1
|
Hu JJ, Deng F, Sun QS, Xiong QM, Min Y, Feng SY, Lin ZB, Chen PH, Hu Z, Wu L, Chen XF, Xie S, Liu WF, Li C, Liu KX. Time-restricted feeding protects against septic liver injury by reshaping gut microbiota and metabolite 3-hydroxybutyrate. Gut Microbes 2025; 17:2486515. [PMID: 40223164 DOI: 10.1080/19490976.2025.2486515] [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: 11/04/2024] [Revised: 02/13/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Liver injury is an independent risk factor for multiple organ dysfunction and high mortality in patients with sepsis. However, the pathological mechanisms and therapeutic strategies for sepsis-associated liver injury have not been fully elucidated. Time-restricted feeding (TRF) is a promising dietary regime, but its role in septic liver injury remains unknown. Using 16S rRNA gene sequencing, Q200 targeted metabolomics, transcriptomics, germ-free mice, Hmgcs2/Lpin1 gene knockout mice, and Aml12 cells experiments, we revealed that TRF can mitigate septic liver injury by modulating the gut microbiota, particularly by increasing Lactobacillus murinus (L. murinus) abundance, which was significantly reduced in septic mice. Further study revealed that live L. murinus could markedly elevate serum levels of metabolite 3-hydroxybutyrate (3-HB) and alleviate sepsis-related injury, while the knockout of the key enzyme for 3-HB synthesis (3-hydroxy-3-methylglutaryl-CoA synthase 2, Hmgcs2) in the liver negated this protective effect. Additionally, serum 3-HB levels were significantly positively correlated with L. murinus abundance and negatively correlated with liver injury indicators in septic patients, demonstrating a strong predictive value for septic liver injury (AUC = 0.8429). Mechanistically, 3-HB significantly inhibited hepatocyte ferroptosis by activating the PI3K/AKT/mTOR/LPIN1 pathway, reducing ACSL4, MDA, LPO, and Fe2+ levels. This study demonstrates that TRF reduces septic liver injury by modulating gut microbiota to increase L. murinus, which elevates 3-HB to activate PI3K/AKT/mTOR/LPIN1 and inhibit hepatocyte ferroptosis. Overall, this study elucidates the protective mechanism of TRF against septic liver injury and identifies 3-HB as a potential therapeutic target and predictive biomarker, thereby providing new insights into the clinical management and diagnosis of septic liver injury.
Collapse
Affiliation(s)
- Jing-Juan Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Fan Deng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Qi-Shun Sun
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Qing-Ming Xiong
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, China
| | - Yue Min
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Si-Yuan Feng
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Ze-Bin Lin
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Peng-Han Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Zhen Hu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Ling Wu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Xiao-Feng Chen
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Sun Xie
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Wei-Feng Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Cai Li
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Precision Anesthesia and Perioperative Organ Protection, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Liu Z, Xu J, Que T, Que S, Valenti L, Zheng S. Molecular Mechanisms of Ischemia/Reperfusion Injury and Graft Dysfunction in Liver Transplantation: Insights from Multi-Omics Studies in Rodent Animal Models. Int J Biol Sci 2025; 21:2135-2154. [PMID: 40083684 PMCID: PMC11900806 DOI: 10.7150/ijbs.109449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 01/25/2025] [Indexed: 03/16/2025] Open
Abstract
Rodent ischemia-reperfusion injury (IRI) and liver transplantation (LT) models play crucial roles in mimicking graft injury and immune rejection, developing therapeutic approaches, and evaluating the efficacy of treatments. The application of integrated multi-omics data and advanced omics techniques like single-cell RNA sequencing in rodent models has expanded researchers' perspectives on pathophysiological processes in LT settings. This review summarizes key molecules and pathways associated with reperfusion injury and prognosis in LT models, highlighting the potential of omics data in understanding and improving transplant outcomes. In addition, we highlight the current challenges and future approaches for the application of omics data in rodent LT models. Cross-species validation with human data will improve therapeutic potential. Finally, further applications combining advanced single-cell, spatial omics technologies and machine learning algorithms will help to identify the key regulatory networks in specific cell populations underlying poor outcomes after LT.
