1
|
Yin XH, Wang XY, Liu SC, Chen XX, Yan L, Li L, Le He G, Yang M, Liu ZK. SIRT5 -mediated desuccinylation of UQCRC2 attenuates osteogenic differentiation of aged BM-MSCs through impairing mitochondrial homeostasis. Cell Signal 2025; 128:111636. [PMID: 39892725 DOI: 10.1016/j.cellsig.2025.111636] [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: 10/14/2024] [Revised: 01/19/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND The osteogenic differentiation potential of bone marrow mesenchymal stem cells (BM-MSCs) is critical for bone regeneration and repair. In recent years, the role of protein succinylation modification in regulating cellular metabolism has garnered increasing attention. However, its mechanism in osteogenic differentiation remains unclear. METHODS Oxygen consumption rate (OCR) and mitochondrial ROS (mtROS) were detected to assess mitochondrial function in BM-MSCs with successive passages. Alizarin red staining and western blot experiments were used to evaluate osteogenic differentiation capacity. Succinylation modification omics and Co-IP detection were conducted to determine SIRT5-mediated desuccinylation of UQCRC2. RESULTS Bioinformatics analysis revealed that sirtuin 5 (SIRT5) expression is upregulated with multiple rounds of BM-MSCs' passages, and is associated with biological pathways such as oxidative phosphorylation (OXPHOS), cellular senescence, and inhibition of osteogenic differentiation. Experiments in vitro confirmed the up-regulation of SIRT5 and the suppression of osteogenic differentiation with the increased times of BM-MSCs' passages. Overexpression of SIRT5 enhanced OXPHOS and elevated mtROS levels, but reduced the expression of Runx2 and osteocalcin, and decreased calcified nodules, thereby inhibiting the osteogenic differentiation of BM-MSCs. SIRT5-mediated desuccinylation of ubiquinol-cytochrome C reductase core protein 2 (UQCRC2) at the site of K250 promoted UQCRC2 translocation from cytoplasm to mitochondria, which enhanced the activity of mitochondrial respiratory complex III. It further increased mtROS, accelerated cellular senescence and inhibited the osteogenic differentiation of BM-MSCs. CONCLUSION SIRT5 reduces succinylation modification of UQCRC2, promotes mitochondrial respiration and mtROS, and thus reduces the osteogenic differentiation ability of BM-MSCs cells. SIRT5 might be a potential target to prevent the suppression of osteogenic differentiation of of BM-MSCs after multiple rounds passages.
Collapse
Affiliation(s)
- Xin Hua Yin
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Xiao Yuan Wang
- Physical Examination Center, Xi'an International Medical Center Hospital, Xi'an, China
| | - Shi Chang Liu
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Xu Xu Chen
- Department of Sports Medicine, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Liang Yan
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Liang Li
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Gao Le He
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Ming Yang
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China.
| | - Zhong Kai Liu
- Department of Spine Surgery, Hong Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China.
| |
Collapse
|
2
|
Becker S, Allen J, Morison ZL, Saeid S, Adderley A, Koskelainen A, Vinberg F. Healing of ischemic injury in the retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.04.621932. [PMID: 39574566 PMCID: PMC11580909 DOI: 10.1101/2024.11.04.621932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Neuro- and retinal degenerative diseases, including Alzheimer's, age-related macular degeneration, stroke, and central retinal artery occlusion, rob millions of their independence. Studying these diseases in human retinas has been hindered by the immediate loss of neuronal activity postmortem. While recent studies restored limited activity in postmortem CNS tissues, synchronized neuronal transmission >30 minutes postmortem remained elusive. Our study overcomes this barrier by reviving and sustaining light signal transmission in human retinas recovered up to four hours and stored 48 hours postmortem. We also establish infrared-based ex vivo imaging for precise sampling, a closed perfusion system for drug testing, and an ex vivo ischemia-reperfusion model in mouse and human retina. This platform enables testing of neuroprotective and neurotoxic effects of drugs targeting oxidative stress and glutamate excitotoxicity. Our advances question the irreversibility of ischemic injury, support preclinical vision restoration studies, offer new insights into treating ischemic CNS injuries, and pave the way for transplantation of human donor eyes. Teaser Reviving light signaling in postmortem human retinas challenges the irreversibility of ischemic injury and advances research to restore vision.
Collapse
|
3
|
Yim WY, Li C, Tong F, Hou J, Chen Y, Liu Z, Wang Z, Geng B, Wang Y, Dong N. Circadian immune system in solid organ transplantation: a review article. Front Immunol 2025; 16:1556057. [PMID: 40098968 PMCID: PMC11911371 DOI: 10.3389/fimmu.2025.1556057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
The innate and adaptive immune systems are intricately regulated by the circadian clock machinery. Recent clinical investigations have shed light on the influence of timing in organ procurement and transplantation on graft survival. In this review, we explore various mechanisms of immunological functions associated with the steps involved in organ transplantation, spanning from surgical harvesting to reperfusion and linking to the circadian rhythm. A deeper understanding of these processes has the potential to extend the principles of chrono-immunotherapy to the realm of organ transplantation, with the aim of enhancing graft durability and improving patient outcomes. This review concludes with some perspectives on future directions in this exciting and still evolving field of research.
Collapse
Affiliation(s)
- Wai Yen Yim
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenghao Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuqiang Tong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jincheng Hou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqi Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zongtao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingchuan Geng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
4
|
Yang J, Duan C, Wang P, Zhang S, Gao Y, Lu S, Ji Y. 4-Octyl Itaconate Alleviates Myocardial Ischemia-Reperfusion Injury Through Promoting Angiogenesis via ERK Signaling Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411554. [PMID: 39836624 PMCID: PMC11904966 DOI: 10.1002/advs.202411554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/13/2024] [Indexed: 01/23/2025]
Abstract
Myocardial ischemia-reperfusion (IR) injury is a critical complication following revascularization therapy for ischemic heart disease. Itaconate, a macrophage-derived metabolite, has been implicated in inflammation and metabolic regulation. This study investigates the protective role of itaconate derivatives against IR injury. Using a mice model of IR injury, the impact of 7-day 4-Octyl itaconate (4-OI) administration on cardiac function is assessed. Exogenous administration of 4-OI significantly reduces myocardial damage, enhances angiogenesis, and alleviates myocardial hypoxia injury during reperfusion. RNA sequencing and molecular docking techniques are used to find the target of itaconate, and changes in cardiac function are observed in Immune-Responsive Gene1 (IRG1) global knockout mice. In cell culture studies, 4-OI promotes endothelial cell proliferation and migration, mediated by Mitogen-Activated Protein Kinases (MAPK) signaling pathway activation, particularly through Extracellular Signal-Regulated Kinase (ERK) signaling. Inhibition of ERK blocks these beneficial effects on endothelial cells. Furthermore, itaconate synthesis inhibition worsens myocardial damage, which is mitigated by 4-OI supplementation. The results indicate that 4-OI promotes angiogenesis by activating MAPK signaling via FMS-like tyrosine kinase 1 (Flt1), highlighting its potential as a therapeutic strategy for myocardial IR injury.
Collapse
Affiliation(s)
- Jiqin Yang
- Key Laboratory of Cardiovascular and Cerebrovascular MedicineSchool of PharmacyNanjing Medical University101 Longmian Avenue, Jiangning DistrictNanjing211166P. R. China
| | - Chenqi Duan
- Key Laboratory of Cardiovascular and Cerebrovascular MedicineSchool of PharmacyNanjing Medical University101 Longmian Avenue, Jiangning DistrictNanjing211166P. R. China
| | - Peng Wang
- Key Laboratory of Cardiovascular and Cerebrovascular MedicineSchool of PharmacyNanjing Medical University101 Longmian Avenue, Jiangning DistrictNanjing211166P. R. China
| | - Sijia Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Harbin Medical UniversityHarbinHeilongjiang150081P. R. China
| | - Yuanqing Gao
- Key Laboratory of Cardiovascular and Cerebrovascular MedicineSchool of PharmacyNanjing Medical University101 Longmian Avenue, Jiangning DistrictNanjing211166P. R. China
| | - Shan Lu
- Key Laboratory of Cardiovascular and Cerebrovascular MedicineSchool of PharmacyNanjing Medical University101 Longmian Avenue, Jiangning DistrictNanjing211166P. R. China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular MedicineSchool of PharmacyNanjing Medical University101 Longmian Avenue, Jiangning DistrictNanjing211166P. R. China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Harbin Medical UniversityHarbinHeilongjiang150081P. R. China
| |
Collapse
|
5
|
Nengroo MA, Klein AT, Carr HS, Vidal-Cruchez O, Sahu U, McGrail DJ, Sahni N, Gao P, Asara JM, Shah H, Mendillo ML, Ben-Sahra I. Succinate dehydrogenase activity supports de novo purine synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640389. [PMID: 40060604 PMCID: PMC11888382 DOI: 10.1101/2025.02.26.640389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
The de novo purine synthesis pathway is fundamental for nucleic acid production and cellular energetics, yet the role of mitochondrial metabolism in modulating this process remains underexplored. In many cancers, metabolic reprogramming supports rapid proliferation and survival, but the specific contributions of the tricarboxylic acid (TCA) cycle enzymes to nucleotide biosynthesis are not fully understood. Here, we demonstrate that the TCA cycle enzyme succinate dehydrogenase (SDH) is essential for maintaining optimal de novo purine synthesis in normal and cancer cells. Genetic or pharmacological inhibition of SDH markedly attenuates purine synthesis, leading to a significant reduction in cell proliferation. Mechanistically, SDH inhibition causes an accumulation of succinate, which directly impairs the purine biosynthetic pathway. In response, cancer cells compensate by upregulating the purine salvage pathway, a metabolic adaptation that represents a potential therapeutic vulnerability. Notably, co-inhibition of SDH and the purine salvage pathway induces pronounced antiproliferative and antitumoral effects in preclinical models. These findings not only reveal a signaling role for mitochondrial succinate in regulating nucleotide metabolism but also provide a promising therapeutic strategy for targeting metabolic dependencies in cancer.
Collapse
Affiliation(s)
- Mushtaq A Nengroo
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Austin T Klein
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Heather S Carr
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Olivia Vidal-Cruchez
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Umakant Sahu
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Daniel J McGrail
- Center for Immunotherapy and Precision Immuno Oncology, Cleveland Clinic, Cleveland, OH, 441796, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Peng Gao
- Metabolomics Core Facility, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, 60611, USA
| | - John M Asara
- Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Hardik Shah
- Metabolomics Platform, University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL, 60637, USA
| | - Marc L Mendillo
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago IL, 60611 USA
| |
Collapse
|
6
|
Zhang C, Chang X, Zhao D, He Y, Dong G, Gao L. Mitochondria and myocardial ischemia/reperfusion injury: Effects of Chinese herbal medicine and the underlying mechanisms. J Pharm Anal 2025; 15:101051. [PMID: 39931135 PMCID: PMC11808734 DOI: 10.1016/j.jpha.2024.101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 02/03/2025] Open
Abstract
Ischemic heart disease (IHD) is associated with high morbidity and mortality rates. Reperfusion therapy is the best treatment option for this condition. However, reperfusion can aggravate myocardial damage through a phenomenon known as myocardial ischemia/reperfusion (I/R) injury, which has recently gained the attention of researchers. Several studies have shown that Chinese herbal medicines and their natural monomeric components exert therapeutic effects against I/R injury. This review outlines the current knowledge on the pathological mechanisms through which mitochondria participate in I/R injury, focusing on the issues related to energy metabolism, mitochondrial quality control disorders, oxidative stress, and calcium. The mechanisms by which mitochondria mediate cell death have also been discussed. To develop a resource for the prevention and management of clinical myocardial I/R damage, we compiled the most recent research on the effects of Chinese herbal remedies and their monomer components.
Collapse
Affiliation(s)
- Chuxin Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xing Chang
- Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, 100053, China
| | - Dandan Zhao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu He
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Guangtong Dong
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lin Gao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| |
Collapse
|
7
|
Cillo U, Lonati C, Bertacco A, Magnini L, Battistin M, Borsetto L, Dazzi F, Al-Adra D, Gringeri E, Bacci ML, Schlegel A, Dondossola D. A proof-of-concept study in small and large animal models for coupling liver normothermic machine perfusion with mesenchymal stromal cell bioreactors. Nat Commun 2025; 16:283. [PMID: 39746966 PMCID: PMC11697227 DOI: 10.1038/s41467-024-55217-7] [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: 08/24/2023] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
To fully harness mesenchymal-stromal-cells (MSCs)' benefits during Normothermic Machine Perfusion (NMP), we developed an advanced NMP platform coupled with a MSC-bioreactor and investigated its bio-molecular effects and clinical feasibility using rat and porcine models. The study involved three work packages: 1) Development (n = 5): MSC-bioreactors were subjected to 4 h-liverless perfusion; 2) Rat model (n = 10): livers were perfused for 4 h on the MSC-bioreactor-circuit or with the standard platform; 3) Porcine model (n = 6): livers were perfused using a clinical device integrated with a MSC-bioreactor or in its standard setup. MSCs showed intact stem-core properties after liverless-NMP. Liver NMP induced specific, liver-tailored, changes in MSCs' secretome. Rat livers exposed to bioreactor-based perfusion produced more bile, released less damage and pro-inflammatory biomarkers, and showed improved mithocondrial function than those subjected to standard NMP. MSC-bioreactor integration into a clinical device resulted in no machine failure and perfusion-related injury. This proof-of-concept study presents a novel MSC-based liver NMP platform that could reduce the deleterious effects of ischemia/reperfusion before transplantation.
