperfusion-induced renal injury. Finally, in the preclinical pig kidney transplant model, intravenous injection of GC7 before kidney removal significantly improved graft function recovery and late graft function and reduced interstitial fibrosis after transplant. This unconventional signaling pathway offers an innovative therapeutic target for treating hypoxic-ischemic human diseases and organ transplantation.
Collapse
Affiliation(s)
- Nicolas Melis
- Laboratoire de Physio-Médecine Moléculaire, Centre National de la Recherche Scientifique-UMR7370
| | - Isabelle Rubera
- Laboratoire de Physio-Médecine Moléculaire, Centre National de la Recherche Scientifique-UMR7370
| | - Marc Cougnon
- Laboratoire de Physio-Médecine Moléculaire, Centre National de la Recherche Scientifique-UMR7370
| | - Sébastien Giraud
- Centre Hospitalo Universitaire Poitiers, Service de Biochimie, Poitiers, France
- Institut National de la Santé et de la Recherche Médicale U1082 Ischémie Reperfusion en Transplantation d'Organes Mécanismes et Innovations Thérapeutiques, Poitiers, France
- Faculté de Médecine et de Pharmacie, Université de Poitiers, Poitiers, France; and
| | - Baharia Mograbi
- Institut de Recherche sur le Cancer, Centre National de la Recherche Scientifique-UMR7284, Institut National de la Santé et de la Recherche Médicale U1081
| | - Amine Belaid
- Institut de Recherche sur le Cancer, Centre National de la Recherche Scientifique-UMR7284, Institut National de la Santé et de la Recherche Médicale U1081
| | - Didier F Pisani
- Institute of Biology Valrose, Centre National de la Recherche Scientifique-UMR7277 Institut National de la Santé et de la Recherche Médicale U1091
| | - Stephan M Huber
- Department of Radiation Oncology, University of Tübingen, Tuebingen, Germany
| | | | - Konstantina Fragaki
- Institut de Recherche sur le Cancer, Centre National de la Recherche Scientifique-UMR7284, Institut National de la Santé et de la Recherche Médicale U1081
| | - Nicolas Blondeau
- Institut de Physiologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique Unité Mixte de Recherche UMR7275, University Nice-Sophia Antipolis, Nice, France
| | - Paul Vigne
- Institute of Biology Valrose, Centre National de la Recherche Scientifique-UMR7277 Institut National de la Santé et de la Recherche Médicale U1091
| | - Christian Frelin
- Institute of Biology Valrose, Centre National de la Recherche Scientifique-UMR7277 Institut National de la Santé et de la Recherche Médicale U1091
| | - Thierry Hauet
- Centre Hospitalo Universitaire Poitiers, Service de Biochimie, Poitiers, France
- Institut National de la Santé et de la Recherche Médicale U1082 Ischémie Reperfusion en Transplantation d'Organes Mécanismes et Innovations Thérapeutiques, Poitiers, France
- Faculté de Médecine et de Pharmacie, Université de Poitiers, Poitiers, France; and
| | - Christophe Duranton
- Laboratoire de Physio-Médecine Moléculaire, Centre National de la Recherche Scientifique-UMR7370
| | - Michel Tauc
- Laboratoire de Physio-Médecine Moléculaire, Centre National de la Recherche Scientifique-UMR7370,
| |
Collapse
|
21
|
Tillet S, Giraud S, Kerforne T, Saint-Yves T, Joffrion S, Goujon JM, Cau J, Mauco G, Petitou M, Hauet T. Inhibition of coagulation proteases Xa and IIa decreases ischemia-reperfusion injuries in a preclinical renal transplantation model. Transl Res 2016; 178:95-106.e1. [PMID: 27513209 DOI: 10.1016/j.trsl.2016.07.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 07/13/2016] [Accepted: 07/15/2016] [Indexed: 11/30/2022]
Abstract
Coagulation is an important pathway in the pathophysiology of ischemia-reperfusion injuries. In particular, deceased after circulatory death (DCD) donors undergo a no-flow period, a strong activator of coagulation. Hence, therapies influencing the coagulation cascade must be developed. We evaluated the effect of a new highly specific and effective anti-Xa/IIa molecule, with an integrated innovative antidote site (EP217609), in a porcine preclinical model mimicking injuries observed in DCD donor kidney transplantation. Kidneys were clamped for 60 minutes (warm ischemia), then flushed and preserved for 24 hours at 4°C in University of Wisconsin (UW) solution (supplemented or not). EP217609-supplemented UW solution (UW-EP), compared with unfractionated heparin-supplemented UW solution (UW-UFH) or UW alone (UW). A mechanistic investigation was conducted in vitro: addition of EP217609 to endothelial cells during hypoxia at 4°C in the UW solution inhibited thrombin generation during reoxygenation at 37°C in human plasma and reduced tumor necrosis factor alpha, intercellular adhesion molecule 1, and vascular cell adhesion molecule 1 messenger RNA cell expressions. In vivo, function recovery was markedly improved in the UW-EP group. Interestingly, levels of thrombin-antithrombin complexes (reflecting thrombin generation) were reduced 60 minutes after reperfusion in the UW-EP group. In addition, 3 months after transplantation, lower fibrosis, epithelial-mesenchymal transition, inflammation, and leukocyte infiltration were observed. Using this new dual anticoagulant, anti-Xa/IIa activity during kidney flush and preservation is protected by reducing thrombin generation at revascularization, improving early function recovery, and decreasing chronic lesions. Such an easy-to-deploy clinical strategy could improve marginal graft outcome.
Collapse
Affiliation(s)
- Solenne Tillet
- Inserm U1082 IRTOMIT, Poitiers, France; Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France
| | - Sébastien Giraud
- Inserm U1082 IRTOMIT, Poitiers, France; Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France; CHU Poitiers, Service de Biochimie, Poitiers, France
| | - Thomas Kerforne
- Inserm U1082 IRTOMIT, Poitiers, France; Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France; CHU Poitiers, Département d'Anesthésie-Réanimation, Poitiers, France
| | - Thibaut Saint-Yves
- Inserm U1082 IRTOMIT, Poitiers, France; CH d'Angoulème, Service de Chirurgie Urologie, Angoulème, France; CHU de Poitiers, Service d'Urologie, Pôle DUNE, Poitiers, France
| | - Sandrine Joffrion
- Inserm U1082 IRTOMIT, Poitiers, France; CHU Poitiers, Service de Biochimie, Poitiers, France
| | - Jean-Michel Goujon
- Inserm U1082 IRTOMIT, Poitiers, France; Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France; CHU de Poitiers, Service d'Anapathomopathologie, Poitiers, France
| | | | - Gérard Mauco
- Inserm U1082 IRTOMIT, Poitiers, France; Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France; CHU Poitiers, Service de Biochimie, Poitiers, France
| | | | - Thierry Hauet
- Inserm U1082 IRTOMIT, Poitiers, France; Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers, France; CHU Poitiers, Service de Biochimie, Poitiers, France; IBiSA Plateforme 'plate-forme MOdélisation Préclinique-Innovation Chirurgicale et Technologique (MOPICT), INRA Domaine Expérimental du Magneraud, Surgères, France; FHU SUPORT 'SUrvival oPtimization in ORgan Transplantation', Poitiers, France.