Collapse
Affiliation(s)
- Zhengtao Liu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Shulan Hospital (Hangzhou), Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Xu
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Que
- Birth Defects Prevention and Control Institute, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | | | - Luca Valenti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Precision Medicine, Biological Resource Center Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Shusen Zheng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Shulan Hospital (Hangzhou), Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Organ Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
3
|
Xu J, Chen S, Liu D, Zhang Q, Luo T, Zhu J, Zhou L, Lin Y, Pan H, Chen Y, Zhao Q, Wang T, Andrea S, Nashan B, Stefan TG, Cai C, Cui J, He X, Guo Z. Suppression of Hepatocyte Ferroptosis via USP19-Mediated Deubiquitination of SLC7A11 in Ischemia-Free Liver Transplantation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406200. [PMID: 39574305 PMCID: PMC11809379 DOI: 10.1002/advs.202406200] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/17/2024] [Indexed: 02/11/2025]
Abstract
Ischemia-free liver transplantation (IFLT) is developed as a novel clinical approach to avoid ischemia-reperfusion injury (IRI). This study aims to identify the most distinguished programmed cell death pathway in grafts undergoing IFLT versus conventional liver transplantation (CLT) and to explore the underlying mechanism. Ferroptosis is the most distinct programmed cell death form between IFLT and CLT grafts. Among various cell death inhibitors, the ferroptosis inhibitor (Ferrostain-1) is the most effective one to prevent hepatocytes from damage induced by oxygen deprivation/reoxygenation (OGD/R). Hepatocyte ferroptosis is significantly alleviated in IFLT versus CLT grafts in both human beings and pigs. Ubiquitination enzyme screening identifies augmented amounts of ubiquitin-specific protease 19 (USP19) in IFLT versus CLT grafts. The upregulation of USP19 in the grafts is correlated with reduced pathological Suzuki's score, lower post-transplant peak liver enzyme level, and less early allograft dysfunction in liver transplant recipients. USP19 overexpression mitigates post-transplant liver injury in mice. Mechanistically, USP19 inhibits the degradation of solute carrier family 7 member 11 (SLC7A11) by removing its K63-linked ubiquitin chains. Notably, USP19 overexpression reduces ferroptosis and IRI in a SLC7A11-dependent manner in mice. Collectively, USP19-mediated suppression of hepatocyte ferroptosis via deubiquitinating SLC7A11 is a key mechanism by which IFLT abrogates graft IRI.
Collapse
Affiliation(s)
- Jinghong Xu
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Department of AnesthesiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Shirui Chen
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Di Liu
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Qi Zhang
- Department of Thyroid and Breast SurgeryThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230601China
| | - Tao Luo
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Jiaxing Zhu
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Liang Zhou
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Yuan Lin
- Department of PathologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Hongyu Pan
- Department of PathologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Yichao Chen
- Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Qiang Zhao
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Tielong Wang
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Schlegel Andrea
- Transplantation CenterDigestive Disease and Surgery Institute and Department of ImmunologyLerner Research Institute, Cleveland ClinicClevelandOhio44113USA
| | - Björn Nashan
- Organ Transplant CenterThe First Affiliated Hospital of the University of Science and Technology of ChinaHefeiAnhui230001China
| | - Tullius G. Stefan
- Division of Transplant SurgeryBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Changjie Cai
- Department of Critical CareThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Jun Cui
- School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdong510080China
| | - Xiaoshun He
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
| | - Zhiyong Guo
- Organ Transplant CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510080China
- Guangdong Provincial Key Laboratory of Organ MedicineGuangzhouGuangdong510080China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation)GuangzhouGuangdong510080China
- NHC Key Laboratory of Assisted CirculationSun Yat‐sen UniversityGuangzhouGuangdong510080China
| |
Collapse
|
4
|
Ishikawa K, Murao A, Aziz M, Wang P. Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 (MOP3) attenuates inflammation and improves survival in hepatic ischemia/reperfusion injury. Surgery 2025; 178:108872. [PMID: 39455391 PMCID: PMC11717596 DOI: 10.1016/j.surg.2024.09.029] [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: 06/21/2024] [Revised: 08/20/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024]
Abstract
INTRODUCTION Hepatic ischemia/reperfusion injury is a severe clinical condition leading to high mortality as the result of excessive inflammation, partially triggered by released damage-associated molecular patterns. Extracellular cold-inducible RNA-binding protein is a new damage-associated molecular pattern. Current clinical management of hepatic ischemia/reperfusion injury is limited to supportive therapy, necessitating the development of novel and effective treatment strategies. Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 is a newly invented oligopeptide originating from milk fat globule-epidermal growth factor-VIII. This peptide acts as an opsonic compound that specifically binds to extracellular cold-inducible RNA-binding protein to facilitate its clearance by phagocytes, thereby attenuating inflammation. In this study, we hypothesized that milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 attenuated hepatic ischemia/reperfusion injury by inhibiting extracellular cold-inducible RNA-binding protein-induced inflammation in Kupffer cells. METHODS We treated Kupffer cells isolated from male C57BL/6 mice with extracellular cold-inducible RNA-binding protein and various doses of milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 for 4 hours, then measured cytokines in the culture supernatants. In addition, mice underwent 70% hepatic ischemia for 60 minutes immediately followed by the intravenous administration of either vehicle or milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3. Blood and ischemic liver tissues were collected 24 hours later, and inflammatory markers including cytokines, liver enzymes, chemokines, myeloperoxidase activity, and Z-DNA-binding protein 1 were measured. Hepatic tissue damage and cell death were evaluated histologically. Survival rates were monitored for 10 days posthepatic ischemia/reperfusion. RESULTS The release of interleukin-6 and tumor necrosis factor-α from extracellular cold-inducible RNA-binding protein-challenged Kupffer cells was significantly reduced by milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 in a dose-dependent manner. In hepatic ischemia/reperfusion mice, milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 treatment significantly decreased serum levels of extracellular cold-inducible RNA-binding protein, interleukin-6, tumor necrosis factor-α, aspartate aminotransferase, alanine aminotransferase, and lactate dehydrogenase. Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 treatment also significantly reduced mRNA levels of interleukin-6, tumor necrosis factor-α, interleukin-1β, Z-DNA-binding protein 1, and chemokine macrophage inflammatory protein-2, as well as myeloperoxidase activity in hepatic tissues. Histologic evaluation demonstrated that treatment with milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 significantly attenuated tissue damage and cell death in the liver of hepatic ischemia/reperfusion mice. Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 treatment significantly improved the survival rate of hepatic ischemia/reperfusion mice. CONCLUSION Milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 significantly attenuated inflammation and liver tissue damage and improved survival after hepatic ischemia/reperfusion. Thus, milk fat globule-epidermal growth factor-VIII-derived oligopeptide 3 holds promise as a potential future therapeutic strategy for hepatic ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Kouhei Ishikawa
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York
| |
Collapse
|
5
|
Lyon A, Agius T, Macarthur MR, Kiesworo K, Stavart L, Allagnat F, Mitchell SJ, Riella LV, Uygun K, Yeh H, Déglise S, Golshayan D, Longchamp A. Dietary or pharmacological inhibition of insulin-like growth factor-1 protects from renal ischemia-reperfusion injury in mice. iScience 2024; 27:111256. [PMID: 39759002 PMCID: PMC11700642 DOI: 10.1016/j.isci.2024.111256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/29/2024] [Accepted: 10/23/2024] [Indexed: 01/07/2025] Open
Abstract
One-week protein restriction (PR) limits ischemia-reperfusion (IR) damages and improves metabolic fitness. Similarly, longer-term calory restriction results in increased lifespan, partly via reduced insulin-like growth factor (IGF)-1. However, the influence of short-term PR on IGF-1 and its impact on IR are unknown. PR was achieved in mice via one-week carbohydrate loading and/or through a low-protein diet. PR decreased IGF-1 circulating levels as well as renal and hepatic expression. Upon renal IR, serum IGF-1 positively correlated with renal dysfunction and tissular damages, independently of sex and age. Exogenous IGF-1 administration abrogated PR benefits during IR, while IGF-1 receptor inhibition with linsitinib was protective. IGF-1 was associated with a reduction in forkhead box O (FoxO), and AMP-activated protein kinase (AMPK) signaling pathways previously demonstrated to improve IR resilience in various organs. These data support dietary or pharmacological reduction of IGF-1 signaling to mitigate IR injury prior to solid organ transplantation and beyond.
Collapse
Affiliation(s)
- Arnaud Lyon
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Transplantation Center and Transplantation Immunopathology Laboratory, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas Agius
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael R. Macarthur
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Kevin Kiesworo
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Louis Stavart
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Transplantation Center and Transplantation Immunopathology Laboratory, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | | | - Leonardo V. Riella
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Korkut Uygun
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi Yeh
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sebastien Déglise
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Déla Golshayan
- Transplantation Center and Transplantation Immunopathology Laboratory, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
You Y, Chen S, Deng H, Xing X, Tang B, Wu Y, Lei E. Remifentanil represses oxidative stress to relieve hepatic ischemia/reperfusion injury via regulating BACH1/PRDX1 axis. Clin Res Hepatol Gastroenterol 2024; 48:102422. [PMID: 39025461 DOI: 10.1016/j.clinre.2024.102422] [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: 05/14/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Hepatic ischemia-reperfusion injury (HIRI) is a major cause of liver dysfunction after clinical liver surgery, which seriously affects the prognosis of patients. Remifentanil (RE) has been verified to attenuate HIRI. However, its therapeutic mechanism is still unclear. This study aimed to explore the protective mechanism of RE against HIRI. METHODS A mouse HIRI model and an in vitro model of hypoxia/reoxygenation (H/R)-stimulated AML12 hepatocytes were established. Liver histopathological changes were evaluated by hematoxylin and eosin (HE) staining. Oxidative stress damage was assessed by malondialdehyde (MDA), superoxide dismutase (SOD), and reactive oxygen species (ROS) levels. Liver function was determined by serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH). and adenosine triphosphate (ATP) levels. Cell counting kit-8 (CCK-8) assessed cell viability. Apoptosis was measured by terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL) and flow cytometry. The levels of inflammatory factors were detected by enzyme-linked immunosorbent assay (ELISA) kits. The differentially expressed genes were evaluated by mRNA microarray analysis. Western blotting and real-time quantitative polymerase chain reaction (RT-qPCR) were conducted to detect molecule expression. The binding of BTB and CNC homology 1 (BACH1) to peroxiredoxin 1 (PRDX1) was validated by chromatin immunoprecipitation (ChIP) and dual luciferase reporter assay. RESULTS RE treatment improved liver function, and repressed oxidative stress damage and apoptosis in HIRI mice. Nine differentially expressed genes in the liver tissues of HIRI mice were selected by microarray analysis, among which BACH1 was down-regulated and PRDX1 was up-regulated after RE treatment. In addition, BACH1 directly bound to the promoter region of PRDX1 to inhibit its transcription and expression, which led to oxidative stress injury. BACH1 overexpression or PRDX1 silencing could counteract the beneficial effects of RE against HIRI. CONCLUSION RE suppressed oxidative stress injury and inflammation via inactivation of the BACH1/PRDX1 axis, thereby ameliorating HIRI. Our findings enrich the understanding of the protective mechanisms of RE against HIRI, and provide novel evidence for its clinical application.