Collapse
Affiliation(s)
- Umberto Cillo
- Hepato-Biliary-Pancreatic Surgery and Liver Transplant Unit, General Surgery 2, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Caterina Lonati
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20100, Milan, Italy.
| | - Alessandra Bertacco
- Hepato-Biliary-Pancreatic Surgery and Liver Transplant Unit, General Surgery 2, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Lucrezia Magnini
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20100, Milan, Italy
| | - Michele Battistin
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20100, Milan, Italy
| | - Lara Borsetto
- Hepato-Biliary-Pancreatic Surgery and Liver Transplant Unit, General Surgery 2, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Francesco Dazzi
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
| | - David Al-Adra
- Division of Transplantation, Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Enrico Gringeri
- Hepato-Biliary-Pancreatic Surgery and Liver Transplant Unit, General Surgery 2, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Maria Laura Bacci
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Andrea Schlegel
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Pace 9, 20100, Milan, Italy
- Transplantation Center, Digestive Disease and Surgery Institute, Department of Immunity and Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Daniele Dondossola
- General and Liver Transplant Surgery Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via Francesco Sforza 35, 20100, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Via Francesco Sforza 35, 20100, Milan, Italy
| |
Collapse
|
8
|
Tian L, Liu Q, Guo H, Zang H, Li Y. Fighting ischemia-reperfusion injury: Focusing on mitochondria-derived ferroptosis. Mitochondrion 2024; 79:101974. [PMID: 39461581 DOI: 10.1016/j.mito.2024.101974] [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: 07/04/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of mortality and morbidity. Current treatments for IRI have limited efficacy and novel therapeutic strategies are needed. Mitochondrial dysfunction not only initiates IRI but also plays a significant role in ferroptosis pathogenesis. Recent studies have highlighted that targeting mitochondrial pathways is a promising therapeutic approach for ferroptosis-induced IRI. The association between ferroptosis and IRI has been reviewed many times, but our review provides the first comprehensive overview with a focus on recent mitochondrial research. First, we present the role of mitochondria in ferroptosis. Then, we summarize the evidence on mitochondrial manipulation of ferroptosis in IRI and review recent therapeutic strategies aimed at targeting mitochondria-related ferroptosis to mitigate IRI. We hope our review will provide new ideas for the treatment of IRI and accelerate the transition from bench to bedside.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, China
| | - Hong Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Honggang Zang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
9
|
Ilzorkina AI, Belosludtseva NV, Semenova AA, Dubinin MV, Belosludtsev KN. The Effect of TRO19622 (Olesoxime) on the Functional Activity of Isolated Mitochondria and Cell Viability. Biophysics (Nagoya-shi) 2024; 69:630-638. [DOI: 10.1134/s0006350924700714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 03/15/2024] [Accepted: 06/19/2024] [Indexed: 05/04/2025] Open
|
10
|
Sorby-Adams A, Prime TA, Miljkovic JL, Prag HA, Krieg T, Murphy MP. A model of mitochondrial superoxide production during ischaemia-reperfusion injury for therapeutic development and mechanistic understanding. Redox Biol 2024; 72:103161. [PMID: 38677214 PMCID: PMC11066467 DOI: 10.1016/j.redox.2024.103161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/01/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
Ischaemia-reperfusion (IR) injury is the paradoxical consequence of the rapid restoration of blood flow to an ischaemic organ. Although reperfusion is essential for tissue survival in conditions such as myocardial infarction and stroke, the excessive production of mitochondrial reactive oxygen species (ROS) upon reperfusion initiates the oxidative damage that underlies IR injury, by causing cell death and inflammation. This ROS production is caused by an accumulation of the mitochondrial metabolite succinate during ischaemia, followed by its rapid oxidation upon reperfusion by succinate dehydrogenase (SDH), driving superoxide production at complex I by reverse electron transport. Inhibitors of SDH, such as malonate, show therapeutic potential by decreasing succinate oxidation and superoxide production upon reperfusion. To better understand the mechanism of mitochondrial ROS production upon reperfusion and to assess potential therapies, we set up an in vitro model of IR injury. For this, isolated mitochondria were incubated anoxically with succinate to mimic ischaemia and then rapidly reoxygenated to replicate reperfusion, driving a burst of ROS formation. Using this system, we assess the factors that contribute to the magnitude of mitochondrial ROS production in heart, brain, and kidney mitochondria, as well as screening for inhibitors of succinate oxidation with therapeutic potential.
Collapse
Affiliation(s)
- Annabel Sorby-Adams
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge, CB2 0XY, UK
| | - Tracy A Prime
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge, CB2 0XY, UK
| | - Jan Lj Miljkovic
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge, CB2 0XY, UK
| | - Hiran A Prag
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge, CB2 0XY, UK; Department of Medicine, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
11
|
Marques E, Kramer R, Ryan DG. Multifaceted mitochondria in innate immunity. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:6. [PMID: 38812744 PMCID: PMC11129950 DOI: 10.1038/s44324-024-00008-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/14/2024] [Indexed: 05/31/2024]
Abstract
The ability of mitochondria to transform the energy we obtain from food into cell phosphorylation potential has long been appreciated. However, recent decades have seen an evolution in our understanding of mitochondria, highlighting their significance as key signal-transducing organelles with essential roles in immunity that extend beyond their bioenergetic function. Importantly, mitochondria retain bacterial motifs as a remnant of their endosymbiotic origin that are recognised by innate immune cells to trigger inflammation and participate in anti-microbial defence. This review aims to explore how mitochondrial physiology, spanning from oxidative phosphorylation (OxPhos) to signalling of mitochondrial nucleic acids, metabolites, and lipids, influences the effector functions of phagocytes. These myriad effector functions include macrophage polarisation, efferocytosis, anti-bactericidal activity, antigen presentation, immune signalling, and cytokine regulation. Strict regulation of these processes is critical for organismal homeostasis that when disrupted may cause injury or contribute to disease. Thus, the expanding body of literature, which continues to highlight the central role of mitochondria in the innate immune system, may provide insights for the development of the next generation of therapies for inflammatory diseases.
Collapse
Affiliation(s)
- Eloïse Marques
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Robbin Kramer
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Dylan G. Ryan
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
12
|
Ray S, Parmentier C, Kawamura M, Ganesh S, Nogueira E, Novoa FC, Hobeika C, Chu T, Kalimuthu SN, Selzner M, Reichman TW. Reanimating Pancreatic Grafts Subjected to Prolonged Cold Ischemic Injury Using Normothermic Ex Vivo Perfusion. Transplant Direct 2024; 10:e1620. [PMID: 38617463 PMCID: PMC11013695 DOI: 10.1097/txd.0000000000001620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 04/16/2024] Open
Abstract
Background Pancreas transplant volumes are limited because of poor utilization of "extended criteria grafts." Prolonged cold ischemia is a risk factor associated with poor allograft survival. We aimed to establish the feasibility of transplantation using grafts subjected to prolonged cold ischemia and determine whether these grafts could be optimized using normothermic ex vivo perfusion (NEVP) in a porcine model. Methods The study population consisted of 35 to 40 kg male Yorkshire pigs in an allotransplantation model with a 3-d survival plan for recipients. Control grafts were subjected to cold storage (CS) in a University of Wisconsin solution for 21 to 24 h (n = 6), whereas the test group received an additional 3 h NEVP after CS of 21 h (n = 5). Results The 3-d survival was 60% in the NEVP arm versus 0% in the control arm (P = 0.008; log rank). Graft parenchyma was 60% to 70% preserved in the NEVP arm at necropsy on gross appearance. In addition, the islet function was well preserved, and both the pancreas (including the islets) and the duodenal morphology were maintained histologically. The intravenous glucose tolerance test on the day of euthanasia was in the normoglycemic range for 80% of cases in the NEVP arm. Conclusions Optimization of pancreas grafts exposed to extended CS with NEVP seems promising at rescuing and reanimating these grafts for transplantation, resulting in significantly improved survival in a porcine pancreas transplant model.
Collapse
Affiliation(s)
- Samrat Ray
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Catherine Parmentier
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Masataka Kawamura
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Sujani Ganesh
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Emmanuel Nogueira
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Francisco Calderon Novoa
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Christian Hobeika
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Tunpang Chu
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
| | - Sangeetha N. Kalimuthu
- Department of Laboratory Medicine and Pathobiology, University Health Network, Toronto, ON, Canada
| | - Markus Selzner
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Trevor W. Reichman
- Department of Multiorgan Transplantation, Ajmera Transplant Centre, Toronto General Hospital, University Health Network, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
13
|
Cenik I, Van Slambrouck J, Provoost AL, Barbarossa A, Vanluyten C, Boelhouwer C, Vanaudenaerde BM, Vos R, Pirenne J, Van Raemdonck DE, Ceulemans LJ. Controlled Hypothermic Storage for Lung Preservation: Leaving the Ice Age Behind. Transpl Int 2024; 37:12601. [PMID: 38694492 PMCID: PMC11062243 DOI: 10.3389/ti.2024.12601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/06/2024] [Indexed: 05/04/2024]
Abstract
Controlled hypothermic storage (CHS) is a recent advance in lung transplantation (LTx) allowing preservation at temperatures higher than those achieved with traditional ice storage. The mechanisms explaining the benefits of CHS compared to conventional static ice storage (SIS) remain unclear and clinical data on safety and feasibility of lung CHS are limited. Therefore, we aimed to provide a focus review on animal experiments, molecular mechanisms, CHS devices, current clinical experience, and potential future benefits of CHS. Rabbit, canine and porcine experiments showed superior lung physiology after prolonged storage at 10°C vs. ≤4°C. In recent molecular analyses of lung CHS, better protection of mitochondrial health and higher levels of antioxidative metabolites were observed. The acquired insights into the underlying mechanisms and development of CHS devices allowed clinical application and research using CHS for lung preservation. The initial findings are promising; however, further data collection and analysis are required to draw more robust conclusions. Extended lung preservation with CHS may provide benefits to both recipients and healthcare personnel. Reduced time pressure between procurement and transplantation introduces flexibility allowing better decision-making and overnight bridging by delaying transplantation to daytime without compromising outcome.
Collapse
Affiliation(s)
- Ismail Cenik
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Jan Van Slambrouck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - An-Lies Provoost
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Annalisa Barbarossa
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Cedric Vanluyten
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Caroline Boelhouwer
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | | | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Pulmonology, University Hospitals Leuven, Leuven, Belgium
| | - Jacques Pirenne
- Abdominal Transplant Surgery, University Hospitals Leuven, Leuven, Belgium
- Immunology and Transplantation, Department of Microbiology, KU Leuven, Leuven, Belgium
| | - Dirk E. Van Raemdonck
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Laurens J. Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Wancewicz B, Pergande MR, Zhu Y, Gao Z, Shi Z, Plouff K, Ge Y. Comprehensive Metabolomic Analysis of Human Heart Tissue Enabled by Parallel Metabolite Extraction and High-Resolution Mass Spectrometry. Anal Chem 2024; 96:5781-5789. [PMID: 38568106 PMCID: PMC11057979 DOI: 10.1021/acs.analchem.3c04353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The heart contracts incessantly and requires a constant supply of energy, utilizing numerous metabolic substrates, such as fatty acids, carbohydrates, lipids, and amino acids, to supply its high energy demands. Therefore, a comprehensive analysis of various metabolites is urgently needed for understanding cardiac metabolism; however, complete metabolome analyses remain challenging due to the broad range of metabolite polarities, which makes extraction and detection difficult. Herein, we implemented parallel metabolite extractions and high-resolution mass spectrometry (MS)-based methods to obtain a comprehensive analysis of the human heart metabolome. To capture the diverse range of metabolite polarities, we first performed six parallel liquid-liquid extractions (three monophasic, two biphasic, and one triphasic) of healthy human donor heart tissue. Next, we utilized two complementary MS platforms for metabolite detection: direct-infusion ultrahigh-resolution Fourier-transform ion cyclotron resonance (DI-FTICR) and high-resolution liquid chromatography quadrupole time-of-flight tandem MS (LC-Q-TOF-MS/MS). Using DI-FTICR MS, 9644 metabolic features were detected where 7156 were assigned a molecular formula and 1107 were annotated by accurate mass assignment. Using LC-Q-TOF-MS/MS, 21,428 metabolic features were detected where 285 metabolites were identified based on fragmentation matching against publicly available libraries. Collectively, 1340 heart metabolites were identified in this study, which span a wide range of polarities including polar (benzenoids, carbohydrates, and nucleosides) as well as nonpolar (phosphatidylcholines, acylcarnitines, and fatty acids) compounds. The results from this study will provide critical knowledge regarding the selection of appropriate extraction and MS detection methods for the analysis of the diverse classes of human heart metabolites.
Collapse
Affiliation(s)
- Benjamin Wancewicz
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Melissa R. Pergande
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Yanlong Zhu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Zhan Gao
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Zhuoxin Shi
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Kylie Plouff
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| |
Collapse
|
15
|
Tabata Fukushima C, Dancil IS, Clary H, Shah N, Nadtochiy SM, Brookes PS. Reactive oxygen species generation by reverse electron transfer at mitochondrial complex I under simulated early reperfusion conditions. Redox Biol 2024; 70:103047. [PMID: 38295577 PMCID: PMC10844975 DOI: 10.1016/j.redox.2024.103047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/09/2024] [Accepted: 01/15/2024] [Indexed: 02/02/2024] Open
Abstract
Ischemic tissues accumulate succinate, which is rapidly oxidized upon reperfusion, driving a burst of mitochondrial reactive oxygen species (ROS) generation that triggers cell death. In isolated mitochondria with succinate as the sole metabolic substrate under non-phosphorylating conditions, 90 % of ROS generation is from reverse electron transfer (RET) at the Q site of respiratory complex I (Cx-I). Together, these observations suggest Cx-I RET is the source of pathologic ROS in reperfusion injury. However, numerous factors present in early reperfusion may impact Cx-I RET, including: (i) High [NADH]; (ii) High [lactate]; (iii) Mildly acidic pH; (iv) Defined ATP/ADP ratios; (v) Presence of the nucleosides adenosine and inosine; and (vi) Defined free [Ca2+]. Herein, experiments with mouse cardiac mitochondria revealed that under simulated early reperfusion conditions including these factors, total mitochondrial ROS generation was only 56 ± 17 % of that seen with succinate alone (mean ± 95 % confidence intervals). Of this ROS, only 52 ± 20 % was assignable to Cx-I RET. A further 14 ± 7 % could be assigned to complex III, with the remainder (34 ± 11 %) likely originating from other ROS sources upstream of the Cx-I Q site. Together, these data suggest the relative contribution of Cx-I RET ROS to reperfusion injury may be overestimated, and other ROS sources may contribute a significant fraction of ROS in early reperfusion.