| |
Collapse
|
22
|
Kerforne T, Favreau F, Thuillier R, Hauet T, Pinsard M. [Toward a customized preservation for each kidney graft?]. Nephrol Ther 2016; 12:437-442. [PMID: 27720135 DOI: 10.1016/j.nephro.2016.02.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 01/24/2023]
Abstract
The increased number of patients in waiting list for renal transplantation requires the establishment of recommendations regarding graft preservation techniques. The preservation method impacts graft function and survival particularly in case of extended criteria donors. Based on our experience, the aim of this review is to establish a decisional diagram to draw graft management to 5years in relation to donor type and graft quality. Novel biomarkers are necessary to evaluate graft quality. Nuclear magnetic resonance or transcriptomic analyses are promising. Thus, good quality organs will be preserved in static condition associated to hypothermia; while grafts from extended criteria donors need to be assessed early during dynamic perfusion through an evaluation of perfusion solution to discriminate: good organs, with acceptable risks without perfusion conditions modifications; tolerable risk grafts for which it will be recommended to use a supplementation of perfusion solution with oxygen or pharmacologic additives such as mitochondrion protectors or oxygen carriers; and elevated risks graft which will not be used. This diagram based on experimental data needs to be assessed in clinical trials but highlights the crucial role of kidney graft quality assessment for its management and placed dynamic perfusion preservation as the protocol of choice for extended criteria donors.
Collapse
Affiliation(s)
- Thomas Kerforne
- Inserm, U1082, IRTOMIT, ischémie-reperfusion en transplantation d'organe, mécanismes et innovations thérapeutiques, faculté de médecine et de pharmacie de Poitiers, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France; Service de réanimation chirurgicale, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France
| | - Frédéric Favreau
- Inserm, U1082, IRTOMIT, ischémie-reperfusion en transplantation d'organe, mécanismes et innovations thérapeutiques, faculté de médecine et de pharmacie de Poitiers, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France; Laboratoire de biochimie, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France
| | - Raphaël Thuillier
- Inserm, U1082, IRTOMIT, ischémie-reperfusion en transplantation d'organe, mécanismes et innovations thérapeutiques, faculté de médecine et de pharmacie de Poitiers, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France; Laboratoire de biochimie, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France
| | - Thierry Hauet
- Inserm, U1082, IRTOMIT, ischémie-reperfusion en transplantation d'organe, mécanismes et innovations thérapeutiques, faculté de médecine et de pharmacie de Poitiers, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France; Laboratoire de biochimie, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France; Inra, UE1372 GenESI, plateforme labellisée IBiSA MOPICT, BP 52, Saint-Pierre d'Amilly, 17700 Surgères, France; Fédération hospitalo-universitaire suport, CS90577, 86021 Poitiers, France.
| | - Michel Pinsard
- Inserm, U1082, IRTOMIT, ischémie-reperfusion en transplantation d'organe, mécanismes et innovations thérapeutiques, faculté de médecine et de pharmacie de Poitiers, université de Poitiers, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France; Service de réanimation chirurgicale, CHU de Poitiers, 2, rue de la Milétrie, CS90577, 86021 Poitiers, France
| |
Collapse
|
23
|
Delpech PO, Thuillier R, SaintYves T, Danion J, Le Pape S, van Amersfoort ES, Oortwijn B, Blancho G, Hauet T. Inhibition of complement improves graft outcome in a pig model of kidney autotransplantation. J Transl Med 2016; 14:277. [PMID: 27663514 PMCID: PMC5035455 DOI: 10.1186/s12967-016-1013-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/16/2016] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Ischemia reperfusion injury (IRI) induced immune response is a critical issue in transplantation. Complement and contact system activation are among its key mechanisms. STUDY DESIGN We investigated the benefits of pre-reperfusion treatment with recombinant human C1INH (rhC1INH), inhibitor of both complement and contact activation, in a pig model of kidney autotransplantation, subjecting the organ to 60 min warm ischemia prior to 24 h static preservation to maximize damage. RESULTS Serum creatinine measurement showed that treated animals recovered glomerular function quicker than the Vehicle group. However, no difference was observed in tubular function recovery, and elevated level of urinary NGal (Neutrophil gelatinase-associated lipocalin) and plasma AST (Aspartate Aminotransferase) were detected, indicating that treatment did not influence IRI-mediated tubular cell necrosis. Regarding chronic graft outcome, rhC1INH significantly prevented fibrosis development and improved function. Immunohistochemistry and western blot showed decreased invasion by macrophages and T lymphocytes, and reduction of epithelial to mesenchymal transition. We determined the effect of treatment on complement activation with immunofluorescence analyses at 30 min post reperfusion, showing an inhibition of C4d deposition and MBL staining in treated animals. CONCLUSIONS In this model, the inhibition of complement activation by rhC1INH at reperfusion, while not completely counteracting IRI, limited immune system activation, significantly improving graft outcome on the short and long term.
Collapse
Affiliation(s)
- Pierre-Olivier Delpech
- Département d'Urologie, CHU de Poitiers, 86000, Poitiers, France.,Inserm U1082, 86000, Poitiers, France
| | - Raphael Thuillier
- Inserm U1082, 86000, Poitiers, France.,Service de Biochimie, CHU Poitiers, 86000, Poitiers, France.,Faculté de Médecine et de Pharmacie, Université de Poitiers, 86000, Poitiers, France.,Fédération Hospitalo-Universitaire SUPORT, 86000, Poitiers, France
| | | | - Jerome Danion
- Service de Chirurgie Viscérale, CHU de Poitiers, 86000, Poitiers, France
| | - Sylvain Le Pape
- Inserm U1082, 86000, Poitiers, France.,Faculté de Médecine et de Pharmacie, Université de Poitiers, 86000, Poitiers, France
| | | | | | - Gilles Blancho
- Institut de Transplantation Urologie et Néphrologie (ITUN), CHU de Nantes, Faculté de Médecine et des Techniques Médicales de Nantes, Université de Nantes, Inserm U1064, 44000, Nantes, France
| | - Thierry Hauet
- Inserm U1082, 86000, Poitiers, France. .,Service de Biochimie, CHU Poitiers, 86000, Poitiers, France. .,Faculté de Médecine et de Pharmacie, Université de Poitiers, 86000, Poitiers, France. .,Fédération Hospitalo-Universitaire SUPORT, 86000, Poitiers, France. .,Institut National de La Recherche Agronomique, Unité Expérimentale Génétique, Expérimentations et Systèmes Innovants, Domaine Expérimental Du Magneraud, Plateforme IBiSA 'MOPICT', 17700, Surgères, France. .,INSERM U1082, CHU de Poitiers, 2 Rue de La Miletrie, 86021, Poitiers Cedex, France.