Collapse
Affiliation(s)
- Yujuan You
- Department of Anesthesiology, The 2(nd) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, PR China.
| | - Shoulin Chen
- Department of Anesthesiology, The 2(nd) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, PR China.
| | - Huanling Deng
- Department of Anesthesiology, The 2(nd) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, PR China.
| | - Xianliang Xing
- Department of Anesthesiology, The 2(nd) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, PR China.
| | - Binquan Tang
- Department of Anesthesiology, The 2(nd) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, PR China.
| | - Yiguo Wu
- Department of Blood Transfusion, The 2(nd) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330008, PR China.
| | - Enjun Lei
- Department of Anesthesiology, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, PR China.
| |
Collapse
|
7
|
Whalen C, Verma A, Kurashima K, Carter J, Nazzal H, Jain A. Novel Models for Assessing and Pathophysiology of Hepatic Ischemia-Reperfusion Injury Mechanisms. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1507. [PMID: 39336548 PMCID: PMC11434406 DOI: 10.3390/medicina60091507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Hepatic ischemia-reperfusion injury (IRI) is a major cause of postoperative hepatic dysfunction and liver failure involving cellular damage to previously ischemic tissues to which blood flow is restored. The reestablishment of blood flow is essential for salvaging ischemic tissues. The reperfusion itself, however, can paradoxically lead to further cellular damage, which involves a multi-factorial process resulting in extensive tissue damage, which can threaten the function and viability of the liver and other organ systems. The following review outlines multiple models for in-lab analysis of the various hepatic IRI mechanisms, including murine, porcine, cell lines, and machine perfusion models.
Collapse
Affiliation(s)
- Connor Whalen
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Arun Verma
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Kento Kurashima
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Jasmine Carter
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Hala Nazzal
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Ajay Jain
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, MO 63104, USA
| |
Collapse
|
8
|
Chen H, Ellis BW, Dinicu AT, Mojoudi M, Wilks BT, Tessier SN, Toner M, Uygun K, Uygun BE. Polyethylene glycol and caspase inhibitor emricasan alleviate cold injury in primary rat hepatocytes. Cryobiology 2024; 116:104926. [PMID: 38880369 PMCID: PMC11374468 DOI: 10.1016/j.cryobiol.2024.104926] [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] [Received: 12/01/2023] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Current methods of storing explanted donor livers at 4 °C in University of Wisconsin (UW) solution result in loss of graft function and ultimately lead to less-than-ideal outcomes post transplantation. Our lab has previously shown that supplementing UW solution with 35-kilodalton polyethylene glycol (PEG) has membrane stabilizing effects for cold stored primary rat hepatocytes in suspension. Expanding on past studies, we here investigate if PEG has the same beneficial effects in an adherent primary rat hepatocyte cold storage model. In addition, we investigated the extent of cold-induced apoptosis through treating cold-stored hepatocytes with pan caspase inhibitor emricasan. In parallel to storage at the current cold storage standard of 4 °C, we investigated the effects of lowering the storage temperature to -4 °C, at which the storage solution remains ice-free due to the supercooling phenomenon. We show the addition of 5 % PEG to the storage medium significantly reduced the release of lactate dehydrogenase (LDH) in plated rat hepatocytes and a combinatorial treatment with emricasan maintains hepatocyte viability and morphology following recovery from cold storage. These results show that cold-stored hepatocytes undergo multiple mechanisms of cold-induced injury and that PEG and emricasan treatment in combination with supercooling may improve cell and organ preservation.
Collapse
Affiliation(s)
- Huyun Chen
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Children's Boston, Boston, MA, USA
| | - Bradley W Ellis
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Children's Boston, Boston, MA, USA
| | - Antonia T Dinicu
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Children's Boston, Boston, MA, USA
| | - Mohammadreza Mojoudi
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Children's Boston, Boston, MA, USA
| | - Benjamin T Wilks
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Children's Boston, Boston, MA, USA
| | - Shannon N Tessier
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Children's Boston, Boston, MA, USA
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Children's Boston, Boston, MA, USA
| | - Korkut Uygun
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Children's Boston, Boston, MA, USA
| | - Basak E Uygun
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Shriners Children's Boston, Boston, MA, USA.