Collapse
Affiliation(s)
- Caio Tabata Fukushima
- Departments of Anesthesiology, University of Rochester Medical Center, USA; Departments of Biochemistry, University of Rochester Medical Center, USA; Pharmacology and Physiology, University of Rochester Medical Center, USA
| | - Ian-Shika Dancil
- Departments of Anesthesiology, University of Rochester Medical Center, USA
| | - Hannah Clary
- Departments of Biochemistry, University of Rochester Medical Center, USA
| | - Nidhi Shah
- Pharmacology and Physiology, University of Rochester Medical Center, USA
| | - Sergiy M Nadtochiy
- Departments of Anesthesiology, University of Rochester Medical Center, USA
| | - Paul S Brookes
- Departments of Anesthesiology, University of Rochester Medical Center, USA; Pharmacology and Physiology, University of Rochester Medical Center, USA.
| |
Collapse
|
16
|
Bundgaard A, Borowiec BG, Lau GY. Are reactive oxygen species always bad? Lessons from hypoxic ectotherms. J Exp Biol 2024; 227:jeb246549. [PMID: 38533673 DOI: 10.1242/jeb.246549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Oxygen (O2) is required for aerobic energy metabolism but can produce reactive oxygen species (ROS), which are a wide variety of oxidant molecules with a range of biological functions from causing cell damage (oxidative distress) to cell signalling (oxidative eustress). The balance between the rate and amount of ROS generated and the capacity for scavenging systems to remove them is affected by several biological and environmental factors, including oxygen availability. Ectotherms, and in particular hypoxia-tolerant ectotherms, are hypothesized to avoid oxidative damage caused by hypoxia, although it is unclear whether this translates to an increase in ecological fitness. In this Review, we highlight the differences between oxidative distress and eustress, the current mechanistic understanding of the two and how they may affect ectothermic physiology. We discuss the evidence of occurrence of oxidative damage with hypoxia in ectotherms, and that ectotherms may avoid oxidative damage through (1) high levels of antioxidant and scavenging systems and/or (2) low(ering) levels of ROS generation. We argue that the disagreements in the literature as to how hypoxia affects antioxidant enzyme activity and the variable metabolism of ectotherms makes the latter strategy more amenable to ectotherm physiology. Finally, we argue that observed changes in ROS production and oxidative status with hypoxia may be a signalling mechanism and an adaptive strategy for ectotherms encountering hypoxia.
Collapse
Affiliation(s)
- Amanda Bundgaard
- University of Cologne, CECAD, Joseph-Stelzmann-Straße 26, DE-50931 Köln, Germany
- Aarhus University, Department of Biology, CF Moellers Alle 3, DK-8000 Aarhus C, Denmark
| | - Brittney G Borowiec
- Wilfrid Laurier University, Department of Biology, 75 University Ave. W., Waterloo, ON, Canada, N2L 3C5
| | - Gigi Y Lau
- University of British Columbia, Department of Zoology, 6270 University Blvd, Vancouver, BC, Canada, V6T 1Z4
| |
Collapse
|
17
|
Flores Carvalho M, Boteon YL, Guarrera JV, Modi PR, Lladó L, Lurje G, Kasahara M, Dutkowski P, Schlegel A. Obstacles to implement machine perfusion technology in routine clinical practice of transplantation: Why are we not there yet? Hepatology 2024; 79:713-730. [PMID: 37013926 DOI: 10.1097/hep.0000000000000394] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/05/2023] [Indexed: 04/05/2023]
Abstract
Machine perfusion of solid human organs is an old technique, and the basic principles were presented as early as 1855 by Claude Barnard. More than 50 years ago, the first perfusion system was used in clinical kidney transplantation. Despite the well-known benefits of dynamic organ preservation and significant medical and technical development in the last decades, perfusion devices are still not in routine use. This article describes the various challenges to implement this technology in practice, critically analyzing the role of all involved stakeholders, including clinicians, hospitals, regulatory, and industry, on the background of regional differences worldwide. The clinical need for this technology is discussed first, followed by the current status of research and the impact of costs and regulations. Considering the need for strong collaborations between clinical users, regulatory bodies, and industry, integrated road maps and pathways required to achieve a wider implementation are presented. The role of research development, clear regulatory pathways, and the need for more flexible reimbursement schemes is discussed together with potential solutions to address the most relevant hurdles. This article paints an overall picture of the current liver perfusion landscape and highlights the role of clinical, regulatory, and financial stakeholders worldwide.
Collapse
Affiliation(s)
- Mauricio Flores Carvalho
- Department of Clinical and Experimental Medicine, Hepatobiliary Unit, University of Florence, AOU Careggi, Florence, Italy
| | - Yuri L Boteon
- Liver Unit, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - James V Guarrera
- Division of Abdominal Transplant Surgery, Rutgers New Jersey Medical School, Department of Surgery, Newark, New Jersey, USA
| | - Pranjal R Modi
- Department of Transplantation Surgery, Institute of Kidney Diseases and Research Center and Dr. H L Trivedi Institute of Transplantation Sciences (IKDRC-ITS), Ahmedabad, India
| | - Laura Lladó
- Liver Transplant Unit, Hospital Universitario de Bellvitge, Barcelona, Spain
| | - Georg Lurje
- Department of Surgery, Campus Charité Mitte, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mureo Kasahara
- Transplantation Center, National Center for Child Health and Development, Tokyo, Japan
| | - Philipp Dutkowski
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Andrea Schlegel
- Department of Clinical and Experimental Medicine, Hepatobiliary Unit, University of Florence, AOU Careggi, Florence, Italy
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Center for Preclinical Research, 20100 Milan, Italy
| |
Collapse
|
18
|
Carlström M, Rannier Ribeiro Antonino Carvalho L, Guimaraes D, Boeder A, Schiffer TA. Dimethyl malonate preserves renal and mitochondrial functions following ischemia-reperfusion via inhibition of succinate dehydrogenase. Redox Biol 2024; 69:102984. [PMID: 38061207 PMCID: PMC10749277 DOI: 10.1016/j.redox.2023.102984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI), often experienced at the intensive care units, is associated with high morbidity/mortality where ischemia-reperfusion injury is a main causative factor. Succinate accumulation during ischemia contributes to the excessive generation of reactive oxygen species at reperfusion. Inhibition of succinate dehydrogenase has been associated with protective outcome in cardiac ischemia-reperfusion after 24h, but the effects on kidney and mitochondrial functions are less well studied. AIM To investigate the therapeutic potential of succinate dehydrogenase inhibition, by using dimethyl malonate (DMM), on kidney and mitochondria functions in a mouse model of AKI. METHODS Male C57BL/6J mice were pre-treated with DMM or placebo, i.p. 30min prior to bilateral renal ischemia (20min). After 3-days of reperfusion, glomerular filtration rate (GFR) was calculated from plasma clearance of FITC-inulin. Kidney mitochondria was isolated and mass specific and intrinsic mitochondrial function were evaluated by high resolution respirometry. Kidney sections were stained (i.e., hematoxylin-eosin and TUNEL) and analyzed for histopathological evaluation of injuries and apotosis, respectively. NADPH oxidase activity in kidney and human proximal tubular cell-line (HK2) were measured luminometrically. RESULTS DMM treatment improved GFR (p < 0.05) and reduced levels of blood urea nitrogen (p < 0.01) compared to untreated animals, which was associated with lower degree of ischemia-reperfusion-induced tubular injuries (P < 0.001) and apoptosis (P < 0.01). These therapeutic renal effects were linked with improved mitochondrial function, both mass-specific and intrinsic. Finally, DMM treatment prevented ischemia-reperfusion-induced NADPH oxidase activity in the kidney (p < 0.001), which was showed also in HK2 cells exposed to hypoxia and reoxygenation (P < 0.01). CONCLUSION Inhibition of succinate dehydrogenase with DMM, in conjunction with the ischemia-reperfusion phase, significantly improved both renal and mitochondrial functions. These findings may have clinical implications for future therapeutic strategies to prevent development of AKI and associated adverse complications, especially in high risk hospitalized patients.
Collapse
Affiliation(s)
- Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Drielle Guimaraes
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ariela Boeder
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Tomas A Schiffer
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
19
|
Rossignol G, Muller X, Brunet TA, Bidault V, Hervieu V, Clement Y, Ayciriex S, Mabrut JY, Salvador A, Mohkam K. Comprehensive bile acid pool analysis during ex-vivo liver perfusion in a porcine model of ischemia-reperfusion injury. Sci Rep 2024; 14:2384. [PMID: 38286808 PMCID: PMC10824768 DOI: 10.1038/s41598-024-52504-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024] Open
Abstract
Bile acids (BA) are key for liver regeneration and injury. This study aims at analyzing the changes in the BA pool induced by ischemia-reperfusion (IRI) and investigates the impact of hypothermic oxygenated perfusion (HOPE) on the BA pool compared to static cold storage (SCS). In a porcine model of IRI, liver grafts underwent 30 min of asystolic warm ischemia followed by 6 h of SCS (n = 6) ± 2 h of HOPE (n = 6) and 2 h of ex-situ warm reperfusion. The BA pool in bile samples was analyzed with liquid chromatography coupled with tandem mass spectrometry. We identified 16 BA and observed significant changes in response to ischemia-reperfusion, which were associated with both protective and injury mechanisms. Second, HOPE-treated liver grafts exhibited a more protective BA phenotype, characterized by a more hydrophilic BA pool compared to SCS. Key BA, such as GlycoCholic Acid, were identified and were associated with a decreased transaminase release and improved lactate clearance during reperfusion. Partial Least Square-Discriminant Analysis revealed a distinct injury profile for the HOPE group. In conclusion, the BA pool changes with liver graft IRI, and preservation with HOPE results in a protective BA phenotype compared to SCS.
Collapse
Affiliation(s)
- Guillaume Rossignol
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Lyon, France.
- Department of Pediatric Surgery and Liver Transplantation, Femme Mere Enfant University Hospital, Lyon, France.
- The Cancer Research Center of Lyon, INSERM U1052, Lyon, France.
- ED 340 BMIC, Claude Bernard Lyon 1 University, Villeurbanne, France.
- Institute of Analytical Sciences, CNRS UMR 5280, Claude Bernard University Lyon 1, Villeurbanne, France.
| | - Xavier Muller
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Lyon, France.
- The Cancer Research Center of Lyon, INSERM U1052, Lyon, France.
- ED 340 BMIC, Claude Bernard Lyon 1 University, Villeurbanne, France.
| | - Thomas Alexandre Brunet
- Institute of Analytical Sciences, CNRS UMR 5280, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Valeska Bidault
- Department of Pediatric Surgery and Liver Transplantation, Femme Mere Enfant University Hospital, Lyon, France
| | - Valerie Hervieu
- Department of Pathology, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Villeurbanne, Lyon, France
| | - Yohann Clement
- Institute of Analytical Sciences, CNRS UMR 5280, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Sophie Ayciriex
- Institute of Analytical Sciences, CNRS UMR 5280, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Jean-Yves Mabrut
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Lyon, France
- The Cancer Research Center of Lyon, INSERM U1052, Lyon, France
| | - Arnaud Salvador
- Institute of Analytical Sciences, CNRS UMR 5280, Claude Bernard University Lyon 1, Villeurbanne, France
| | - Kayvan Mohkam
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Lyon, France
- Department of Pediatric Surgery and Liver Transplantation, Femme Mere Enfant University Hospital, Lyon, France
- The Cancer Research Center of Lyon, INSERM U1052, Lyon, France
| |
Collapse
|
20
|
Chullo G, Panisello-Rosello A, Marquez N, Colmenero J, Brunet M, Pera M, Rosello-Catafau J, Bataller R, García-Valdecasas JC, Fundora Y. Focusing on Ischemic Reperfusion Injury in the New Era of Dynamic Machine Perfusion in Liver Transplantation. Int J Mol Sci 2024; 25:1117. [PMID: 38256190 PMCID: PMC10816079 DOI: 10.3390/ijms25021117] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Liver transplantation is the most effective treatment for end-stage liver disease. Transplant indications have been progressively increasing, with a huge discrepancy between the supply and demand of optimal organs. In this context, the use of extended criteria donor grafts has gained importance, even though these grafts are more susceptible to ischemic reperfusion injury (IRI). Hepatic IRI is an inherent and inevitable consequence of all liver transplants; it involves ischemia-mediated cellular damage exacerbated upon reperfusion and its severity directly affects graft function and post-transplant complications. Strategies for organ preservation have been constantly improving since they first emerged. The current gold standard for preservation is perfusion solutions and static cold storage. However, novel approaches that allow extended preservation times, organ evaluation, and their treatment, which could increase the number of viable organs for transplantation, are currently under investigation. This review discusses the mechanisms associated with IRI, describes existing strategies for liver preservation, and emphasizes novel developments and challenges for effective organ preservation and optimization.
Collapse
Affiliation(s)
- Gabriela Chullo
- Service of Digestive, Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (G.C.); (M.P.); (J.C.G.-V.)
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| | - Arnau Panisello-Rosello
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| | - Noel Marquez
- Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain;
| | - Jordi Colmenero
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
- Liver Transplant Unit, Service of Hepatology, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepaticas y digestives (CIBERehd), University of Barcelona, 08036 Barcelona, Spain
| | - Merce Brunet
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
- Centro de Investigación Biomédica en Red de Enfermedades hepaticas y digestives (CIBERehd), University of Barcelona, 08036 Barcelona, Spain
- Pharmacology and Toxicology Laboratory, Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, Hospital Clinic of Barcelona, 08036 Barcelona, Spain
| | - Miguel Pera
- Service of Digestive, Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (G.C.); (M.P.); (J.C.G.-V.)
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| | - Joan Rosello-Catafau
- Experimental Pathology, Institut d’Investigacions Biomèdiques de Barcelona-Consejo Superior de Investigaciones Científicas (IBB-CSIC), 08036 Barcelona, Spain;
| | - Ramon Bataller
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
- Liver Transplant Unit, Service of Hepatology, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepaticas y digestives (CIBERehd), University of Barcelona, 08036 Barcelona, Spain
| | - Juan Carlos García-Valdecasas
- Service of Digestive, Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (G.C.); (M.P.); (J.C.G.-V.)
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| | - Yiliam Fundora
- Service of Digestive, Hepato-Pancreatico-Biliary and Liver Transplant Surgery, Institut Clínic de Malalties Digestives i Metabòliques (ICMDM), Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (G.C.); (M.P.); (J.C.G.-V.)