| |
Collapse
|
24
|
Yu ZX, Qi S, Lasaro MA, Bouchard K, Dow C, Moore K, Wu Z, Barama A, Xu J, Johnson K, Marozsan AJ, Wang Y. Targeting Complement Pathways During Cold Ischemia and Reperfusion Prevents Delayed Graft Function. Am J Transplant 2016; 16:2589-97. [PMID: 27003920 DOI: 10.1111/ajt.13797] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/11/2016] [Indexed: 01/25/2023]
Abstract
The complement system plays a critical role in ischemia-reperfusion injury (IRI)-mediated delayed graft function (DGF). To better understand the roles of complement activation pathways in IRI in kidney transplantation, donor kidneys were treated ex vivo with terminal complement pathway (TP) inhibitor, anti-rat C5 mAb 18A10, or complement alternative pathway (AP) inhibitor TT30 for 28 h at 4°C pretransplantation in a syngeneic kidney transplantation rat model. All 18A10- and 67% of TT30-pretreated grafts, but only 16.7% of isotype control-pretreated grafts, survived beyond day 21 (p < 0.01). Inhibitor treatment in the final 45 min of 28-h cold ischemia (CI) similarly improved graft survival. Systemic posttransplant treatment with 18A10 resulted in 60% increased graft survival beyond day 21 (p < 0.01), while no TT30-treated rat survived > 6 days. Our results demonstrate that AP plays a prominent role during CI and that blocking either the AP or, more effectively the TP prevents ischemic injury and subsequent DGF. Multiple complement pathways may be activated and contribute to reperfusion injury; blocking the TP, but not the AP, posttransplant is effective in preventing reperfusion injury and increasing graft survival. These results demonstrate the feasibility of using complement inhibitors for prevention of DGF in humans.
Collapse
Affiliation(s)
- Z X Yu
- Pre-Clinical Sciences, Alexion Pharmaceuticals, Inc., Cheshire, CT, Canada
| | - S Qi
- Research Centre, Centre Hospitalier de l'Université de Montréal, Department of Surgery, Hôpital Notre Dame and Université de Montréal, Montréal, Quebec, Canada
| | - M A Lasaro
- Pre-Clinical Sciences, Alexion Pharmaceuticals, Inc., Cheshire, CT, Canada
| | - K Bouchard
- Pre-Clinical Sciences, Alexion Pharmaceuticals, Inc., Cheshire, CT, Canada
| | - C Dow
- Pre-Clinical Sciences, Alexion Pharmaceuticals, Inc., Cheshire, CT, Canada
| | - K Moore
- Pre-Clinical Sciences, Alexion Pharmaceuticals, Inc., Cheshire, CT, Canada
| | - Z Wu
- Research Centre, Centre Hospitalier de l'Université de Montréal, Department of Surgery, Hôpital Notre Dame and Université de Montréal, Montréal, Quebec, Canada
| | - A Barama
- Research Centre, Centre Hospitalier de l'Université de Montréal, Department of Surgery, Hôpital Notre Dame and Université de Montréal, Montréal, Quebec, Canada
| | - J Xu
- Research Centre, Centre Hospitalier de l'Université de Montréal, Department of Surgery, Hôpital Notre Dame and Université de Montréal, Montréal, Quebec, Canada
| | - K Johnson
- Pre-Clinical Sciences, Alexion Pharmaceuticals, Inc., Cheshire, CT, Canada
| | - A J Marozsan
- Pre-Clinical Sciences, Alexion Pharmaceuticals, Inc., Cheshire, CT, Canada
| | - Y Wang
- Pre-Clinical Sciences, Alexion Pharmaceuticals, Inc., Cheshire, CT, Canada
| |
Collapse
|
25
|
Coagulation Factors in the Interstitial Space. Protein Sci 2016. [DOI: 10.1201/9781315374307-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
26
|
de Ridder GG, Lundblad RL, Pizzo SV. Actions of thrombin in the interstitium. J Thromb Haemost 2016; 14:40-7. [PMID: 26564405 DOI: 10.1111/jth.13191] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Indexed: 01/14/2023]
Abstract
Thrombin is a pleiotropic enzyme best known for its contribution to fibrin formation and platelet aggregation during vascular hemostasis. There is increasing evidence to suggest a role for thrombin in the development of interstitial fibrosis, but interstitial thrombin has not been demonstrated by the direct determination of activity. Rather its presence is inferred by products of thrombin action such as fibrin and activated fibroblasts. This review will focus on possible mechanisms of thrombin formation in the interstitial space, the possible actions of thrombin, processes regulating thrombin activity in the interstitial space, and evidence supporting a role for thrombin in fibrosis.
Collapse
Affiliation(s)
- G G de Ridder
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - R L Lundblad
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
| | - S V Pizzo
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
27
|
Woodside KJ, Goldfarb DA, Rabets JC, Sanchez EQ, Lebovitz DJ, Schulak JA, Fung JJ, Eghtesad B. Enhancing kidney function with thrombolytic therapy following donation after cardiac death: a multicenter quasi-blinded prospective randomized trial. Clin Transplant 2015; 29:1173-80. [PMID: 26448622 DOI: 10.1111/ctr.12647] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2015] [Indexed: 12/21/2022]
Abstract
Kidneys from donors after cardiac death (DCD) are at risk for inferior outcomes, possibly due to microthrombi and additional warm ischemia. We describe an organ procurement organization-wide trial utilizing thrombolytic tissue plasminogen activator (tPA) during machine pulsatile perfusion (MPP). A kidney from each recovered kidney pair was prospectively randomized to receive tPA (50 mg Alteplase) or no tPA (control) in the MPP perfusate. From 2011 to 2013, 24 kidneys were placed with enrolled recipients from 19 DCD kidney donors. There were no significant differences for absolute values of flow or resistance while undergoing MPP between the groups, nor rates of achieving discrete flow and resistance targets. While there was a trend toward lower creatinine and higher glomerular filtration rates in the tPA group at 3, 6, 9, and 12 months, these differences were not significant. Delayed graft function (DGF) rates were 41.7% in the tPA group vs. 58.4% in the control group (OR 0.51, 95%CI 0.10-2.59, p = 0.68). Death-censored graft survival was similar between the groups. In this pilot study, encouraging trends are seen in kidney allograft function independent of MPP parameters following DCD kidney transplantation for those kidneys receiving thrombolytic tPA and MPP, compared with standard MPP.