| |
Collapse
|
9
|
Eldafashi N, Waaz S, Ali TFS, Zaki MYW, Nazmy MH, Fathy M. The protective role of two oxindole derivatives is mediated by modulating NLRP3/caspase-1 and PI3K/AKT pathways in a preclinical animal model of hepatic ischemia reperfusion injury. Life Sci 2024; 352:122872. [PMID: 38942361 DOI: 10.1016/j.lfs.2024.122872] [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: 04/25/2024] [Revised: 06/05/2024] [Accepted: 06/23/2024] [Indexed: 06/30/2024]
Abstract
Aim Hepatic ischemia reperfusion injury (HIRI) is a leading cause of mortality post liver transplantation, hypovolemic shock and trauma. In this study, we tested, on molecular bases, the possible protective role of two different derivatives of 2-oxindole in a preclinical model of HIRI in rats. MAIN METHODS HIRI was operated in male Wistar albino rats and prophylactic treatment with oxindole-curcumin (Coxi) or oxindole-vanillin (Voxi) was carried out before the operation. The biochemical and histopathological investigations, in addition to the mechanistic characterizations of the effect of the tested drugs were performed. KEY FINDINGS HIRI was assured with elevated liver enzymes and marked changes in histopathological features, inflammatory response and oxidative stress. Pretreatment with Coxi and Voxi improved the hepatic histopathological alterations, reduced the elevated serum liver enzymes level and hepatic Malondialdehyde (MDA) content, increased the hepatic Superoxide Dismutase (SOD) activity and reduced Glutathione (GSH) content, downregulated the expression of TNF-α, IL-6, Nod-Like Receptor p3 (NLRP3), Cleaved caspase1, Cleaved caspase 3 proteins, alongside the expression level of IL-1β, ICAM-1, VCAM-1 and BAX genes, attenuated NF-кB p-P65 Ser536 and Myeloperoxidase (MPO)-positive neutrophils, and activated the PI3K/AKT pathway. SIGNIFICANCE Coxi and Voxi have promising hepatoprotective activity against HIRI in rats through ameliorating the biochemical and histopathological alterations, attenuating inflammatory and oxidative stress status by modulating the inflammatory TNF-α/ICAM-1, the pyroptosis NLRP3/Caspase-1, and the antioxidant PI3K/AKT pathways.
Collapse
Affiliation(s)
- Nardeen Eldafashi
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
| | - Shaimaa Waaz
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
| | - Taha F S Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
| | - Marco Y W Zaki
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
| | - Maiiada Hassan Nazmy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
| | - Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.
| |
Collapse
|
10
|
George J, Lu Y, Tsuchishima M, Tsutsumi M. Cellular and molecular mechanisms of hepatic ischemia-reperfusion injury: The role of oxidative stress and therapeutic approaches. Redox Biol 2024; 75:103258. [PMID: 38970988 PMCID: PMC11279328 DOI: 10.1016/j.redox.2024.103258] [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: 03/25/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024] Open
Abstract
Ischemia-reperfusion (IR) or reoxygenation injury is the paradoxical exacerbation of cellular impairment following restoration of blood flow after a period of ischemia during surgical procedures or other conditions. Acute interruption of blood supply to the liver and subsequent reperfusion can result in hepatocyte injury, apoptosis, and necrosis. Since the liver requires a continuous supply of oxygen for many biochemical reactions, any obstruction of blood flow can rapidly lead to hepatic hypoxia, which could quickly progress to absolute anoxia. Reoxygenation results in the increased generation of reactive oxygen species and oxidative stress, which lead to the enhanced production of proinflammatory cytokines, chemokines, and other signaling molecules. Consequent acute inflammatory cascades lead to significant impairment of hepatocytes and nonparenchymal cells. Furthermore, the expression of several vascular growth factors results in the heterogeneous closure of numerous hepatic sinusoids, which leads to reduced oxygen supply in certain areas of the liver even after reperfusion. Therefore, it is vital to identify appropriate therapeutic modalities to mitigate hepatic IR injury and subsequent tissue damage. This review covers all the major aspects of cellular and molecular mechanisms underlying the pathogenesis of hepatic ischemia-reperfusion injury, with special emphasis on oxidative stress, associated inflammation and complications, and prospective therapeutic approaches.
Collapse
Affiliation(s)
- Joseph George
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA; Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan; Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa, 920-0293, Japan.
| | - Yongke Lu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Mutsumi Tsuchishima
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Mikihiro Tsutsumi
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan; Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa, 920-0293, Japan
| |
Collapse
|
11
|
Aboelez MO, Ezelarab HAA, Alotaibi G, Abouzed DEE. Inflammatory setting, therapeutic strategies targeting some pro-inflammatory cytokines and pathways in mitigating ischemia/reperfusion-induced hepatic injury: a comprehensive review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6299-6315. [PMID: 38643452 DOI: 10.1007/s00210-024-03074-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 03/28/2024] [Indexed: 04/22/2024]
Abstract
Ischemia/reperfusion injury (IRI) is a key determining agent in the pathophysiology of clinical organ dysfunction. It is characterized by an aseptic local inflammatory reaction due to a decrease in blood supply, hence deprivation of dependent oxygen and nutrients. In instances of liver transplantation, this injury may have irreversible implications, resulting in eventual organ rejection. The deterioration associated with IRI is affected by the hepatic health status and various factors such as alterations in metabolism, oxidative stress, and pro-inflammatory cytokines. The primary cause of inflammation is the initial immune response of pro-inflammatory cytokines, while Kupffer cells (KFCs) and neutrophil-produced chemokines also play a significant role. Upon reperfusion, the activation of inflammatory responses can elicit further cellular damage and organ dysfunction. This review discusses the interplay between chemokines, pro-inflammatory cytokines, and other inflammatory mediators that contribute to the damage to hepatocytes and liver failure in rats following IR. Furthermore, it delves into the impact of anti-inflammatory therapies in safeguarding against liver failure and hepatocellular damage in rats following IR. This review investigates the correlation between cytokine factors and liver dysfunction via examining databases, such as PubMed, Google Scholar, Science Direct, Egyptian Knowledge Bank (EKB), and Research Gate.