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain; (J.C.); (M.B.); (R.B.)
| |
Collapse
|
21
|
Muller X, Rossignol G, Couillerot J, Breton A, Hervieu V, Lesurtel M, Mohkam K, Mabrut JY. A Single Preservation Solution for Static Cold Storage and Hypothermic Oxygenated Perfusion of Marginal Liver Grafts: A Preclinical Study. Transplantation 2024; 108:175-183. [PMID: 37410580 DOI: 10.1097/tp.0000000000004714] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
BACKGROUND Hypothermic oxygenated perfusion (HOPE) improves outcomes of marginal liver grafts. However, to date, no preservation solution exists for both static cold storage (SCS) and HOPE. METHODS After 30 min of asystolic warm ischemia, porcine livers underwent 6 h of SCS followed by 2 h of HOPE. Liver grafts were either preserved with a single preservation solution (IGL2) designed for SCS and HOPE (IGL2-Machine Perfusion Solution [MPS] group, n = 6) or with the gold-standard University of Wisconsin designed for for SCS and Belzer MPS designed for HOPE (MPS group, n = 5). All liver grafts underwent warm reperfusion with whole autologous blood for 2 h, and surrogate markers of hepatic ischemia-reperfusion injury (IRI) were assessed in the hepatocyte, cholangiocyte, vascular, and immunological compartments. RESULTS After 2 h of warm reperfusion, livers in the IGL2-MPS group showed no significant differences in transaminase release (aspartate aminotransferase: 65.58 versus 104.9 UI/L/100 g liver; P = 0.178), lactate clearance, and histological IRI compared with livers in the MPS group. There were no significant differences in biliary acid composition, bile production, and histological biliary IRI. Mitochondrial and endothelial damage was also not significantly different and resulted in similar hepatic inflammasome activation. CONCLUSIONS This preclinical study shows that a novel IGL2 allows for the safe preservation of marginal liver grafts with SCS and HOPE. Hepatic IRI was comparable with the current gold standard of combining 2 different preservation solutions (University of Wisconsin + Belzer MPS). These data pave the way for a phase I first-in-human study and it is a first step toward tailored preservation solutions for machine perfusion of liver grafts.
Collapse
Affiliation(s)
- Xavier Muller
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
- Hepatology Institute of Lyon, INSERM U1052, Lyon, France
- Ecole Doctorale 340, Biologie Moléculaire et Intégrative, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Guillaume Rossignol
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
- Hepatology Institute of Lyon, INSERM U1052, Lyon, France
- Ecole Doctorale 340, Biologie Moléculaire et Intégrative, Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Joris Couillerot
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
- Hepatology Institute of Lyon, INSERM U1052, Lyon, France
| | - Antoine Breton
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
- Hepatology Institute of Lyon, INSERM U1052, Lyon, France
| | - Valérie Hervieu
- Department of Pathology, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, Villeurbanne, Lyon, France
| | - Mickaël Lesurtel
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
| | - Kayvan Mohkam
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
- Hepatology Institute of Lyon, INSERM U1052, Lyon, France
| | - Jean-Yves Mabrut
- Department of General Surgery and Liver Transplantation, Croix Rousse University Hospital, Hospices Civils de Lyon, University of Lyon I, Lyon, France
- Hepatology Institute of Lyon, INSERM U1052, Lyon, France
| |
Collapse
|
22
|
Lopez-Schenk R, Collins NL, Schenk NA, Beard DA. Integrated Functions of Cardiac Energetics, Mechanics, and Purine Nucleotide Metabolism. Compr Physiol 2023; 14:5345-5369. [PMID: 38158366 PMCID: PMC10956446 DOI: 10.1002/cphy.c230011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Purine nucleotides play central roles in energy metabolism in the heart. Most fundamentally, the free energy of hydrolysis of the adenine nucleotide adenosine triphosphate (ATP) provides the thermodynamic driving force for numerous cellular processes including the actin-myosin crossbridge cycle. Perturbations to ATP supply and/or demand in the myocardium lead to changes in the homeostatic balance between purine nucleotide synthesis, degradation, and salvage, potentially affecting myocardial energetics and, consequently, myocardial mechanics. Indeed, both acute myocardial ischemia and decompensatory remodeling of the myocardium in heart failure are associated with depletion of myocardial adenine nucleotides and with impaired myocardial mechanical function. Yet there remain gaps in the understanding of mechanistic links between adenine nucleotide degradation and contractile dysfunction in heart disease. The scope of this article is to: (i) review current knowledge of the pathways of purine nucleotide depletion and salvage in acute ischemia and in chronic heart disease; (ii) review hypothesized mechanisms linking myocardial mechanics and energetics with myocardial adenine nucleotide regulation; and (iii) highlight potential targets for treating myocardial metabolic and mechanical dysfunction associated with these pathways. It is hypothesized that an imbalance in the degradation, salvage, and synthesis of adenine nucleotides leads to a net loss of adenine nucleotides in both acute ischemia and under chronic high-demand conditions associated with the development of heart failure. This reduction in adenine nucleotide levels results in reduced myocardial ATP and increased myocardial inorganic phosphate. Both of these changes have the potential to directly impact tension development and mechanical work at the cellular level. © 2024 American Physiological Society. Compr Physiol 14:5345-5369, 2024.
Collapse
Affiliation(s)
- Rachel Lopez-Schenk
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicole L Collins
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Noah A Schenk
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel A Beard
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
23
|
Eden J, Breuer E, Birrer D, Müller M, Pfister M, Mayr H, Sun K, Widmer J, Huwyler F, Ungethüm U, Humar B, Gupta A, Schiess S, Wendt M, Immer F, Elmer A, Meierhofer D, Schlegel A, Dutkowski P. Screening for mitochondrial function before use-routine liver assessment during hypothermic oxygenated perfusion impacts liver utilization. EBioMedicine 2023; 98:104857. [PMID: 37918219 PMCID: PMC10641151 DOI: 10.1016/j.ebiom.2023.104857] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/15/2023] [Accepted: 10/15/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND To report on a concept of liver assessment during ex situ hypothermic oxygenated perfusion (HOPE) and its significant impact on liver utilization. METHODS An analysis of prospectively collected data on donation after circulatory death (DCD) livers, treated by HOPE at our institution, during a 11-year period between January 2012 and December 2022. FINDINGS Four hundred and fifteen DCD Maastricht III livers were offered during the study period in Switzerland, resulting in 249 liver transplants. Of those, we performed 158 DCD III liver transplants at our institution, with 1-year patient survival and death censored graft survival (death with functioning graft) of 87 and 89%, respectively, thus comparable to benchmark graft survivals of ideal DBD and DCD liver transplants (89% and 86%). Correspondingly, graft loss for primary non-function or cholangiopathy was overall low, i.e., 7/158 (4.4%) and 11/158 (6.9%), despite more than 82% of DCD liver grafts ranked high (6-10 points) or futile risk (>10 points) according to the UK-DCD score. Consistently, death censored graft survival was not different between low-, high-risk or futile DCD III livers. The key behind these achievements was the careful development and implementation of a routine perfusate assessment of mitochondrial biomarkers for injury and function, i.e., release of flavin mononucleotide from complex I, perfusate NADH, and mitochondrial CO2 production during HOPE, allowing a more objective interpretation of liver quality on a subcellular level, compared to donor derived data. INTERPRETATION HOPE after cold storage is a highly suitable and easy to perform perfusion approach, which allows reliable liver graft assessment, enabling surgeons to make a fact based decision on whether or not to implant the organ. HOPE-treatment should be combined with viability assessment particularly when used for high-risk organs, including DCD livers or organs with relevant steatosis. FUNDING This study was supported by the Swiss National Foundation (SNF) grant 320030_189055/1 to PD.
Collapse
Affiliation(s)
- Janina Eden
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Eva Breuer
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Dominique Birrer
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Matteo Müller
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Matthias Pfister
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Hemma Mayr
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Keyue Sun
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Jeannette Widmer
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Florian Huwyler
- Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Udo Ungethüm
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Bostjan Humar
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Anurag Gupta
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Stefanie Schiess
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Martin Wendt
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| | - Franz Immer
- Swisstransplant, The Swiss National Foundation for Organ Donation and Transplantation, Effingerstrasse 1, Bern 3011, Switzerland
| | - Andreas Elmer
- Swisstransplant, The Swiss National Foundation for Organ Donation and Transplantation, Effingerstrasse 1, Bern 3011, Switzerland
| | - David Meierhofer
- Max Planck Institute for Molecular Genetics, Mass Spectrometry Facility, Berlin 14195, Germany
| | - Andrea Schlegel
- Transplantation Center, Digestive Disease and Surgery Institute and Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Philipp Dutkowski
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland.
| |
Collapse
|
24
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
25
|
Lu W, Park NR, TeSlaa T, Jankowski CS, Samarah L, McReynolds M, Xing X, Schembri J, Woolf MT, Rabinowitz JD, Davidson SM. Acidic Methanol Treatment Facilitates Matrix-Assisted Laser Desorption Ionization-Mass Spectrometry Imaging of Energy Metabolism. Anal Chem 2023; 95:14879-14888. [PMID: 37756255 PMCID: PMC10568533 DOI: 10.1021/acs.analchem.3c01875] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023]
Abstract
Detection of small molecule metabolites (SMM), particularly those involved in energy metabolism using MALDI-mass spectrometry imaging (MSI), is challenging due to factors including ion suppression from other analytes present (e.g., proteins and lipids). One potential solution to enhance SMM detection is to remove analytes that cause ion suppression from tissue sections before matrix deposition through solvent washes. Here, we systematically investigated solvent treatment conditions to improve SMM signal and preserve metabolite localization. Washing with acidic methanol significantly enhances the detection of phosphate-containing metabolites involved in energy metabolism. The improved detection is due to removing lipids and highly polar metabolites that cause ion suppression and denaturing proteins that release bound phosphate-containing metabolites. Stable isotope infusions of [13C6]nicotinamide coupled to MALDI-MSI ("Iso-imaging") in the kidney reveal patterns that indicate blood vessels, medulla, outer stripe, and cortex. We also observed different ATP:ADP raw signals across mouse kidney regions, consistent with regional differences in glucose metabolism favoring either gluconeogenesis or glycolysis. In mouse muscle, Iso-imaging using [13C6]glucose shows high glycolytic flux from infused circulating glucose in type 1 and 2a fibers (soleus) and relatively lower glycolytic flux in type 2b fiber type (gastrocnemius). Thus, improved detection of phosphate-containing metabolites due to acidic methanol treatment combined with isotope tracing provides an improved way to probe energy metabolism with spatial resolution in vivo.
Collapse
Affiliation(s)
- Wenyun Lu
- Lewis
Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, United States
- Department
of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Noel R. Park
- Lewis
Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, United States
| | - Tara TeSlaa
- Department
of Molecular and Medical Pharmacology, University
of California Los Angeles, Los Angeles, California 90095, United States
| | - Connor S.R. Jankowski
- Lewis
Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, United States
| | - Laith Samarah
- Lewis
Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, United States
| | - Melanie McReynolds
- Department
of Biochemistry and Molecular Biology, The
Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Xi Xing
- Lewis
Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, United States
- Department
of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Jessica Schembri
- Lewis
Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, United States
| | - Morgan T. Woolf
- Department
of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Joshua D. Rabinowitz
- Lewis
Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, United States
- Rutgers
Cancer Institute of New Jersey (CINJ), Rutgers
University, New Brunswick, New Jersey 08901, United States
- Department
of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
- Ludwig
Institute for Cancer Research, Princeton
University, Princeton, New Jersey 08544, United States
| | - Shawn M. Davidson
- Lewis
Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, United States
- Rutgers
Cancer Institute of New Jersey (CINJ), Rutgers
University, New Brunswick, New Jersey 08901, United States
| |
Collapse
|
26
|
Tokarska-Schlattner M, Zeaiter N, Cunin V, Attia S, Meunier C, Kay L, Achouri A, Hiriart-Bryant E, Couturier K, Tellier C, El Harras A, Elena-Herrmann B, Khochbin S, Le Gouellec A, Schlattner U. Multi-Method Quantification of Acetyl-Coenzyme A and Further Acyl-Coenzyme A Species in Normal and Ischemic Rat Liver. Int J Mol Sci 2023; 24:14957. [PMID: 37834405 PMCID: PMC10573920 DOI: 10.3390/ijms241914957] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/29/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
Thioesters of coenzyme A (CoA) carrying different acyl chains (acyl-CoAs) are central intermediates of many metabolic pathways and donor molecules for protein lysine acylation. Acyl-CoA species largely differ in terms of cellular concentrations and physico-chemical properties, rendering their analysis challenging. Here, we compare several approaches to quantify cellular acyl-CoA concentrations in normal and ischemic rat liver, using HPLC and LC-MS/MS for multi-acyl-CoA analysis, as well as NMR, fluorimetric and spectrophotometric techniques for the quantification of acetyl-CoAs. In particular, we describe a simple LC-MS/MS protocol that is suitable for the relative quantification of short and medium-chain acyl-CoA species. We show that ischemia induces specific changes in the short-chain acyl-CoA relative concentrations, while mild ischemia (1-2 min), although reducing succinyl-CoA, has little effects on acetyl-CoA, and even increases some acyl-CoA species upstream of the tricarboxylic acid cycle. In contrast, advanced ischemia (5-6 min) also reduces acetyl-CoA levels. Our approach provides the keys to accessing the acyl-CoA metabolome for a more in-depth analysis of metabolism, protein acylation and epigenetics.
Collapse
Affiliation(s)
- Malgorzata Tokarska-Schlattner
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Nour Zeaiter
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Valérie Cunin
- University Grenoble Alpes, CNRS UMR 5525, Laboratory TIMC—Translational Microbiology, Evolution, Engineering (TREE), Service de Biochimie, Biologie Moléculaire et Toxicologie Environnementale, CHU Grenoble-Alpes, 38058 Grenoble, France; (V.C.); (C.M.); (A.L.G.)
| | - Stéphane Attia
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Cécile Meunier
- University Grenoble Alpes, CNRS UMR 5525, Laboratory TIMC—Translational Microbiology, Evolution, Engineering (TREE), Service de Biochimie, Biologie Moléculaire et Toxicologie Environnementale, CHU Grenoble-Alpes, 38058 Grenoble, France; (V.C.); (C.M.); (A.L.G.)
| | - Laurence Kay
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Amel Achouri
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Edwige Hiriart-Bryant
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Karine Couturier
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Cindy Tellier
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
| | - Abderrafek El Harras
- University Grenoble Alpes, Inserm U1209 and CNRS UMR5309, Institute for Advanced Biosciences (IAB), 38058 Grenoble, France; (A.E.H.); (B.E.-H.); (S.K.)
| | - Bénédicte Elena-Herrmann
- University Grenoble Alpes, Inserm U1209 and CNRS UMR5309, Institute for Advanced Biosciences (IAB), 38058 Grenoble, France; (A.E.H.); (B.E.-H.); (S.K.)
| | - Saadi Khochbin
- University Grenoble Alpes, Inserm U1209 and CNRS UMR5309, Institute for Advanced Biosciences (IAB), 38058 Grenoble, France; (A.E.H.); (B.E.-H.); (S.K.)
| | - Audrey Le Gouellec
- University Grenoble Alpes, CNRS UMR 5525, Laboratory TIMC—Translational Microbiology, Evolution, Engineering (TREE), Service de Biochimie, Biologie Moléculaire et Toxicologie Environnementale, CHU Grenoble-Alpes, 38058 Grenoble, France; (V.C.); (C.M.); (A.L.G.)
| | - Uwe Schlattner
- University Grenoble Alpes, Inserm U1055, Laboratory of Fundamental and Applied Bioenergetics (LBFA), 38058 Grenoble, France; (N.Z.); (S.A.); (L.K.); (A.A.); (E.H.-B.); (K.C.); (C.T.)