Collapse
Affiliation(s)
- Kenneth J Woodside
- Transplant Institute, University Hospitals Case Medical Center, Cleveland, OH, USA
| | | | - John C Rabets
- Transplant Center, Cleveland Clinic, Cleveland, OH, USA
| | - Edmund Q Sanchez
- Transplant Institute, University Hospitals Case Medical Center, Cleveland, OH, USA
| | - Daniel J Lebovitz
- Lifebanc, Cleveland, OH, USA.,Critical Care, Akron Children's Hospital, Akron, OH, USA
| | - James A Schulak
- Transplant Institute, University Hospitals Case Medical Center, Cleveland, OH, USA
| | - John J Fung
- Transplant Center, Cleveland Clinic, Cleveland, OH, USA
| | | |
Collapse
|
28
|
|
29
|
Strategies to optimize kidney recovery and preservation in transplantation: specific aspects in pediatric transplantation. Pediatr Nephrol 2015; 30:1243-54. [PMID: 25185880 DOI: 10.1007/s00467-014-2924-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 07/09/2014] [Accepted: 07/21/2014] [Indexed: 01/08/2023]
Abstract
In renal transplantation, live donor kidney grafts are associated with optimum success rates due to the shorter period of ischemia during the surgical procedure. The current shortage of donor organs for adult patients has caused a shift towards deceased donors, often with co-morbidity factors, whose organs are more sensitive to ischemia-reperfusion injury, which is unavoidable during transplantation. Donor management is pivotal to kidney graft survival through the control of the ischemia-reperfusion sequence, which is known to stimulate numerous deleterious or regenerative pathways. Although the key role of endothelial cells has been established, the complexity of the injury, associated with stimulation of different cell signaling pathways, such as unfolded protein response and cell death, prevents the definition of a unique therapeutic target. Preclinical transplant models in large animals are necessary to establish relationships and kinetics and have already contributed to the improvement of organ preservation. Therapeutic strategies using mesenchymal stem cells to induce allograft tolerance are promising advances in the treatment of the pediatric recipient in terms of reducing/withdrawing immunosuppressive therapy. In this review we focus on the different donor management strategies in kidney graft conditioning and on graft preservation consequences by highlighting the role of endothelial cells. We also propose strategies for preventing ischemia-reperfusion, such as cell therapy.
Collapse
|
30
|
Pontrelli P, Cariello M, Rascio F, Gigante M, Verrienti R, Tataranni T, Castellano G, Divella C, Ranieri E, Stallone G, Gesualdo L, Grandaliano G. Thrombin may modulate dendritic cell activation in kidney transplant recipients with delayed graft function. Nephrol Dial Transplant 2015; 30:1480-7. [DOI: 10.1093/ndt/gfv129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/08/2015] [Indexed: 02/02/2023] Open
|
31
|
Abstract
As the impact of ischemia reperfusion injury on graft outcome is now well defined, efforts are made towards decreasing these lesions, typically through the improvement of preservation techniques. The use of pharmacological supplements which could be compatible with any preservation solution used by the transplant center and target specific pathways of IR is an interesting strategy to improve graft quality. However, the extensive number of studies showing the benefits a molecule in an animal model of IR without thorough mechanistic determination of the effects of this agent make it difficult to opt for specific pharmaceutical intervention. Herein we expose studies which demonstrate the benefits of several molecules relying on a thorough mechanical analysis of the events occurring during preservation, both at the cellular and the systemic levels. We believe this approach is the most appropriate to truly understand the potential benefits of a molecule and particularly to design a comprehensive pharmaceutical regiment, with several agents acting synergistically against IR, to improve organ preservation and graft outcome.
Collapse
Affiliation(s)
- T Saint Yves
- Inserm U1082, université de Poitiers, CHU La Miletrie, 86000 Poitiers, France; Service d'Urologie, CHU La Miletrie, 86000 Poitiers, France
| | - P-O Delpech
- Inserm U1082, université de Poitiers, CHU La Miletrie, 86000 Poitiers, France; Service d'Urologie, CHU La Miletrie, 86000 Poitiers, France
| | - S Giraud
- Inserm U1082, université de Poitiers, CHU La Miletrie, 86000 Poitiers, France
| | - R Thuillier
- Inserm U1082, université de Poitiers, CHU La Miletrie, 86000 Poitiers, France.
| | - T Hauet
- Inserm U1082, université de Poitiers, CHU La Miletrie, 86000 Poitiers, France
| |
Collapse
|
32
|
Abstract
Ischemia reperfusion injury occurs in the kidney when blood supply is interrupted in clinical settings such as kidney transplantation or nephron sparing surgery for renal tumors. These lesions lead to acute kidney injury (AKI) a detrimental situation associated with impaired short-term allograft function (delayed graft function or primary non function) but also long-term transplant survival through the onset of chronic allograft nephropathy. The present review details the cellular and molecular consequences of ischemia reperfusion in a native kidney as well as in a kidney graft after cold ischemia time, giving a comprehensive description of biological pathways involved during the phase of ischemia and during the reperfusion period where the rapid return to normoxia leads to a large burst of reactive oxygen species along with a dramatic reduction in antioxidant defenses. This work also focuses on the distinct susceptibilities of kidney cells to ischemia (endothelial vs epithelial) and the outcome of acute kidney injury.
Collapse
|
33
|
Chen J, Vemuri C, Palekar RU, Gaut JP, Goette M, Hu L, Cui G, Zhang H, Wickline SA. Antithrombin nanoparticles improve kidney reperfusion and protect kidney function after ischemia-reperfusion injury. Am J Physiol Renal Physiol 2015; 308:F765-73. [PMID: 25651565 DOI: 10.1152/ajprenal.00457.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 01/20/2015] [Indexed: 01/24/2023] Open
Abstract
In the extension phase of acute kidney injury, microvascular thrombosis, inflammation, vasoconstriction, and vascular endothelial cell dysfunction promote progressive damage to renal parenchyma after reperfusion. In this study, we hypothesized that direct targeting and pharmaceutical knockdown of activated thrombin at the sites of injury with a selective nanoparticle (NP)-based thrombin inhibitor, PPACK (phenylalanine-proline-arginine-chloromethylketone), would improve kidney reperfusion and protect renal function after transient warm ischemia in rodent models. Saline- or plain NP-treated animals were employed as controls. In vivo 19F magnetic resonance imaging revealed that kidney nonreperfusion was evident within 3 h after global kidney reperfusion at 34 ± 13% area in the saline group and 43 ± 12% area in the plain NP group and substantially reduced to 17 ± 4% (∼50% decrease, P < 0.05) in the PPACK NP pretreatment group. PPACK NP pretreatment prevented an increase in serum creatinine concentration within 24 h after ischemia-reperfusion, reflecting preserved renal function. Histologic analysis illustrated substantially reduced intrarenal thrombin accumulation within 24 h after reperfusion for PPACK NP-treated kidneys (0.11% ± 0.06%) compared with saline-treated kidneys (0.58 ± 0.37%). These results suggest a direct role for thrombin in the pathophysiology of AKI and a nanomedicine-based preventative strategy for improving kidney reperfusion after transient warm ischemia.