Collapse
Affiliation(s)
- Moustafa O Aboelez
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sohag University, Sohag, 82524, Egypt.
| | - Hend A A Ezelarab
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minya, 61519, Egypt.
| | - Ghallab Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Al-Dawadmi Campus, 11961, Al-Dawadmi, Saudi Arabia
| | - Deiaa E Elsayed Abouzed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sohag University, Sohag, 82524, Egypt
| |
Collapse
|
12
|
Song S, Li R, Wu C, Dong J, Wang P. EFFECTS OF HYPERBARIC OXYGEN THERAPY ON INTESTINAL ISCHEMIA-REPERFUSION AND ITS MECHANISM. Shock 2024; 61:650-659. [PMID: 38113056 DOI: 10.1097/shk.0000000000002287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
ABSTRACT Ischemia can cause reversible or irreversible cell or tissue damage, and reperfusion after ischemia not only has no therapeutic effect but also aggravates cell damage. Notably, gut tissue is highly susceptible to ischemia-reperfusion (IR) injury under many adverse health conditions. Intestinal IR (IIR) is an important pathophysiological process in critical clinical diseases. Therefore, it is necessary to identify better therapeutic methods for relieving intestinal ischemia and hypoxia. Hyperbaric oxygenation refers to the intermittent inhalation of 100% oxygen in an environment greater than 1 atm pressure, which can better increase the oxygen level in the tissue and change the inflammatory pathway. Currently, it can have a positive effect on hypoxia and ischemic diseases. Related studies have suggested that hyperbaric oxygen can significantly reduce ischemia-hypoxic injury to the brain, spinal cord, kidney, and myocardium. This article reviews the pathogenesis of IR and the current treatment measures, and further points out that hyperbaric oxygen has a better effect in IR. We found that not only improved hypoxia but also regulated IR induced injury in a certain way. From the perspective of clinical application, these changes and the application of hyperbaric oxygen therapy have important implications for treatment, especially IIR.
Collapse
Affiliation(s)
- Shurui Song
- Department of Emergency Surgery, The Affiliated Hospital of Qing Dao University, Qing Dao, PR China
| | - Ruojing Li
- Department of Emergency Surgery, The Affiliated Hospital of Qing Dao University, Qing Dao, PR China
| | - Changliang Wu
- Department of Emergency Surgery, The Affiliated Hospital of Qing Dao University, Qing Dao, PR China
| | | | - Peige Wang
- Department of Emergency Surgery, The Affiliated Hospital of Qing Dao University, Qing Dao, PR China
| |
Collapse
|
13
|
Kim JW, Kim YJ. The evidence-based multifaceted roles of hepatic stellate cells in liver diseases: A concise review. Life Sci 2024; 344:122547. [PMID: 38460810 DOI: 10.1016/j.lfs.2024.122547] [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: 12/25/2023] [Revised: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 03/11/2024]
Abstract
Hepatic stellate cells (HSCs) play central roles in liver disease pathogenesis, spanning steatosis to cirrhosis and hepatocellular carcinoma. These cells, located in the liver's sinusoidal space of Disse, transition from a quiescent, vitamin A-rich state to an activated, myofibroblast-like phenotype in response to liver injury. This activation results from a complex interplay of cytokines, growth factors, and oxidative stress, leading to excessive collagen deposition and liver fibrosis, a hallmark of chronic liver diseases. Recently, HSCs have gained recognition for their dynamic, multifaceted roles in liver health and disease. Attention has shifted toward their involvement in various liver conditions, including acute liver injury, alcoholic and non-alcoholic fatty liver disease, and liver regeneration. This review aims to explore diverse functions of HSCs in these acute or chronic liver pathologies, with a focus on their roles beyond fibrogenesis. HSCs exhibit a wide range of actions, including lipid storage, immunomodulation, and interactions with other hepatic and extrahepatic cells, making them pivotal in the hepatic microenvironment. Understanding HSC involvement in the progression of liver diseases can offer novel insights into pathogenic mechanisms and guide targeted therapeutic strategies for various liver conditions.