- Institut Universitaire de France (IUF), 75231 Paris, France
| |
Collapse
|
27
|
Wang L, Qiu S, Li X, Zhang Y, Huo M, Shi J. Myocardial-Targeting Tannic Cerium Nanocatalyst Attenuates Ischemia/Reperfusion Injury. Angew Chem Int Ed Engl 2023; 62:e202305576. [PMID: 37368480 DOI: 10.1002/anie.202305576] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 06/28/2023]
Abstract
Ischemic heart disease (IHD) is one of the leading causes of death worldwide. Medications or surgery have been considered as effective protocols to treat IHD for decades. Yet the reperfusion of the blood flow frequently leads to the generation of excessive reactive oxygen species (ROS), causing prominent and irreversible damage to the cardiomyocytes. In the present work, tannic acid-assembled tetravalent cerium (TA-Ce) nanocatalysts with appealing cardiomyocyte-targeting and antioxidation capability have been synthesized and applied for the effective and biocompatible ischemia/reperfusion injury therapeutics. TA-Ce nanocatalysts could effectively rescue the cardiomyocytes from oxidative stress induced by H2 O2 challenge as well as oxygen-glucose deprivation in vitro. In the murine ischemia/reperfusion model, cardiac accumulation and intracellular ROS scavenging could be achieved against the pathology, substantially reducing the myocardial infarct area and recovering heart functionality. This work illuminates the design of nanocatalytic metal complexes and their therapeutic prospects in ischemic heart diseases with high effectiveness and biocompatibility, paving the way for the clinical translation from bench to bedside.
Collapse
Affiliation(s)
- Liping Wang
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Shuwen Qiu
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Xi Li
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yabing Zhang
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Minfeng Huo
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Jianlin Shi
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences (2021RU012), Shanghai, 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| |
Collapse
|
28
|
Wancewicz B, Pergande MR, Zhu Y, Gao Z, Shi Z, Plouff K, Ge Y. Comprehensive Metabolomic Analysis of Human Heart Tissue Enabled by Parallel Metabolite Extraction and High-Resolution Mass Spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.15.558013. [PMID: 37745334 PMCID: PMC10516009 DOI: 10.1101/2023.09.15.558013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The heart contracts incessantly and requires a constant supply of energy, utilizing numerous metabolic substrates such as fatty acids, carbohydrates, lipids, and amino acids to supply its high energy demands. Therefore, a comprehensive analysis of various metabolites is urgently needed for understanding cardiac metabolism; however, complete metabolome analyses remain challenging due to the broad range of metabolite polarities which makes extraction and detection difficult. Herein, we implemented parallel metabolite extractions and high-resolution mass spectrometry (MS)-based methods to obtain a comprehensive analysis of the human heart metabolome. To capture the diverse range of metabolite polarities, we first performed six parallel liquid-liquid extractions (three monophasic, two biphasic, and one triphasic extractions) of healthy human donor heart tissue. Next, we utilized two complementary MS platforms for metabolite detection - direct-infusion ultrahigh-resolution Fourier-transform ion cyclotron resonance (DI-FTICR) and high-resolution liquid chromatography quadrupole time-of-flight tandem MS (LC-Q-TOF MS/MS). Using DI-FTICR MS, 9,521 metabolic features were detected where 7,699 were assigned a chemical formula and 1,756 were assigned an annotated by accurate mass assignment. Using LC-Q-TOF MS/MS, 21,428 metabolic features were detected where 626 metabolites were identified based on fragmentation matching against publicly available libraries. Collectively, 2276 heart metabolites were identified in this study which span a wide range of polarities including polar (benzenoids, alkaloids and derivatives and nucleosides) as well as non-polar (phosphatidylcholines, acylcarnitines, and fatty acids) compounds. The results of this study will provide critical knowledge regarding the selection of appropriate extraction and MS detection methods for the analysis of the diverse classes of human heart metabolites.
Collapse
Affiliation(s)
- Benjamin Wancewicz
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Melissa R. Pergande
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Yanlong Zhu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Zhan Gao
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Zhuoxin Shi
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Kylie Plouff
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| |
Collapse
|
29
|
Tingle SJ, Dobbins JJ, Thompson ER, Figueiredo RS, Mahendran B, Pandanaboyana S, Wilson C. Machine perfusion in liver transplantation. Cochrane Database Syst Rev 2023; 9:CD014685. [PMID: 37698189 PMCID: PMC10496129 DOI: 10.1002/14651858.cd014685.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
BACKGROUND Liver transplantation is the only chance of cure for people with end-stage liver disease and some people with advanced liver cancers or acute liver failure. The increasing prevalence of these conditions drives demand and necessitates the increasing use of donated livers which have traditionally been considered suboptimal. Several novel machine perfusion preservation technologies have been developed, which attempt to ameliorate some of the deleterious effects of ischaemia reperfusion injury. Machine perfusion technology aims to improve organ quality, thereby improving outcomes in recipients of suboptimal livers when compared to traditional static cold storage (SCS; ice box). OBJECTIVES To evaluate the effects of different methods of machine perfusion (including hypothermic oxygenated machine perfusion (HOPE), normothermic machine perfusion (NMP), controlled oxygenated rewarming, and normothermic regional perfusion) versus each other or versus static cold storage (SCS) in people undergoing liver transplantation. SEARCH METHODS We used standard, extensive Cochrane search methods. The latest search date was 10 January 2023. SELECTION CRITERIA We included randomised clinical trials which compared different methods of machine perfusion, either with each other or with SCS. Studies comparing HOPE via both hepatic artery and portal vein, or via portal vein only, were grouped. The protocol detailed that we also planned to include quasi-randomised studies to assess treatment harms. DATA COLLECTION AND ANALYSIS We used standard Cochrane methods. Our primary outcomes were 1. overall participant survival, 2. quality of life, and 3. serious adverse events. Secondary outcomes were 4. graft survival, 5. ischaemic biliary complications, 6. primary non-function of the graft, 7. early allograft function, 8. non-serious adverse events, 9. transplant utilisation, and 10. transaminase release during the first week post-transplant. We assessed bias using Cochrane's RoB 2 tool and used GRADE to assess certainty of evidence. MAIN RESULTS We included seven randomised trials (1024 transplant recipients from 1301 randomised/included livers). All trials were parallel two-group trials; four compared HOPE versus SCS, and three compared NMP versus SCS. No trials used normothermic regional perfusion. When compared with SCS, it was uncertain whether overall participant survival was improved with either HOPE (hazard ratio (HR) 0.91, 95% confidence interval (CI) 0.42 to 1.98; P = 0.81, I2 = 0%; 4 trials, 482 recipients; low-certainty evidence due to imprecision because of low number of events) or NMP (HR 1.08, 95% CI 0.31 to 3.80; P = 0.90; 1 trial, 222 recipients; very low-certainty evidence due to imprecision and risk of bias). No trials reported quality of life. When compared with SCS alone, HOPE was associated with improvement in the following clinically relevant outcomes: graft survival (HR 0.45, 95% CI 0.23 to 0.87; P = 0.02, I2 = 0%; 4 trials, 482 recipients; high-certainty evidence), serious adverse events in extended criteria DBD liver transplants (OR 0.45, 95% CI 0.22 to 0.91; P = 0.03, I2 = 0%; 2 trials, 156 participants; moderate-certainty evidence) and clinically significant ischaemic cholangiopathy in recipients of DCD livers (OR 0.31, 95% CI 0.11 to 0.92; P = 0.03; 1 trial, 156 recipients; high-certainty evidence). In contrast, NMP was not associated with improvement in any of these clinically relevant outcomes. NMP was associated with improved utilisation compared with SCS (one trial found a 50% lower rate of organ discard; P = 0.008), but the reasons underlying this effect are unknown. We identified 11 ongoing studies investigating machine perfusion technologies. AUTHORS' CONCLUSIONS In situations where the decision has been made to transplant a liver donated after circulatory death or donated following brain death, end-ischaemic HOPE will provide superior clinically relevant outcomes compared with SCS alone. Specifically, graft survival is improved (high-certainty evidence), serious adverse events are reduced (moderate-certainty evidence), and in donors after circulatory death, clinically relevant ischaemic biliary complications are reduced (high-certainty evidence). There is no good evidence that NMP has the same benefits over SCS in terms of these clinically relevant outcomes. NMP does appear to improve utilisation of grafts that would otherwise be discarded with SCS; however, the reasons for this, and whether this effect is specific to NMP, is not clear. Further studies into NMP viability criteria and utilisation, as well as head-to-head trials with other perfusion technologies are needed. In the setting of donation following circulatory death transplantation, further trials are needed to assess the effect of these ex situ machine perfusion methods against, or in combination with, normothermic regional perfusion.
Collapse
Affiliation(s)
- Samuel J Tingle
- NIHR Blood and Transplant Research Unit, Newcastle University and Cambridge University, Newcastle upon Tyne, UK
| | | | - Emily R Thompson
- Institute of Transplantation, The Freeman Hospital, Newcastle upon Tyne, UK
| | | | | | - Sanjay Pandanaboyana
- HPB and Liver Transplant Surgery, Freeman Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Colin Wilson
- Institute of Transplantation, The Freeman Hospital, Newcastle upon Tyne, UK
| |
Collapse
|
30
|
Maspero M, Ali K, Cazzaniga B, Yilmaz S, Raj R, Liu Q, Quintini C, Miller C, Hashimoto K, Fairchild RL, Schlegel A. Acute rejection after liver transplantation with machine perfusion versus static cold storage: A systematic review and meta-analysis. Hepatology 2023; 78:835-846. [PMID: 36988381 DOI: 10.1097/hep.0000000000000363] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/27/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND AND AIMS Acute cellular rejection (ACR) is a frequent complication after liver transplantation. By reducing ischemia and graft damage, dynamic preservation techniques may diminish ACR. We performed a systematic review to assess the effect of currently tested organ perfusion (OP) approaches versus static cold storage (SCS) on post-transplant ACR-rates. APPROACH AND RESULTS A systematic search of Medline, Embase, Cochrane Library, and Web of Science was conducted. Studies reporting ACR-rates between OP and SCS and comprising at least 10 liver transplants performed with either hypothermic oxygenated perfusion (HOPE), normothermic machine perfusion, or normothermic regional perfusion were included. Studies with mixed perfusion approaches were excluded. Eight studies were identified (226 patients in OP and 330 in SCS). Six studies were on HOPE, one on normothermic machine perfusion, and one on normothermic regional perfusion. At meta-analysis, OP was associated with a reduction in ACR compared with SCS [OR: 0.55 (95% CI, 0.33-0.91), p =0.02]. This effect remained significant when considering HOPE alone [OR: 0.54 (95% CI, 0.29-1), p =0.05], in a subgroup analysis of studies including only grafts from donation after cardiac death [OR: 0.43 (0.20-0.91) p =0.03], and in HOPE studies with only donation after cardiac death grafts [OR: 0.37 (0.14-1), p =0.05]. CONCLUSIONS Dynamic OP techniques are associated with a reduction in ACR after liver transplantation compared with SCS. PROSPERO registration: CRD42022348356.
Collapse
Affiliation(s)
- Marianna Maspero
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
- University of Milan, Università degli Studi di Milano, Milan, Italy
| | - Khaled Ali
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Beatrice Cazzaniga
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Sumeyye Yilmaz
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Roma Raj
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Qiang Liu
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Cristiano Quintini
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
- Department of Gastroenterology and Hepatology, Digestive Disease Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Charles Miller
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Koji Hashimoto
- Transplantation Center, Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, Ohio
| | - Robert L Fairchild
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Andrea Schlegel
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Centre of Preclinical Research, Milan, Italy
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, Switzerland
| |
Collapse
|
31
|
Prag HA, Murphy MP, Krieg T. Preventing mitochondrial reverse electron transport as a strategy for cardioprotection. Basic Res Cardiol 2023; 118:34. [PMID: 37639068 PMCID: PMC10462584 DOI: 10.1007/s00395-023-01002-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/29/2023]
Abstract
In the context of myocardial infarction, the burst of superoxide generated by reverse electron transport (RET) at complex I in mitochondria is a crucial trigger for damage during ischaemia/reperfusion (I/R) injury. Here we outline the necessary conditions for superoxide production by RET at complex I and how it can occur during reperfusion. In addition, we explore various pathways that are implicated in generating the conditions for RET to occur and suggest potential therapeutic strategies to target RET, aiming to achieve cardioprotection.