Collapse
Affiliation(s)
- Junjie Chen
- Department of Medicine, Washington University, St. Louis, Missouri
| | - Chandu Vemuri
- Department of Medicine, Washington University, St. Louis, Missouri; Department of Surgery, Washington University, St. Louis, Missouri
| | - Rohun U Palekar
- Department of Medicine, Washington University, St. Louis, Missouri; Department of Biomedical Engineering, Washington University, St. Louis, Missouri; and
| | - Joseph P Gaut
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri
| | - Matthew Goette
- Department of Medicine, Washington University, St. Louis, Missouri; Department of Biomedical Engineering, Washington University, St. Louis, Missouri; and
| | - Lingzhi Hu
- Department of Medicine, Washington University, St. Louis, Missouri
| | - Grace Cui
- Department of Medicine, Washington University, St. Louis, Missouri
| | - Huiying Zhang
- Department of Medicine, Washington University, St. Louis, Missouri
| | - Samuel A Wickline
- Department of Medicine, Washington University, St. Louis, Missouri; Department of Biomedical Engineering, Washington University, St. Louis, Missouri; and
| |
Collapse
|
34
|
Tillet S, Giraud S, Delpech PO, Thuillier R, Ameteau V, Goujon JM, Renelier B, Macchi L, Hauet T, Mauco G. Kidney graft outcome using an anti-Xa therapeutic strategy in an experimental model of severe ischaemia-reperfusion injury. Br J Surg 2014; 102:132-42; discussion 142. [PMID: 25402331 DOI: 10.1002/bjs.9662] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/04/2014] [Accepted: 08/28/2014] [Indexed: 12/15/2022]
Abstract
BACKGROUND Deceased after cardiac death donors represent an important source of organs to reduce organ shortage in transplantation. However, these organs are subjected to more ischaemia-reperfusion injury (IRI). Reducing IRI by targeting coagulation is studied here in an experimental model. METHODS The effect of an anti-Xa compound (fondaparinux) was evaluated using an autotransplanted kidney model in pigs. Kidneys were clamped for 60 min (warm ischaemia) and then preserved for 24 h at 4 °C in University of Wisconsin solution (UW). The anti-Xa compound was injected intravenously before warm ischaemia and used during cold storage, and its effects were compared with those of intravenous injection of unfractionated heparin (UFH) before warm ischaemia and use during cold storage, or use of UW alone during cold storage. RESULTS At 3 months after transplantation, anti-Xa treatment improved recovery of renal function and chronic serum creatinine levels compared with UW and UFH (mean(s.e.m.) 89(4), 250(4) and 217(8) µmol/l respectively). The anti-Xa treatment also reduced fibrosis, and decreased tissue expression of markers of the epithelial-mesenchymal transition compared with UW and UFH. Cleaved protease-activated receptor 2 was overexpressed in the UW group compared with the anti-Xa and UFH groups. Leucocyte infiltrates were decreased in the anti-Xa group compared with the UW and UFH groups. Macrophage invasion was also decreased by anticoagulation treatment. CONCLUSION Peritransplant anticoagulation therapy was beneficial to graft outcome, in both the acute and chronic phases. Moreover, specific inhibition of coagulation Xa protease further protected kidney grafts, with better recovery and decreased expression of chronic lesion markers. Surgical relevance The increasing use of marginal donors highlights the importance of organ quality in transplantation. Renal ischaemia-reperfusion injury (IRI), which includes a deleterious activation of coagulation, plays a central role in determining graft quality and outcome. Using an established porcine renal autotransplantation preclinical model with high clinical relevance, the benefits of anticoagulation therapy using an antifactor Xa molecule were evaluated. Peritransplantion anticoagulation treatment, specifically with an anti-Xa compound, protected marginal kidney grafts, improving functional recovery and reducing chronic lesions. This study demonstrates the benefits of anticoagulation therapy at the time of organ collection, particularly for marginal organs, encountered in cases of extended criteria and deceased after circulatory death donors. This anticoagulation strategy could be an important addition to current donor and organ management protocols in order to limit IRI and improve outcome.
Collapse
Affiliation(s)
- S Tillet
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1082, Poitiers, France; Faculté de Médecine et de Pharmacie, Université de Poitiers, Poitiers, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Delpech PO, Thuillier R, Le Pape S, Rossard L, Jayle C, Billault C, Goujon JM, Hauet T. Effects of warm ischaemia combined with cold preservation on the hypoxia-inducible factor 1α pathway in an experimental renal autotransplantation model. Br J Surg 2014; 101:1739-50. [DOI: 10.1002/bjs.9611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/12/2013] [Accepted: 06/10/2014] [Indexed: 01/12/2023]
Abstract
Abstract
Background
The increased use of marginal donors highlights the importance of organ quality in transplantation and the identification of prognostic biomarkers. This experimental study investigated modulation of the hypoxia-inducible factor (HIF) 1α pathway in kidney grafts in relation to different degrees of ischaemia.
Methods
In a porcine autotransplantation model, two different kidney graft protocols were compared: standard 24-h cold storage (CS) and 24-h CS preceded by 1 h warm ischaemia (WI + CS). The renal HIF-1α pathway and tubular dedifferentiation were analysed in the early phase of reperfusion and at 3 months.
Results
There was a relationship between the degree of ischaemic injury and the outcome of the kidney graft. During the first week of reperfusion, WI + CS grafts showed a higher degree of injury. The observed tubular dedifferentiation was associated with delayed HIF-1α expression, and with loss of its role in transcription. In highly injured kidneys, deregulation of the HIF-1α pathway was also observed in the chronic phase, with reduced production of vascular endothelial growth factor (VEGF) A, and upregulation of VEGF receptor 1 (Flt-1) and thrombospondin 1. In addition, these kidneys displayed altered kidney histology and decreased function.
Conclusion
The HIF-1α pathway appears to be abolished early in response to severe ischaemia. A high degree of ischaemic injury also results in chronic activation of the HIF-1α pathway, diverting it away from the beneficial activation of angiogenesis. Further studies on the finely tuned balance of signals in this pathway may provide diagnostic biomarkers that can determine organ quality during kidney transplantation. Surgical relevanceThe increased use of marginal donors has highlighted the importance of organ quality in transplantation. Renal ischaemia–reperfusion injury following transplantation induces graft dysfunction.In a porcine model of renal autotransplantation, the induction of regenerative processes, in response to graded degrees of ischaemia, was studied in the post-transplantation phase. There was early abrogation of the hypoxia-inducible factor (HIF) 1α pathway in response to severe ischaemia. High degrees of ischaemic injury induced chronic activation of the HIF-1α pathway, diverting it from the beneficial activation of angiogenesis.Identification of the mechanisms involved in renal regeneration, such as those related to the HIF-1α pathway, are important as these mechanisms can be used to identify novel therapeutic targets or develop diagnostic biomarkers to determine organ quality early in the transplantation process.