Collapse
Affiliation(s)
- Jong-Won Kim
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Yu Ji Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Medical School, Jeonbuk National University, Research Institute of Clinical Medicine of Jeonbuk National University - Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea.
| |
Collapse
|
14
|
Gao Y, Wang M, Qin R, Zhao C, Gong J. METTL3 Deficiency Aggravates Hepatic Ischemia/Reperfusion Injury in Mice by Activating the MAPK Signaling Pathway. Int J Med Sci 2024; 21:1037-1048. [PMID: 38774758 PMCID: PMC11103385 DOI: 10.7150/ijms.94177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/05/2024] [Indexed: 05/24/2024] Open
Abstract
Background: Inflammatory responses, apoptosis, and oxidative stress, are key factors that contribute to hepatic ischemia/reperfusion (I/R) injury, which may lead to the failure of liver surgeries, such as hepatectomy and liver transplantation. The N6-methyladenosine (m6A) modification has been implicated in multiple biological processes, and its specific role and mechanism in hepatic I/R injury require further investigation. Methods: Dot blotting analysis was used to profile m6A levels in liver tissues at different reperfusion time points in hepatic I/R mouse models. Hepatocyte-specific METTL3 knockdown (HKD) mice were used to determine the function of METTL3 during hepatic I/R. RNA sequencing and western blotting were performed to assess the potential signaling pathways involved with the deficiency of METTL3. Finally, AAV8-TBG-METTL3 was injected through the tail vein to further elucidate the role of METTL3 in hepatic I/R injury. Results: The m6A modification levels and the expression of METTL3 were upregulated in mouse livers during hepatic I/R injury. METTL3 deficiency led to an exacerbated inflammatory response and increased cell death during hepatic I/R, whereas overexpression of METTL3 reduced the extent of liver injury. Bioinformatic analysis revealed that the MAPK pathway was significantly enriched in the livers of METTL3-deficient mice. METTL3 protected the liver from I/R injury, possibly by inhibiting the phosphorylation of JNK and ERK, but not P38. Conclusions: METTL3 deficiency aggravates hepatic I/R injury in mice by activating the MAPK signaling pathway. METTL3 may be a potential therapeutic target in hepatic I/R injury.
Collapse
Affiliation(s)
- Yang Gao
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Renyi Qin
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Chunle Zhao
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| | - Jun Gong
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan, 430030, Hubei, China
| |
Collapse
|
15
|
Nakano R, Chogahara I, Ohira M, Imaoka K, Sato S, Bekki T, Sato K, Imaoka Y, Marlen D, Tanaka Y, Ohdan H. Atherosclerosis Deteriorates Liver Ischemia/Reperfusion Injury Via Interferon Regulatory Factor-1 Overexpression in a Murine Model. Transplant Proc 2024; 56:678-685. [PMID: 38433025 DOI: 10.1016/j.transproceed.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/04/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Abdominal aortic calcification (AAC) is associated with cardiovascular-related mortality, along with an elevated risk of coronary, cerebrovascular, and cardiovascular events. Notably, AAC is strongly associated with poor overall and recurrence free survival posthepatectomy for hepatocellular carcinoma. Despite the acknowledged significance of atherosclerosis in systemic inflammation, its response to ischemia/reperfusion injury (IRI) remains poorly elucidated. In this study, we aimed to clarify the impact of atherosclerosis on the liver immune system using a warm IRI mouse model. METHODS Injury was induced in an atherosclerotic mouse model (ApoE-/-) or C57BL/6J wild-type (WT) mice through 70% clamping for 1 hour and analyzed after 6 hours of reperfusion. RESULTS Elevated serum levels of aspartate and alanine aminotransferase, along with histological assessment, indicated considerable damage in the livers of ApoE-/- mice than that in WT mice. This indicates a substantial contribution of atherosclerosis to IRI. Furthermore, T and natural killer (NK) cells in ApoE-/- mouse livers displayed a more inflammatory phenotype than those in WT mouse livers. Reverse transcription-polymerase chain reaction analysis revealed a significant upregulation of interleukin (IL)-15 and its transcriptional regulator, interferon regulatory factor-1 (IRF-1) in ApoE-/- mouse livers compared with that in WT mouse livers. CONCLUSIONS These findings suggest that in an atherosclerotic mouse model, atherosclerosis can mirror intrahepatic immunity, particularly activating liver NK and T cells through IL-15 production, thereby exacerbating hepatic damage. The upregulation of IL-15 expression is associated with IRF-1 overexpression.
Collapse
Affiliation(s)
- Ryosuke Nakano
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Ichiya Chogahara
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Masahiro Ohira
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan; Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Minami-ku, Hiroshima, Japan.
| | - Kouki Imaoka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Saki Sato
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Tomoaki Bekki
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Koki Sato
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Yuki Imaoka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Doskali Marlen
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| |
Collapse
|
16
|
Cabanes-Creus M, Liao SHY, Gale Navarro R, Knight M, Nazareth D, Lau NS, Ly M, Zhu E, Roca-Pinilla R, Bugallo Delgado R, Vicente AF, Baltazar G, Westhaus A, Merjane J, Crawford M, McCaughan GW, Unzu C, González-Aseguinolaza G, Alexander IE, Pulitano C, Lisowski L. Harnessing whole human liver ex situ normothermic perfusion for preclinical AAV vector evaluation. Nat Commun 2024; 15:1876. [PMID: 38485924 PMCID: PMC10940703 DOI: 10.1038/s41467-024-46194-y] [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: 07/11/2023] [Accepted: 02/19/2024] [Indexed: 03/18/2024] Open
Abstract
Developing clinically predictive model systems for evaluating gene transfer and gene editing technologies has become increasingly important in the era of personalized medicine. Liver-directed gene therapies present a unique challenge due to the complexity of the human liver. In this work, we describe the application of whole human liver explants in an ex situ normothermic perfusion system to evaluate a set of fourteen natural and bioengineered adeno-associated viral (AAV) vectors directly in human liver, in the presence and absence of neutralizing human sera. Under non-neutralizing conditions, the recently developed AAV variants, AAV-SYD12 and AAV-LK03, emerged as the most functional variants in terms of cellular uptake and transgene expression. However, when assessed in the presence of human plasma containing anti-AAV neutralizing antibodies (NAbs), vectors of human origin, specifically those derived from AAV2/AAV3b, were extensively neutralized, whereas AAV8- derived variants performed efficiently. This study demonstrates the potential of using normothermic liver perfusion as a model for early-stage testing of liver-focused gene therapies. The results offer preliminary insights that could help inform the development of more effective translational strategies.