Collapse
Affiliation(s)
- Hiran A Prag
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Michael P Murphy
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, CB2 0XY, UK.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
32
|
Jones RE, Gruszczyk AV, Schmidt C, Hammersley DJ, Mach L, Lee M, Wong J, Yang M, Hatipoglu S, Lota AS, Barnett SN, Toscano-Rivalta R, Owen R, Raja S, De Robertis F, Smail H, De-Souza A, Stock U, Kellman P, Griffin J, Dumas ME, Martin JL, Saeb-Parsy K, Vazir A, Cleland JGF, Pennell DJ, Bhudia SK, Halliday BP, Noseda M, Frezza C, Murphy MP, Prasad SK. Assessment of left ventricular tissue mitochondrial bioenergetics in patients with stable coronary artery disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:733-745. [PMID: 38666037 PMCID: PMC11041759 DOI: 10.1038/s44161-023-00312-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 06/29/2023] [Indexed: 04/28/2024]
Abstract
Recurrent myocardial ischemia can lead to left ventricular (LV) dysfunction in patients with coronary artery disease (CAD). In this observational cohort study, we assessed for chronic metabolomic and transcriptomic adaptations within LV myocardium of patients undergoing coronary artery bypass grafting. During surgery, paired transmural LV biopsies were acquired on the beating heart from regions with and without evidence of inducible ischemia on preoperative stress perfusion cardiovascular magnetic resonance. From 33 patients, 63 biopsies were acquired, compared to analysis of LV samples from 11 donor hearts. The global myocardial adenosine triphosphate (ATP):adenosine diphosphate (ADP) ratio was reduced in patients with CAD as compared to donor LV tissue, with increased expression of oxidative phosphorylation (OXPHOS) genes encoding the electron transport chain complexes across multiple cell types. Paired analyses of biopsies obtained from LV segments with or without inducible ischemia revealed no significant difference in the ATP:ADP ratio, broader metabolic profile or expression of ventricular cardiomyocyte genes implicated in OXPHOS. Differential metabolite analysis suggested dysregulation of several intermediates in patients with reduced LV ejection fraction, including succinate. Overall, our results suggest that viable myocardium in patients with stable CAD has global alterations in bioenergetic and transcriptional profile without large regional differences between areas with or without inducible ischemia.
Collapse
Affiliation(s)
- Richard E. Jones
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
- Anglia Ruskin University, Chelmsford, UK
- Essex Cardiothoracic Centre, Basildon, UK
| | - Anja V. Gruszczyk
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | | | - Daniel J. Hammersley
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Lukas Mach
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Michael Lee
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Joyce Wong
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Ming Yang
- MRC Cancer Unit, University of Cambridge, Cambridge, UK
- University of Cologne, CECAD, Cologne, Germany
| | - Suzan Hatipoglu
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Amrit S. Lota
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Sam N. Barnett
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Ruth Owen
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Shahzad Raja
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Fabio De Robertis
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Hassiba Smail
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Anthony De-Souza
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Ulrich Stock
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Peter Kellman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA
| | - Julian Griffin
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Marc-Emmanuel Dumas
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- European Genomic Institute of Diabetes, INSERM U1283, CNRS 8199, Institut Pasteur de Lille, Lille University Hospital, University of Lille, Lille, France
- McGill Genome Centre, McGill University, Montréal, QC Canada
| | - Jack L. Martin
- Department of Surgery and Cambridge NIHR Biomedical Research Centre, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery and Cambridge NIHR Biomedical Research Centre, Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Ali Vazir
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | | | - Dudley J. Pennell
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Sunil K. Bhudia
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Brian P. Halliday
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Sanjay K. Prasad
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| |
Collapse
|
33
|
Hautbergue T, Laverdure F, Van SD, Vallee A, Sanchis-Borja M, Decante B, Gaillard M, Junot C, Fenaille F, Mercier O, Colsch B, Guihaire J. Metabolomic profiling of cardiac allografts after controlled circulatory death. J Heart Lung Transplant 2023; 42:870-879. [PMID: 36931989 DOI: 10.1016/j.healun.2023.02.1492] [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: 09/08/2022] [Revised: 01/17/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Assessment of myocardial viability during ex situ heart perfusion (ESHP) is based on the measurement of lactate concentrations. As this provides with limited information, we sought to investigate the metabolic signature associated with donation after circulatory death (DCD) and the impact of ESHP on the myocardial metabolome. METHODS Porcine hearts were retrieved either after warm ischemia (DCD group, N = 6); after brain-stem death (BSD group, N = 6); or without DCD nor BSD (Control group, N = 6). Hearts were perfused using normothermic oxygenated blood for 240 minutes. Plasma and myocardial samples were collected respectively every 30 and 60 minutes, and analyzed by an untargeted metabolomic approach using liquid chromatography coupled to high-resolution mass spectrometry. RESULTS Median duration of warm ischemia was 23 minutes [19-29] in DCD animals. Lactate level within myocardial biopsies was not significantly different between groups at T0 (p = 0.281), and remained stable over the 4-hour period of ESHP. More than 300 metabolites were detected in plasma and heart biopsy samples. Compared to BSD animals, metabolomics changes involving energy and nucleotide metabolisms were observed in plasma samples of DCD animals before initiation of ESHP, whereas 2 metabolites (inosine monophosphate and methylbutyrate) exhibited concentration changes in biopsy samples. Normalization of DCD metabolic profile was remarkable after 4 hours of ESHP. CONCLUSION A specific metabolic profile was observed in DCD hearts, mainly characterized by an increased nucleotide catabolism. DCD and BSD metabolomes proved normalized during ESHP. Complementary investigations are needed to correlate these findings to cardiac performances.
Collapse
Affiliation(s)
- Thaïs Hautbergue
- Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Paris-Saclay University, CEA, INRAE, Gif-sur-Yvette, France
| | - Florent Laverdure
- Department of Anesthesiology and Intensive Care, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Paris-Saclay University, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France; Preclinical Research Laboratory, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France
| | - Simon Dang Van
- Preclinical Research Laboratory, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France
| | - Aurelien Vallee
- Preclinical Research Laboratory, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France; Department of Cardiac Surgery, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France
| | - Mateo Sanchis-Borja
- Preclinical Research Laboratory, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France
| | - Benoît Decante
- Preclinical Research Laboratory, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France
| | - Maïra Gaillard
- Preclinical Research Laboratory, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France; Department of Cardiac Surgery, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France
| | - Christophe Junot
- Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Paris-Saclay University, CEA, INRAE, Gif-sur-Yvette, France
| | - François Fenaille
- Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Paris-Saclay University, CEA, INRAE, Gif-sur-Yvette, France
| | - Olaf Mercier
- Preclinical Research Laboratory, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France; Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France; INSERM UMR_S 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies, Paris-Saclay University, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; Paris-Saclay University School of Medicine, Le Kremlin-Bicêtre, France
| | - Benoit Colsch
- Département Médicaments et Technologies pour la Santé (DMTS), MetaboHUB, Paris-Saclay University, CEA, INRAE, Gif-sur-Yvette, France
| | - Julien Guihaire
- Preclinical Research Laboratory, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France; Department of Cardiac Surgery, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France; Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Paris-Saclay University, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, Pulmonary Hypertension National Referral Center, Le Plessis Robinson, France.
| |
Collapse
|
34
|
Flam E, Arany Z. Metabolite signaling in the heart. NATURE CARDIOVASCULAR RESEARCH 2023; 2:504-516. [PMID: 39195876 DOI: 10.1038/s44161-023-00270-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/29/2023] [Indexed: 08/29/2024]
Abstract
The heart is the most metabolically active organ in the body, sustaining a continuous and high flux of nutrient catabolism via oxidative phosphorylation. The nature and relative contribution of these fuels have been studied extensively for decades. By contrast, less attention has been placed on how intermediate metabolites generated from this catabolism affect intracellular signaling. Numerous metabolites, including intermediates of glycolysis and the tricarboxylic acid (TCA) cycle, nucleotides, amino acids, fatty acids and ketones, are increasingly appreciated to affect signaling in the heart, via various mechanisms ranging from protein-metabolite interactions to modifying epigenetic marks. We review here the current state of knowledge of intermediate metabolite signaling in the heart.
Collapse
Affiliation(s)
- Emily Flam
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zolt Arany
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Bundgaard A, Gruszczyk AV, Prag HA, Williams C, McIntyre A, Ruhr IM, James AM, Galli GLJ, Murphy MP, Fago A. Low production of mitochondrial reactive oxygen species after anoxia and reoxygenation in turtle hearts. J Exp Biol 2023; 226:jeb245516. [PMID: 37066839 PMCID: PMC10184768 DOI: 10.1242/jeb.245516] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/03/2023] [Indexed: 04/18/2023]
Abstract
Extremely anoxia-tolerant animals, such as freshwater turtles, survive anoxia and reoxygenation without sustaining tissue damage to their hearts. In contrast, for mammals, the ischemia-reperfusion (IR) injury that leads to tissue damage during a heart attack is initiated by a burst of superoxide (O2·-) production from the mitochondrial respiratory chain upon reperfusion of ischemic tissue. Whether turtles avoid oxidative tissue damage because of an absence of mitochondrial superoxide production upon reoxygenation, or because the turtle heart is particularly protected against this damage, is unclear. Here, we investigated whether there was an increase in mitochondrial O2·- production upon the reoxygenation of anoxic red-eared slider turtle hearts in vivo and in vitro. This was done by measuring the production of H2O2, the dismutation product of O2·-, using the mitochondria-targeted mass-spectrometric probe in vivo MitoB, while in parallel assessing changes in the metabolites driving mitochondrial O2·- production, succinate, ATP and ADP levels during anoxia, and H2O2 consumption and production rates of isolated heart mitochondria. We found that there was no excess production of in vivo H2O2 during 1 h of reoxygenation in turtles after 3 h anoxia at room temperature, suggesting that turtle hearts most likely do not suffer oxidative injury after anoxia because their mitochondria produce no excess O2·- upon reoxygenation. Instead, our data support the conclusion that both the low levels of succinate accumulation and the maintenance of ADP levels in the anoxic turtle heart are key factors in preventing the surge of O2·- production upon reoxygenation.
Collapse
Affiliation(s)
- Amanda Bundgaard
- CECAD, University of Cologne, 50931 Cologne, Germany
- Department of Biology, Aarhus University, DK-8000 Aarhus, Denmark
| | - Anja V. Gruszczyk
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Hiran A. Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | | | - Angela McIntyre
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Ilan M. Ruhr
- Faculty of Medical and Human Sciences, University of Manchester, Manchester M13 9PL, UK
| | - Andrew M. James
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Gina L. J. Galli
- Faculty of Medical and Human Sciences, University of Manchester, Manchester M13 9PL, UK
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Angela Fago
- Department of Biology, Aarhus University, DK-8000 Aarhus, Denmark
| |
Collapse
|
36
|
Castejon-Vega B, Cordero MD, Sanz A. How the Disruption of Mitochondrial Redox Signalling Contributes to Ageing. Antioxidants (Basel) 2023; 12:831. [PMID: 37107206 PMCID: PMC10135186 DOI: 10.3390/antiox12040831] [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: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
In the past, mitochondrial reactive oxygen species (mtROS) were considered a byproduct of cellular metabolism. Due to the capacity of mtROS to cause oxidative damage, they were proposed as the main drivers of ageing and age-related diseases. Today, we know that mtROS are cellular messengers instrumental in maintaining cellular homeostasis. As cellular messengers, they are produced in specific places at specific times, and the intensity and duration of the ROS signal determine the downstream effects of mitochondrial redox signalling. We do not know yet all the processes for which mtROS are important, but we have learnt that they are essential in decisions that affect cellular differentiation, proliferation and survival. On top of causing damage due to their capacity to oxidize cellular components, mtROS contribute to the onset of degenerative diseases when redox signalling becomes dysregulated. Here, we review the best-characterized signalling pathways in which mtROS participate and those pathological processes in which they are involved. We focus on how mtROS signalling is altered during ageing and discuss whether the accumulation of damaged mitochondria without signalling capacity is a cause or a consequence of ageing.
Collapse
Affiliation(s)
- Beatriz Castejon-Vega
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Mario D. Cordero
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain
| | - Alberto Sanz
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
37
|
Parente A, Cho HD, Kim KH, Schlegel A. Association between Hepatocellular Carcinoma Recurrence and Graft Size in Living Donor Liver Transplantation: A Systematic Review. Int J Mol Sci 2023; 24:6224. [PMID: 37047199 PMCID: PMC10093934 DOI: 10.3390/ijms24076224] [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: 02/21/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
The aim of this work was to assess the association between graft-to-recipient weight ratio (GRWR) in adult-to-adult living donor liver transplantation (LDLT) and hepatocellular carcinoma (HCC) recurrence. A search of the MEDLINE and EMBASE databases was performed until December 2022 for studies comparing different GRWRs in the prognosis of HCC recipients in LDLT. Data were pooled to evaluate 1- and 3-year survival rates. We identified three studies, including a total of 782 patients (168 GRWR < 0.8 vs. 614 GRWR ≥ 0.8%). The pooled overall survival was 85% and 77% at one year and 90% and 83% at three years for GRWR < 0.8 and GRWR ≥ 0.8, respectively. The largest series found that, in patients within Milan criteria, the GRWR was not associated with lower oncological outcomes. However, patients with HCC outside the Milan criteria with a GRWR < 0.8% had lower survival and higher tumor recurrence rates. The GRWR < 0.8% appears to be associated with lower survival rates in HCC recipients, particularly for candidates with tumors outside established HCC criteria. Although the data are scarce, the results of this study suggest that considering the individual GRWR not only as risk factor for small-for-size-syndrome but also as contributor to HCC recurrence in patients undergoing LDLT would be beneficial. Novel perfusion technologies and pharmacological interventions may contribute to improving outcomes.
Collapse
Affiliation(s)
- Alessandro Parente
- HPB and Transplant Unit, Department of Surgical Science, University of Rome Tor Vergata, 00133 Rome, Italy
- Division of Hepatobiliary and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Hwui-Dong Cho
- Division of Hepatobiliary and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Ki-Hun Kim
- Division of Hepatobiliary and Liver Transplantation, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Andrea Schlegel
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Centre of Preclinical Research, 20122 Milan, Italy
- Department of Surgery and Transplantation, Swiss HPB Centre, University Hospital Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
38
|
Lamanilao GG, Dogan M, Patel PS, Azim S, Patel DS, Bhattacharya SK, Eason JD, Kuscu C, Kuscu C, Bajwa A. Key hepatoprotective roles of mitochondria in liver regeneration. Am J Physiol Gastrointest Liver Physiol 2023; 324:G207-G218. [PMID: 36648139 PMCID: PMC9988520 DOI: 10.1152/ajpgi.00220.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/28/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
Treatment of advanced liver disease using surgical modalities is possible due to the liver's innate ability to regenerate following resection. Several key cellular events in the regenerative process converge at the mitochondria, implicating their crucial roles in liver regeneration. Mitochondria enable the regenerating liver to meet massive metabolic demands by coordinating energy production to drive cellular proliferative processes and vital homeostatic functions. Mitochondria are also involved in terminating the regenerative process by mediating apoptosis. Studies have shown that attenuation of mitochondrial activity results in delayed liver regeneration, and liver failure following resection is associated with mitochondrial dysfunction. Emerging mitochondria therapy (i.e., mitotherapy) strategies involve isolating healthy donor mitochondria for transplantation into diseased organs to promote regeneration. This review highlights mitochondria's inherent role in liver regeneration.