Collapse
Affiliation(s)
- P O Delpech
- Faculty of Medicine and Pharmacy, University of Poitiers, France
| | - R Thuillier
- Faculty of Medicine and Pharmacy, University of Poitiers, France
- Department of Biochemistry, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - S Le Pape
- Faculty of Medicine and Pharmacy, University of Poitiers, France
| | - L Rossard
- Faculty of Medicine and Pharmacy, University of Poitiers, France
| | - C Jayle
- Faculty of Medicine and Pharmacy, University of Poitiers, France
| | - C Billault
- Faculty of Medicine and Pharmacy, University of Poitiers, France
| | - J M Goujon
- Faculty of Medicine and Pharmacy, University of Poitiers, France
- Department of Pathology, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - T Hauet
- Faculty of Medicine and Pharmacy, University of Poitiers, France
- Department of Biochemistry, Centre Hospitalier Universitaire de Poitiers, Poitiers, France
- National Institute for Agricultural Research (INRA), UE1372 GenESI, Platform IBiSA, Domaine Expérimental du Magneraud, Surgères, France
- University/Hospital Federation (SUPORT), Limoges, Poitiers and Tours, France
| |
Collapse
|
36
|
Thuillier R, Allain G, Giraud S, Saintyves T, Delpech PO, Couturier P, Billault C, Marchand E, Vaahtera L, Parkkinen J, Hauet T. Cyclodextrin curcumin formulation improves outcome in a preclinical pig model of marginal kidney transplantation. Am J Transplant 2014; 14:1073-83. [PMID: 24618351 DOI: 10.1111/ajt.12661] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 12/27/2013] [Accepted: 12/29/2013] [Indexed: 01/25/2023]
Abstract
Decreasing organ quality is prompting research toward new methods to alleviate ischemia reperfusion injury (IRI). Oxidative stress and nuclear factor kappa beta (NF-κB) activation are well-described elements of IRI. We added cyclodextrin-complexed curcumin (CDC), a potent antioxidant and NF-κB inhibitor, to University of Wisconsin (UW) solution (Belzer's Solution, Viaspan), one of the most effective clinically approved preservative solutions. The effects of CDC were evaluated on pig endothelial cells and in an autologous donation after circulatory death (DCD) kidney transplantation model in large white pigs. CDC allowed rapid and lasting uptake of curcumin into cells. In vitro, CDC decreased mitochondrial loss of function, improved viability and lowered endothelial activation. In vivo, CDC improved function recovery, lowered histological injury and doubled animal survival (83.3% vs. 41.7%). At 3 months, immunohistochemical staining for epithelial-to-mesenchymal transition (EMT) and fibrosis markers was intense in UW grafts while it remained limited in the UW + CDC group. Transcriptional analysis showed that CDC treatment protected against up-regulation of several pathophysiological pathways leading to inflammation, EMT and fibrosis. Thus, use of CDC in a preclinical transplantation model with stringent IRI rescued kidney grafts from an unfavorable prognosis. As curcumin has proved well tolerated and nontoxic, this strategy shows promise for translation to the clinic.
Collapse
Affiliation(s)
- R Thuillier
- Inserm U1082, Faculté de Medecine et Pharmacie, Université de Poitiers, Poitiers, France; Département de Biochimie, CHU de Poitiers, Poitiers, France; FLIRT: Fédération pour L'étude de l'Ischémie Reperfusion en Transplantation, Poitiers, France; COPE: Consortium for Organ Preservation in Europe
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
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
|
38
|
Lu B, Wang C, Wang M, Li W, Chen F, Tracey KJ, Wang H. Molecular mechanism and therapeutic modulation of high mobility group box 1 release and action: an updated review. Expert Rev Clin Immunol 2014; 10:713-27. [PMID: 24746113 DOI: 10.1586/1744666x.2014.909730] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
High mobility group box 1 (HMGB1) is an evolutionarily conserved protein, and is constitutively expressed in virtually all types of cells. Infection and injury converge on common inflammatory responses that are mediated by HMGB1 secreted from immunologically activated immune cells or passively released from pathologically damaged cells. Herein we review the emerging molecular mechanisms underlying the regulation of pathogen-associated molecular patterns (PAMPs)-induced HMGB1 secretion, and summarize many HMGB1-targeting therapeutic strategies for the treatment of infection- and injury-elicited inflammatory diseases. It may well be possible to develop strategies that specifically attenuate damage-associated molecular patterns (DAMPs)-mediated inflammatory responses without compromising the PAMPs-mediated innate immunity for the clinical management of infection- and injury-elicited inflammatory diseases.
Collapse
Affiliation(s)
- Ben Lu
- Department of Hematology, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | | | | | | | | | | | | |
Collapse
|
39
|
Chatauret N, Favreau F, Giraud S, Thierry A, Rossard L, Le Pape S, Lerman LO, Hauet T. Diet-induced increase in plasma oxidized LDL promotes early fibrosis in a renal porcine auto-transplantation model. J Transl Med 2014; 12:76. [PMID: 24655356 PMCID: PMC3994364 DOI: 10.1186/1479-5876-12-76] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 03/12/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND In kidney transplantation, the prevalence of hypercholesterolemia as a co-morbidity factor known to affect graft function, is rising due to the increased number of older donors in response to organ shortage as well as to the hyperlipidemic effects of immunosuppressors in recipient. This study aimed to characterize the effects of hypercholesterolemia on renal graft outcome, investigating the role of oxidized low-density lipoprotein (OxLDL). METHODS In vivo, we used a porcine preclinical model of renal auto-transplantation modulated by two experimental diets: a normal (n = 6) or a hyperlipidemic diet (n = 5) maintained during the 3 month follow-up after the surgical procedure. Kidney function and OxLDL levels were monitored as well as fibrosis, LOX-1 and TGF beta signaling pathways. In vitro, we used human artery endothelial cells subjected to OxLDL to investigate the TGF beta profibrotic pathway and the role of the scavenger receptor LOX-1. RESULTS Hyperlipidemic diet-induced increase in plasma OxLDL levels at the time of surgery correlated with an increase in proteinuria 3 months after transplantation, associated with an early graft fibrosis combined with an activation of renal TGF beta signaling. These data suggest a direct involvement of OxLDL in the hyperlipidemic diet-induced activation of the pro-fibrotic TGF beta pathway which seems to be activated by LOX-1 signaling. These results were supported by studies with endothelial cells incubated in culture medium containing OxLDL promoting TGF beta expression inhibited by LOX-1 antibody. CONCLUSIONS These results implicate OxLDL in the hyperlipidemic diet-promoted fibrosis in transplanted kidneys, suggesting LOX-1 as a potential therapeutic target and reinforce the need to control cholesterol levels in kidney transplant recipients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Thierry Hauet
- INSERM, U1082, Ischémie-reperfusion en transplantation rénale, Université de Poitiers, Faculté de Médecine et de Pharmacie, Poitiers 86000, France.