Collapse
Affiliation(s)
- Gene G Lamanilao
- Department of Surgery, Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Murat Dogan
- Department of Surgery, Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Prisha S Patel
- Department of Surgery, Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Shafquat Azim
- Department of Surgery, Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Disha S Patel
- Department of Legal Studies, Belmont University, Nashville, Tennessee, United States
| | - Syamal K Bhattacharya
- Division of Cardiovascular Diseases, Department of Medicine, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - James D Eason
- Department of Surgery, Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Canan Kuscu
- Department of Surgery, Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Cem Kuscu
- Department of Surgery, Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| | - Amandeep Bajwa
- Department of Surgery, Transplant Research Institute, James D. Eason Transplant Institute, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
- Department of Genetics, Genomics, and Informatics, The University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee, United States
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, United States
| |
Collapse
|
39
|
Lei I, Huang W, Noly PE, Naik S, Ghali M, Liu L, Pagani FD, Abou El Ela A, Pober JS, Pitt B, Platt JL, Cascalho M, Wang Z, Chen YE, Mortensen RM, Tang PC. Metabolic reprogramming by immune-responsive gene 1 up-regulation improves donor heart preservation and function. Sci Transl Med 2023; 15:eade3782. [PMID: 36753565 PMCID: PMC10068866 DOI: 10.1126/scitranslmed.ade3782] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Preservation quality of donor hearts is a key determinant of transplant success. Preservation duration beyond 4 hours is associated with primary graft dysfunction (PGD). Given transport time constraints, geographical limitations exist for donor-recipient matching, leading to donor heart underutilization. Here, we showed that metabolic reprogramming through up-regulation of the enzyme immune response gene 1 (IRG1) and its product itaconate improved heart function after prolonged preservation. Irg1 transcript induction was achieved by adding the histone deacetylase (HDAC) inhibitor valproic acid (VPA) to a histidine-tryptophan-ketoglutarate solution used for donor heart preservation. VPA increased acetylated H3K27 occupancy at the IRG1 enhancer and IRG1 transcript expression in human donor hearts. IRG1 converts aconitate to itaconate, which has both anti-inflammatory and antioxidant properties. Accordingly, our studies showed that Irg1 transcript up-regulation by VPA treatment increased nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) in mice, which was accompanied by increased antioxidant protein expression [hemeoxygenase 1 (HO1) and superoxide dismutase 1 (SOD1)]. Deletion of Irg1 in mice (Irg1-/-) negated the antioxidant and cardioprotective effects of VPA. Consistent with itaconate's ability to inhibit succinate dehydrogenase, VPA treatment of human hearts increased itaconate availability and reduced succinate accumulation during preservation. VPA similarly increased IRG1 expression in pig donor hearts and improved its function in an ex vivo cardiac perfusion system both at the clinical 4-hour preservation threshold and at 10 hours. These results suggest that augmentation of cardioprotective immune-metabolomic pathways may be a promising therapeutic strategy for improving donor heart function in transplantation.
Collapse
Affiliation(s)
- Ienglam Lei
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Wei Huang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pierre Emmanuel Noly
- Department of Cardiac Surgery, Université de Montréal, Montréal, Quebec H1T 1C8, Canada
| | - Suyash Naik
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Miriyam Ghali
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Liu Liu
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francis D Pagani
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ashraf Abou El Ela
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jordan S Pober
- Department of Pathology, Yale University, New Haven, CT 06510, USA
| | - Bertram Pitt
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeffrey L Platt
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marilia Cascalho
- Department of Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhong Wang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard M Mortensen
- Departments of Molecular and Integrative Physiology, Internal Medicine, and Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Paul C Tang
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
40
|
Albert C, Harris M, DiRito J, Shi A, Edwards C, Harkins L, Lysyy T, Kulkarni S, Mulligan DC, Hosgood SA, Watson CJE, Friend PJ, Nicholson ML, Haakinson D, Saeb-Parsy K, Tietjen GT. Honoring the gift: The transformative potential of transplant-declined human organs. Am J Transplant 2023; 23:165-170. [PMID: 36695696 DOI: 10.1016/j.ajt.2022.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/24/2022] [Accepted: 11/13/2022] [Indexed: 01/09/2023]
Abstract
For decades, transplantation has been a life-saving treatment for those fortunate enough to gain access. Nevertheless, many patients die waiting for an organ and countless more never make it onto the waitlist because of a shortage of donor organs. Concurrently, thousands of donated organs are declined for transplant each year because of concerns about poor outcomes post-transplant. The decline of any donated organ-even if medically justified-is tragic for both the donor family and potential recipients. In this Personal Viewpoint, we discuss the need for a new mindset in how we honor the gift of organ donation. We believe that the use of transplant-declined human organs in translational research has the potential to hasten breakthrough discoveries in a multitude of scientific and medical areas. More importantly, such breakthroughs will allow us to properly value every donated organ. We further discuss the many practical challenges that such research presents and offer some possible solutions based on experiences in our own research laboratories. Finally, we share our perspective on what we believe are the necessary next steps to ensure a future where every donated organ realizes its full potential to impact the lives of current and future patients.
Collapse
Affiliation(s)
- Claire Albert
- Yale University, Department of Biomedical Engineering, New Haven, Connecticut, USA
| | - Matthew Harris
- Yale School of Medicine, Department of Surgery, New Haven, Connecticut, USA
| | - Jenna DiRito
- Yale School of Medicine, Department of Surgery, New Haven, Connecticut, USA
| | - Audrey Shi
- Yale School of Medicine, Department of Surgery, New Haven, Connecticut, USA
| | | | - Lauren Harkins
- Yale University, Department of Biomedical Engineering, New Haven, Connecticut, USA
| | - Taras Lysyy
- Yale School of Medicine, Department of Surgery, New Haven, Connecticut, USA
| | - Sanjay Kulkarni
- Yale School of Medicine, Department of Surgery, New Haven, Connecticut, USA
| | - David C Mulligan
- Yale School of Medicine, Department of Surgery, New Haven, Connecticut, USA
| | - Sarah A Hosgood
- Department of Surgery, University of Cambridge, and Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - Christopher J E Watson
- Department of Surgery, University of Cambridge, and Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - Peter J Friend
- University of Oxford, Nuffield Department of Surgical Sciences and the Oxford Transplant Centre, Oxford, UK
| | - Michael L Nicholson
- Department of Surgery, University of Cambridge, and Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - Danielle Haakinson
- Yale School of Medicine, Department of Surgery, New Haven, Connecticut, USA
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, and Cambridge NIHR Biomedical Research Centre, Cambridge, UK.
| | - Gregory T Tietjen
- Yale University, Department of Biomedical Engineering, New Haven, Connecticut, USA; Yale School of Medicine, Department of Surgery, New Haven, Connecticut, USA.
| |
Collapse
|
41
|
Mottahedin A, Prag HA, Dannhorn A, Mair R, Schmidt C, Yang M, Sorby-Adams A, Lee JJ, Burger N, Kulaveerasingam D, Huang MM, Pluchino S, Peruzzotti-Jametti L, Goodwin R, Frezza C, Murphy MP, Krieg T. Targeting succinate metabolism to decrease brain injury upon mechanical thrombectomy treatment of ischemic stroke. Redox Biol 2023; 59:102600. [PMID: 36630820 PMCID: PMC9841348 DOI: 10.1016/j.redox.2023.102600] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/15/2022] [Accepted: 01/02/2023] [Indexed: 01/04/2023] Open
Abstract
Current treatments for acute ischemic stroke aim to reinstate a normal perfusion in the ischemic territory but can also cause significant ischemia-reperfusion (IR) injury. Previous data in experimental models of stroke show that ischemia leads to the accumulation of succinate, and, upon reperfusion, the accumulated succinate is rapidly oxidized by succinate dehydrogenase (SDH) to drive superoxide production at mitochondrial complex I. Despite this process initiating IR injury and causing further tissue damage, the potential of targeting succinate metabolism to minimize IR injury remains unexplored. Using both quantitative and untargeted high-resolution metabolomics, we show a time-dependent accumulation of succinate in both human and mouse brain exposed to ischemia ex vivo. In a mouse model of ischemic stroke/mechanical thrombectomy mass spectrometry imaging (MSI) shows that succinate accumulation is confined to the ischemic region, and that the accumulated succinate is rapidly oxidized upon reperfusion. Targeting succinate oxidation by systemic infusion of the SDH inhibitor malonate upon reperfusion leads to a dose-dependent decrease in acute brain injury. Together these findings support targeting succinate metabolism upon reperfusion to decrease IR injury as a valuable adjunct to mechanical thrombectomy in ischemic stroke.
Collapse
Affiliation(s)
- Amin Mottahedin
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK; Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.
| | - Hiran A Prag
- Department of Medicine, University of Cambridge, Cambridge University Hospitals, Cambridge, UK
| | - Andreas Dannhorn
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R & D, AstraZeneca, Cambridge, UK
| | - Richard Mair
- Division of Neurosurgery, Department of Clinical Neurosciences, Cambridge University Hospitals, Cambridge, UK
| | - Christina Schmidt
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Ming Yang
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Annabel Sorby-Adams
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Jordan J Lee
- Department of Medicine, University of Cambridge, Cambridge University Hospitals, Cambridge, UK
| | - Nils Burger
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Margaret M Huang
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Stefano Pluchino
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences and NIHR Biomedical Research Centre, University of Cambridge, UK
| | - Richard Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R & D, AstraZeneca, Cambridge, UK
| | - Christian Frezza
- CECAD Research Center, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge University Hospitals, Cambridge, UK.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge University Hospitals, Cambridge, UK.
| |
Collapse
|
42
|
Mitochondrial Dysfunction: The Hidden Player in the Pathogenesis of Atherosclerosis? Int J Mol Sci 2023; 24:ijms24021086. [PMID: 36674602 PMCID: PMC9861427 DOI: 10.3390/ijms24021086] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/29/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023] Open
Abstract
Atherosclerosis is a multifactorial inflammatory pathology that involves metabolic processes. Improvements in therapy have drastically reduced the prognosis of cardiovascular disease. Nevertheless, a significant residual risk is still relevant, and is related to unmet therapeutic targets. Endothelial dysfunction and lipid infiltration are the primary causes of atherosclerotic plaque progression. In this contest, mitochondrial dysfunction can affect arterial wall cells, in particular macrophages, smooth muscle cells, lymphocytes, and endothelial cells, causing an increase in reactive oxygen species (ROS), leading to oxidative stress, chronic inflammation, and intracellular lipid deposition. The detection and characterization of mitochondrial DNA (mtDNA) is crucial for assessing mitochondrial defects and should be considered the goal for new future therapeutic interventions. In this review, we will focus on a new idea, based on the analysis of data from many research groups, namely the link between mitochondrial impairment and endothelial dysfunction and, in particular, its effect on atherosclerosis and aging. Therefore, we discuss known and novel mitochondria-targeting therapies in the contest of atherosclerosis.
Collapse
|
43
|
Allen FM, Costa ASH, Gruszczyk AV, Bates GR, Prag HA, Nikitopoulou E, Viscomi C, Frezza C, James AM, Murphy MP. Rapid fractionation of mitochondria from mouse liver and heart reveals in vivo metabolite compartmentation. FEBS Lett 2023; 597:246-261. [PMID: 36217875 PMCID: PMC7614208 DOI: 10.1002/1873-3468.14511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 09/30/2022] [Accepted: 10/07/2022] [Indexed: 02/02/2023]
Abstract
The compartmentation and distribution of metabolites between mitochondria and the rest of the cell is a key parameter of cell signalling and pathology. Here, we have developed a rapid fractionation procedure that enables us to take mouse heart and liver from in vivo and within ~ 30 s stabilise the distribution of metabolites between mitochondria and the cytosol by rapid cooling, homogenisation and dilution. This is followed by centrifugation of mitochondria through an oil layer to separate mitochondrial and cytosolic fractions for subsequent metabolic analysis. Using this procedure revealed the in vivo compartmentation of mitochondrial metabolites and will enable the assessment of the distribution of metabolites between the cytosol and mitochondria during a range of situations in vivo.
Collapse
Affiliation(s)
- Fay M. Allen
- MRC Mitochondrial Biology UnitUniversity of CambridgeUK
| | | | | | | | - Hiran A. Prag
- MRC Mitochondrial Biology UnitUniversity of CambridgeUK
| | | | - Carlo Viscomi
- Department of Biomedical SciencesUniversity of PadovaItaly
| | | | | | - Michael P. Murphy
- MRC Mitochondrial Biology UnitUniversity of CambridgeUK
- Department of MedicineUniversity of CambridgeUK
| |
Collapse
|
44
|
Saemann L, Kohl M, Veres G, Korkmaz‐Icöz S, Großkopf A, Karck M, Simm A, Wenzel F, Szabó G. Prediction Model for Contractile Function of Circulatory Death Donor Hearts Based on Microvascular Flow Shifts During Ex Situ Hypothermic Cardioplegic Machine Perfusion. J Am Heart Assoc 2022; 11:e027146. [PMID: 36382941 PMCID: PMC9851462 DOI: 10.1161/jaha.122.027146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Hearts procured from circulatory death donors (DCD) are predominantly maintained by machine perfusion (MP) with normothermic donor blood. Currently, DCD heart function is evaluated by lactate and visual inspection. We have shown that MP with the cardioplegic, crystalloid Custodiol-N solution is superior to blood perfusion to maintain porcine DCD hearts. However, no method has been developed yet to predict the contractility of DCD hearts after cardioplegic MP. We hypothesize that the shift of microvascular flow during continuous MP with a cardioplegic preservation solution predicts the contractility of DCD hearts. Methods and Results In a pig model, DCD hearts were harvested and maintained by MP with hypothermic, oxygenated Custodiol-N for 4 hours while myocardial microvascular flow was measured by Laser Doppler Flow (LDF) technology. Subsequently, hearts were perfused with blood for 2 hours, and left ventricular contractility was measured after 30 and 120 minutes. Various novel parameters which represent the LDF shift were computed. We used 2 combined LDF shift parameters to identify bivariate prediction models. Using the new prediction models based on LDF shifts, highest r2 for end-systolic pressure was 0.77 (P=0.027), for maximal slope of pressure increment was 0.73 (P=0.037), and for maximal slope of pressure decrement was 0.75 (P=0.032) after 30 minutes of reperfusion. After 120 minutes of reperfusion, highest r2 for end-systolic pressure was 0.81 (P=0.016), for maximal slope of pressure increment was 0.90 (P=0.004), and for maximal slope of pressure decrement was 0.58 (P=0.115). Identical prediction models were identified for maximal slope of pressure increment and for maximal slope of pressure decrement at both time points. Lactate remained constant and therefore was unsuitable for prediction. Conclusions Contractility of DCD hearts after continuous MP with a cardioplegic preservation solution can be predicted by the shift of LDF during MP.
Collapse
Affiliation(s)
- Lars Saemann
- Department of Cardiac Surgery, University Hospital Halle (Saale)University of HalleHalle (Saale)Germany,Department of Cardiac SurgeryUniversity Hospital HeidelbergHeidelbergGermany
| | - Matthias Kohl
- Faculty Medical and Life SciencesFurtwangen UniversityVillingen‐SchwenningenGermany
| | - Gábor Veres
- Department of Cardiac Surgery, University Hospital Halle (Saale)University of HalleHalle (Saale)Germany,Department of Cardiac SurgeryUniversity Hospital HeidelbergHeidelbergGermany
| | - Sevil Korkmaz‐Icöz
- Department of Cardiac Surgery, University Hospital Halle (Saale)University of HalleHalle (Saale)Germany,Department of Cardiac SurgeryUniversity Hospital HeidelbergHeidelbergGermany
| | - Anne Großkopf
- Department of Cardiac Surgery, University Hospital Halle (Saale)University of HalleHalle (Saale)Germany
| | - Matthias Karck
- Department of Cardiac SurgeryUniversity Hospital HeidelbergHeidelbergGermany
| | - Andreas Simm
- Department of Cardiac Surgery, University Hospital Halle (Saale)University of HalleHalle (Saale)Germany
| | - Folker Wenzel
- Faculty Medical and Life SciencesFurtwangen UniversityVillingen‐SchwenningenGermany
| | - Gábor Szabó
- Department of Cardiac Surgery, University Hospital Halle (Saale)University of HalleHalle (Saale)Germany,Department of Cardiac SurgeryUniversity Hospital HeidelbergHeidelbergGermany
| |
Collapse
|
45
|
Cyclophilin D knockout mice do not accumulate succinate during cardiac ischemia. J Mol Cell Cardiol 2022; 173:73-74. [PMID: 36209553 DOI: 10.1016/j.yjmcc.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/07/2022]
|
46
|
Atallah R, Olschewski A, Heinemann A. Succinate at the Crossroad of Metabolism and Angiogenesis: Roles of SDH, HIF1α and SUCNR1. Biomedicines 2022; 10:3089. [PMID: 36551845 PMCID: PMC9775124 DOI: 10.3390/biomedicines10123089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Angiogenesis is an essential process by which new blood vessels develop from existing ones. While adequate angiogenesis is a physiological process during, for example, tissue repair, insufficient and excessive angiogenesis stands on the pathological side. Fine balance between pro- and anti-angiogenic factors in the tissue environment regulates angiogenesis. Identification of these factors and how they function is a pressing topic to develop angiogenesis-targeted therapeutics. During the last decade, exciting data highlighted non-metabolic functions of intermediates of the mitochondrial Krebs cycle including succinate. Among these functions is the contribution of succinate to angiogenesis in various contexts and through different mechanisms. As the concept of targeting metabolism to treat a wide range of diseases is rising, in this review we summarize the mechanisms by which succinate regulates angiogenesis in normal and pathological settings. Gaining a comprehensive insight into how this metabolite functions as an angiogenic signal will provide a useful approach to understand diseases with aberrant or excessive angiogenic background, and may provide strategies to tackle them.
Collapse
Affiliation(s)
- Reham Atallah
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Akos Heinemann
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
47
|
Mantelou AG, Barbouti A, Goussia A, Zacharioudaki A, Papoudou-Bai A, Vlachou C, Kokkoris S, Papalois A, Galaris D, Glantzounis GK. Combined administration of membrane-permeable and impermeable iron-chelating drugs attenuates ischemia/reperfusion-induced hepatic injury. Free Radic Biol Med 2022; 193:227-237. [PMID: 36243210 DOI: 10.1016/j.freeradbiomed.2022.10.266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND The underlying pathophysiological mechanisms of hepatic ischemia-reperfusion (I/R) injury have not been completely elucidated. However, it is well known that oxidative stress, caused by a burst of reactive oxygen species (ROS) production during the reperfusion phase, plays a crucial role. A growing body of evidence indicates that the intracellular availability of free iron represents a requirement for ROS-induced adverse effects, as iron catalyzes the generation of highly reactive free radicals. The aim of this study was to examine whether a combination of iron chelators with varying lipophilicity could offer enhanced protection against I/R by diminishing the conversion of weak oxidants, like H2O2, to extremely reactive ones such as hydroxyl radicals (HO.). METHODS HepG2 cells (hepatocellular carcinoma cell line) were exposed to oxidative stress conditions after pre-treatment with the iron chelators desferrioxamine (DFO) and deferiprone (DFP) alone or in combination. Labile iron pool was estimated using the calcein-acetoxymethyl ester (calcein-AM) method and DNA damage with the comet assay. We subsequently used a rabbit model (male New Zealand white rabbits) of hepatic I/R-induced injury to investigate, by measuring biochemical (ALT, ALT, ALP, γGT) and histological parameters, whether this may be true for in vivo conditions. RESULTS The combination of a membrane-permeable iron chelator (DFP) with a strong membrane-impermeable one (DFO) raises the level of protection in both hepatic cell lines exposed to oxidative stress conditions and hepatic I/R rabbit model. CONCLUSIONS Our results show that combinations of iron chelators with selected lipophilicity and iron-binding properties may represent a valuable strategy to protect against tissue damage during reperfusion after a period of ischemia.
Collapse
Affiliation(s)
- Athina G Mantelou
- HPB Unit, Department of Surgery, University Hospital of Ioannina and Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Anna Goussia
- Department of Pathology, University Hospital of Ioannina and Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | | | - Alexandra Papoudou-Bai
- Department of Pathology, University Hospital of Ioannina and Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Chara Vlachou
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Stelios Kokkoris
- First Department of Critical Care, School of Medicine, National and Kapodistrian University of Athens, Athens, 10676, Greece
| | - Apostolos Papalois
- Experimental, Educational and Research Center ELPEN, Athens, 19009, Greece; European University of Cyprus, School of Medicine, Nicosia, 2404, Cyprus
| | - Dimitrios Galaris
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece
| | - Georgios K Glantzounis
- HPB Unit, Department of Surgery, University Hospital of Ioannina and Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, 45110, Greece.
| |
Collapse
|
48
|
Abstract
Metabolic changes frequently occur in patients with chronic heart failure (HF). Therefore, detailed identification of these metabolic changes, and complementing them, will provide new therapeutic approaches. Here, using a mouse model, we demonstrated that succinyl-CoA levels are reduced in the myocardial mitochondria of hearts undergoing chronic HF, and this reduction impairs mitochondrial oxidative phosphorylation capacity. We identified increased heme synthesis as a cause of this succinyl-CoA reduction and demonstrated a method that can compensate substantially for the increased succinyl-CoA consumption. Reduction in succinyl-CoA levels has also been reported in HF patients. Our results provide an academic basis for the development of new treatment methodologies against HF, which target the altered metabolic activities that occur in HF by nutritional interventions. Heart failure (HF) is a leading cause of death and repeated hospitalizations and often involves cardiac mitochondrial dysfunction. However, the underlying mechanisms largely remain elusive. Here, using a mouse model in which myocardial infarction (MI) was induced by coronary artery ligation, we show the metabolic basis of mitochondrial dysfunction in chronic HF. Four weeks after ligation, MI mice showed a significant decrease in myocardial succinyl-CoA levels, and this decrease impaired the mitochondrial oxidative phosphorylation (OXPHOS) capacity. Heme synthesis and ketolysis, and protein levels of several enzymes consuming succinyl-CoA in these events, were increased in MI mice, while enzymes synthesizing succinyl-CoA from α-ketoglutarate and glutamate were also increased. Furthermore, the ADP-specific subunit of succinyl-CoA synthase was reduced, while its GDP-specific subunit was almost unchanged. Administration of 5-aminolevulinic acid, an intermediate in the pathway from succinyl-CoA to heme synthesis, appreciably restored succinyl-CoA levels and OXPHOS capacity and prevented HF progression in MI mice. Previous reports also suggested the presence of succinyl-CoA metabolism abnormalities in cardiac muscles of HF patients. Our results identified that changes in succinyl-CoA usage in different metabolisms of the mitochondrial energy production system is characteristic to chronic HF, and although similar alterations are known to occur in healthy conditions, such as during strenuous exercise, they may often occur irreversibly in chronic HF leading to a decrease in succinyl-CoA. Consequently, nutritional interventions compensating the succinyl-CoA consumption are expected to be promising strategies to treat HF.
Collapse
|
49
|
Panconesi R, Widmer J, Carvalho MF, Eden J, Dondossola D, Dutkowski P, Schlegel A. Mitochondria and ischemia reperfusion injury. Curr Opin Organ Transplant 2022; 27:434-445. [PMID: 35950880 DOI: 10.1097/mot.0000000000001015] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW This review describes the role of mitochondria in ischemia-reperfusion-injury (IRI). RECENT FINDINGS Mitochondria are the power-house of our cells and play a key role for the success of organ transplantation. With their respiratory chain, mitochondria are the main energy producers, to fuel metabolic processes, control cellular signalling and provide electrochemical integrity. The mitochondrial metabolism is however severely disturbed when ischemia occurs. Cellular energy depletes rapidly and various metabolites, including Succinate accumulate. At reperfusion, reactive oxygen species are immediately released from complex-I and initiate the IRI-cascade of inflammation. Prior to the development of novel therapies, the underlying mechanisms should be explored to target the best possible mitochondrial compound. A clinically relevant treatment should recharge energy and reduce Succinate accumulation before organ implantation. While many interventions focus instead on a specific molecule, which may inhibit downstream IRI-inflammation, mitochondrial protection can be directly achieved through hypothermic oxygenated perfusion (HOPE) before transplantation. SUMMARY Mitochondria are attractive targets for novel molecules to limit IRI-associated inflammation. Although dynamic preservation techniques could serve as delivery tool for new therapeutic interventions, their own inherent mechanism should not only be studied, but considered as key treatment to reduce mitochondrial injury, as seen with the HOPE-approach.
Collapse
Affiliation(s)
- Rebecca Panconesi
- General Surgery 2U-Liver Transplant Unit, Department of Surgery, A.O.U. Città della Salute e della Scienza di Torino, University of Turin, Turin
- Hepatobiliary Unit, Careggi University Hospital, University of Florence, Florence, Italy
| | - Jeannette Widmer
- Swiss HPB and Transplant Center, Department of Visceral Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
| | | | - Janina Eden
- Swiss HPB and Transplant Center, Department of Visceral Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
| | - Daniele Dondossola
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Center for Preclinical Research, Milan, Italy
| | - Philipp Dutkowski
- Swiss HPB and Transplant Center, Department of Visceral Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
| | - Andrea Schlegel
- Hepatobiliary Unit, Careggi University Hospital, University of Florence, Florence, Italy
- Swiss HPB and Transplant Center, Department of Visceral Surgery and Transplantation, University Hospital Zurich, Zurich, Switzerland
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Center for Preclinical Research, Milan, Italy
| |
Collapse
|
50
|
Muller X, Rossignol G, Mohkam K, Mabrut JY. Novel strategies in liver graft preservation - The French perspective. J Visc Surg 2022; 159:389-398. [PMID: 36109331 DOI: 10.1016/j.jviscsurg.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Given the increasing graft shortage, the transplant community is forced to use so called marginal liver grafts with a higher susceptibility to ischemia-reperfusion injury. This exposes the recipient to a higher risk of graft failure and post-transplant complications. While static cold storage remains the gold standard in low-risk transplant scenarios, dynamic preservation strategies may allow to improve outcomes after transplantation of marginal liver grafts. Two dynamic preservation strategies, end-ischemic hypothermic oxygenated perfusion (HOPE) and continuous normothermic machine perfusion (cNMP), have been evaluated in randomized clinical trials. The results show improved preservation of liver grafts after cNMP and reduction of post-transplant biliary complications after HOPE. In comparison to cNMP, HOPE has the advantage of requiring less logistics and expertise with the possibility to return to default static cold storage. Both strategies allow to assess graft viability prior to transplantation and may thus contribute to optimizing graft selection and reducing discard rates. The use of dynamic preservation is rapidly increasing in France and results from a national randomized trial on the use of HOPE in marginal grafts will soon be available. Future applications should focus on controlled donation after circulatory death liver grafts, split grafts and graft treatment during perfusion. The final aim of dynamic liver graft preservation is to improve post-transplant outcomes, increase the number of transplanted grafts and allow expansion of transplant indications.
Collapse
Affiliation(s)
- X Muller
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, Lyon, France; The Lyon Cancer Research Centre, Inserm U1052 UMR 5286, Lyon, France; ED 340 BMIC, Claude-Bernard Lyon 1 University, 69622 Villeurbanne, France.
| | - G Rossignol
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, Lyon, France; The Lyon Cancer Research Centre, Inserm U1052 UMR 5286, Lyon, France; ED 340 BMIC, Claude-Bernard Lyon 1 University, 69622 Villeurbanne, France; Department of Pediatric Surgery and Liver Transplantation, Femme-Mère-Enfant University Hospital, Hospices Civils de Lyon, Lyon, France
| | - K Mohkam
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, Lyon, France; The Lyon Cancer Research Centre, Inserm U1052 UMR 5286, Lyon, France; Department of Pediatric Surgery and Liver Transplantation, Femme-Mère-Enfant University Hospital, Hospices Civils de Lyon, Lyon, France
| | - J Y Mabrut
- Department of General Surgery and Liver Transplantation, Croix-Rousse University Hospital, Hospices Civils de Lyon, Lyon, France; The Lyon Cancer Research Centre, Inserm U1052 UMR 5286, Lyon, France
| |
Collapse
|