| |
Collapse
|
40
|
Schneider DJ. Potential Contribution of Pleiotropic Effects of Direct Anticoagulants to Clinical Benefits. Drug Dev Res 2013. [DOI: 10.1002/ddr.21104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- David J. Schneider
- Cardiology Unit; Cardiovascular Research Institute; Department of Medicine; University of Vermont; Burlington; Vermont; USA
| |
Collapse
|
41
|
Hammad FT, Al-Salam S, Lubbad L. Does aliskiren protect the kidney following ischemia reperfusion injury? Physiol Res 2013; 62:681-90. [PMID: 23869885 DOI: 10.33549/physiolres.932485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The effect of blocking the first and rate-limiting step in renin-angiotensin cascade on the renal function in ischemia reperfusion injury has not been previously investigated. We investigated the effect of aliskiren, the first approved direct oral renin inhibitor, on the alterations in renal functional parameters in this condition. Wistar rats underwent left renal ischemia for 40 min. Group-1 received normal saline whereas Group-2 received aliskiren (30 mg/kg/day) by gavage for 6 days commencing one day before IRI. The hemodynamic and tubular functions and gene expression of neutrophil gelatinase-associated lipocalin (NGAL) and plasminogen activating inhibitor (PAI-1) in the right and left kidneys were measured five days following the IRI. Comparing Group-1 and Group-2, the left renal blood flow was significantly higher in Group-2 (1.28+/-0.36 vs. 0.39+/-0.05, P=0.007). Left kidney glomerular filtration rate was also higher in Group-2 but did not reach statistical significance (0.18+/-0.05 vs. 0.10+/-0.02, P=0.07). The left renal FE(Na) was significantly lower in Group-2 (29.9+/-6.4 vs. 49.7+/-7.8, P=0.03). Aliskiren also caused a significant decrease in the gene expression of both NGAL and PAI-1 in the left ischemic kidney. In conclusions, the administration of aliskiren before and after IRI appears to have ameliorated the IRI effect on the total renal artery blood flow, fractional excretion of sodium and gene expression of both NGAL and PAI-1 indicating a renoprotective effects in IRI.
Collapse
Affiliation(s)
- F T Hammad
- Department of Surgery, College of Medicine and Health Sciences, Al Ain, United Arab Emirates.
| | | | | |
Collapse
|
42
|
Kim JI, Jung KJ, Jang HS, Park KM. Gender-specific role of HDAC11 in kidney ischemia- and reperfusion-induced PAI-1 expression and injury. Am J Physiol Renal Physiol 2013; 305:F61-70. [DOI: 10.1152/ajprenal.00015.2013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Male gender and the male hormone testosterone increase susceptibility to kidney ischemia and reperfusion (I/R) injury, which is associated with inflammatory responses. Possible involvement of histone deacetylase (HDAC) in inflammatory responses has been suggested. We investigated the gender-specific role of HDACs in plasminogen activator inhibitor type-1 (PAI-1) expression and I/R injury. PAI-1 inhibition protected the kidney from I/R-induced inflammation and functional loss. Among HDACs, only HDAC11 negatively regulated PAI-1 expression in I/R-subjected kidney gender specifically and lipopolysaccharide (LPS)-stimulated mouse monocytes/macrophages. HDAC11 gene silencing increased PAI-1 expression. Chromatin immunoprecipitation assay confirmed binding of HDAC11 to the promoter region of PAI-1 and then release by I/R insult or LPS treatment. I/R-induced HDAC11 release was inhibited by orchiectomy and reversed by dihydrotestosterone treatment. Release of HDAC11 increased acetylation of histone H3. In conclusion, male gender and male hormones accelerate I/R-induced decreases in expression and binding of HDAC11, resulting in an increase in PAI-1 expression. These data provide important insight into gender dimorphism offering HDAC11 as a novel target for I/R injury.
Collapse
Affiliation(s)
- Jee In Kim
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
- Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Kyong-Jin Jung
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
| | - Hee-Seong Jang
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
| | - Kwon Moo Park
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
- Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
43
|
Rossard L, Favreau F, Giraud S, Thuillier R, Le Pape S, Goujon JM, Valagier A, Hauet T. Role of warm ischemia on innate and adaptive responses in a preclinical renal auto-transplanted porcine model. J Transl Med 2013; 11:129. [PMID: 23706041 PMCID: PMC3666894 DOI: 10.1186/1479-5876-11-129] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/13/2013] [Indexed: 01/06/2023] Open
Abstract
Background Deceased after cardiac arrest donor are an additional source of kidney graft to overcome graft shortage. Deciphering the respective role of renal warm and cold ischemia is of pivotal interest in the transplantation process. Methods Using a preclinical pig model of renal auto-transplantation, we investigated the consequences of warm and cold ischemia on early innate and adaptive responses as well as graft outcome. Kidneys were subjected to either 60 min-warm ischemia (WI) or auto-transplanted after cold storage for 24 h at 4°C (CS), or both conditions combined (WI + CS). Renal function, immune response and cytokine expression, oxidative stress and cell death were investigated at 3 h, 3 and 7 days (H3, D3 and D7) after reperfusion. At 3 months, we focused on cell infiltration and tissue remodelling. Results WI + CS induced a delayed graft function linked to higher tubular damage. Innate response occurred at D3 associated to a pro-oxidative milieu with a level dependent on the severity of ischemic injury whereas adaptive immune response occurred only at D7 mainly due to CS injuries and aggravated by WI. Graft cellular death was an early event detected at H3 and seems to be one of the first ischemia reperfusion injuries. These early injuries affect graft outcome on renal function, cells infiltration and fibrosis development. Conclusions The results indicate that the severe ischemic insult found in kidneys from deceased after cardiac arrest donor affects kidney outcome and promotes an uncontrolled deleterious innate and adaptive response not inhibited 3 months after reperfusion.
Collapse
|
44
|
Abstract
BACKGROUND Increasing evidence accumulates on the central involvement of microRNAs (miRNAs) in disease pathophysiology. We identified distinctly deregulated miRNAs in human renal allograft biopsies from patients undergoing acute cellular rejection, antibody-mediated rejection (ABMR), and delayed graft function (DGF). METHODS Sixty-five posttransplantation kidney biopsy samples covering 41 cases with acute rejection (15 vascular rejection, 15 interstitial rejection, and 11 ABMR), 14 DGF cases, and 10 protocol biopsies serving as controls were analyzed using the Affymetrix GeneChip miRNA Array. Differentially regulated miRNAs were identified by Student's t test and Bonferroni correction. Target genes for the set of miRNAs were retrieved from miRTarBase (experimentally verified targets) as well as by applying the target prediction routines DIANAmT, miRanda, and Targetscan. RESULTS Patients with acute cellular rejection, ABMR, and DGF discriminate from the control group (protocol biopsies) in unsupervised clustering of miRNA profiles, clearly identifying deregulated miRNAs in rejection and DGF. Angiogenesis, apoptosis, and transforming growth factor-β signaling were identified as relevant pathways in ischemic response following an integrative analysis of miRNA targets and mRNA expression profiles. Inflammation by chemokine and cytokine signaling, T-cell activation, and B-cell activation were identified as relevant in acute rejection accordingly. CONCLUSION These data suggest that distinct miRNA signatures playing a role in specific biological pathways discriminate acute cellular and humoral rejection and DGF. This finding serves as valuable tool for a rational selection of diagnostic, prognostic, and potentially therapeutic molecular targets of posttransplantation events.
Collapse
|
45
|
Thuillier R, Codas R, Marchand E, Lathelize H, Page O, Valagier A, Badet L, Hauet T. Chronic renoprotective effect of pulsatile perfusion machine RM3 and IGL-1 solution in a preclinical kidney transplantation model. J Transl Med 2012; 10:233. [PMID: 23171422 PMCID: PMC3543357 DOI: 10.1186/1479-5876-10-233] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/30/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Machine perfusion (MP) of kidney graft provides benefits against preservation injury, however decreased graft quality requires optimization of the method. We examined the chronic benefits of MP on kidney grafts and the potential improvements provided by IGL-1 solution. METHOD We used an established autotransplantation pig kidney model to study the effects of MP against the deleterious effects of warm ischemia (WI: 60 minutes) followed by 22 hours of cold ischemia in MP or static cold storage (CS) followed by autotransplantation. MPS and IGL-1 solutions were compared. RESULTS Animal survival was higher in MPS-MP and both IGL groups. Creatinine measurement did not discriminate between the groups, however MPS-MP and both IGL groups showed decreased proteinuria. Chronic fibrosis level was equivalent between the groups. RTqPCR and immunohistofluorescent evaluation showed that MP and IGL-1 provided some protection against epithelial to mesenchymal transition and chronic lesions. IGL-1 was protective with both MP and CS, particularly against chronic inflammation, with only small differences between the groups. CONCLUSION IGL-1 used in either machine or static preservation offers similar levels of protection than standard MP. The compatibility of IGL-1 with both machine perfusion and static storage could represent an advantage for clinical teams when choosing the correct solution to use for multi-organ collection. The path towards improving machine perfusion, and organ quality, may involve the optimization of the solution and the correct use of colloids.
Collapse
Affiliation(s)
- Raphael Thuillier
- Inserm U1082, Université de Poitiers, Faculté de Médecine et Pharmacie, Poitiers, 86021, France
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Oliveira FAM, Moraes ACM, Paiva AP, Schinzel V, Correa-Costa M, Semedo P, Castoldi A, Cenedeze MA, Oliveira RSMF, Bastos MG, Câmara NOS, Sanders-Pinheiro H. Low-level laser therapy decreases renal interstitial fibrosis. Photomed Laser Surg 2012; 30:705-13. [PMID: 23134313 DOI: 10.1089/pho.2012.3272] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE the purpose of this study was to investigate the effect of low-level laser therapy (LLLT) on chronic kidney disease (CKD) in a model of unilateral ureteral obstruction (UUO). BACKGROUND DATA Regardless of the etiology, CKD involves progressive widespread tissue fibrosis, tubular atrophy, and loss of kidney function. This process also occurs in kidney allograft. At present, effective therapies for this condition are lacking. We investigated the effects of LLLT on the interstitial fibrosis that occurs after experimental UUO in rats. METHODS The occluded kidney of half of the 32 Wistar rats that underwent UUO received a single intraoperative dose of LLLT (AlGaAs laser, 780 nm, 22.5 J/cm(2), 30 mW, 0.75 W/cm(2), 30 sec on each of nine points). After 14 days, renal fibrosis was assessed by Sirius red staining under polarized light. Immunohistochemical analyses quantitated the renal tissue cells that expressed fibroblast (FSP-1) and myofibroblast (α-SMA) markers. Reverse transcriptase polymerase chain reaction (RT-PCR) was performed to determine the mRNA expression of interleukin (IL)-6, monocyte chemotactic protein-1 (MCP-1), transforming growth factor (TGF)-β1 and Smad3. RESULTS The UUO and LLLT animals had less fibrosis than the UUO animals, as well having decreased expression inflammatory and pro-fibrotic markers. CONCLUSIONS For the first time, we showed that LLLT had a protective effect regarding renal interstitial fibrosis. It is conceivable that by attenuating inflammation, LLLT can prevent tubular activation and transdifferentiation, which are the two processes that mainly drive the renal fibrosis of the UUO model.
Collapse
|
47
|
Geng H, Lan R, Singha PK, Gilchrist A, Weinreb PH, Violette SM, Weinberg JM, Saikumar P, Venkatachalam MA. Lysophosphatidic acid increases proximal tubule cell secretion of profibrotic cytokines PDGF-B and CTGF through LPA2- and Gαq-mediated Rho and αvβ6 integrin-dependent activation of TGF-β. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1236-49. [PMID: 22885106 DOI: 10.1016/j.ajpath.2012.06.035] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Accepted: 06/20/2012] [Indexed: 10/28/2022]
Abstract
After ischemia-reperfusion injury (IRI), kidney tubules show activated transforming growth factor β (TGF-β) signaling and increased expression of profibrotic peptides, platelet-derived growth factor-B (PDGF-B) and connective tissue growth factor (CTGF). If tubule repair after IRI is incomplete, sustained paracrine activity of these peptides can activate interstitial fibroblast progenitors and cause fibrosis. We show that lysophosphatidic acid (LPA), a ubiquitous phospholipid that is increased at sites of injury and inflammation, signals through LPA2 receptors and Gαq proteins of cultured proximal tubule cells to transactivate latent TGF-β in a Rho/Rho-kinase and αvβ6 integrin-dependent manner. Active TGF-β peptide then initiates signaling to increase the production and secretion of PDGF-B and CTGF. In a rat model of IRI, increased TGF-β signaling that was initiated early during reperfusion did not subside during recovery, but progressively increased, causing tubulointerstitial fibrosis. This was accompanied by correspondingly increased LPA2 and β6 integrin proteins and elevated tubule expression of TGF-β1, together with PDGF-B and CTGF. Treatment with a pharmacological TGF-β type I receptor antagonist suppressed TGF-β signaling, decreased the expression of β6 integrin, PDGF-B, and CTGF, and ameliorated fibrosis. We suggest that LPA-initiated autocrine signaling is a potentially important mechanism that gives rise to paracrine profibrotic signaling in injured kidney tubule cells.
Collapse
Affiliation(s)
- Hui Geng
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas 78229, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bon D, Chatauret N, Giraud S, Thuillier R, Favreau F, Hauet T. New strategies to optimize kidney recovery and preservation in transplantation. Nat Rev Nephrol 2012; 8:339-47. [DOI: 10.1038/nrneph.2012.83] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
49
|
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
|
50
|
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
|