Collapse
Affiliation(s)
- Marti Cabanes-Creus
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia
| | - Sophia H Y Liao
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia
| | - Renina Gale Navarro
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia
| | - Maddison Knight
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia
| | - Deborah Nazareth
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia
| | - Ngee-Soon Lau
- Australian National Liver Transplantation Unit, Royal Prince Alfred Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Organ Assessment Repair and Optimisation, Royal Prince Alfred Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Mark Ly
- Australian National Liver Transplantation Unit, Royal Prince Alfred Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Organ Assessment Repair and Optimisation, Royal Prince Alfred Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Erhua Zhu
- Gene Therapy Research Unit, Children's Medical Research Institute and The Children's Hospital at Westmead, Faculty of Medicine and Health, The University of Sydney, and Sydney Children's Hospitals Network, Sydney, Westmead, Australia
| | - Ramon Roca-Pinilla
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia
| | - Ricardo Bugallo Delgado
- Gene Therapy and Regulation of Gene Expression Department, IdiSNA, Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, CIMA, Pamplona, Spain
| | - Ana F Vicente
- Gene Therapy and Regulation of Gene Expression Department, IdiSNA, Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, CIMA, Pamplona, Spain
| | - Grober Baltazar
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia
| | - Adrian Westhaus
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia
| | - Jessica Merjane
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia
| | - Michael Crawford
- Australian National Liver Transplantation Unit, Royal Prince Alfred Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Organ Assessment Repair and Optimisation, Royal Prince Alfred Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Geoffrey W McCaughan
- Australian National Liver Transplantation Unit, Royal Prince Alfred Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Liver Injury and Cancer Program, Centenary Research Institute, A.W Morrow Gastroenterology and Liver Centre, Sydney, Australia
| | - Carmen Unzu
- Gene Therapy and Regulation of Gene Expression Department, IdiSNA, Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, CIMA, Pamplona, Spain
| | - Gloria González-Aseguinolaza
- Gene Therapy and Regulation of Gene Expression Department, IdiSNA, Instituto de Investigación Sanitaria de Navarra, Universidad de Navarra, CIMA, Pamplona, Spain
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute and The Children's Hospital at Westmead, Faculty of Medicine and Health, The University of Sydney, and Sydney Children's Hospitals Network, Sydney, Westmead, Australia
- Discipline of Child and Adolescent Health, The University of Sydney, Sydney Medical School, Faculty of Medicine and Health, Sydney, Westmead, Australia
- Australian Genome Therapeutics Centre, Children's Medical Research Institute and Sydney Children's Hospitals Network, Sydney, Westmead, Australia
| | - Carlo Pulitano
- Australian National Liver Transplantation Unit, Royal Prince Alfred Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Centre for Organ Assessment Repair and Optimisation, Royal Prince Alfred Hospital, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Leszek Lisowski
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, Westmead, Australia.
- Australian Genome Therapeutics Centre, Children's Medical Research Institute and Sydney Children's Hospitals Network, Sydney, Westmead, Australia.
- Military Institute of Medicine - National Research Institute, Laboratory of Molecular Oncology and Innovative Therapies, Warsaw, Poland.
| |
Collapse
|
17
|
Nakatake R, Okuyama T, Ishizaki M, Yanagida H, Kitade H, Yoshizawa K, Nishizawa M, Sekimoto M. Hepatoprotection of a Standardized Extract of Cultured Lentinula edodes Mycelia against Liver Injury Induced by Ischemia-Reperfusion and Partial Hepatectomy. Nutrients 2024; 16:256. [PMID: 38257149 PMCID: PMC10820669 DOI: 10.3390/nu16020256] [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: 12/06/2023] [Revised: 01/08/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
A standardized extract of cultured Lentinula edodes mycelia (ECLM, AHCC®) has been shown to have beneficial effects on organ metabolism. ECLM has been indicated to have liver protective properties by suppressing inflammatory responses. The pathogenesis of hepatic ischemia-reperfusion injury is thought to involve the induction of inflammatory mediators. However, whether ECLM affects inflammatory mediators caused by warm hepatic ischemia-reperfusion injury and partial hepatectomy (HIRI+PH) has not been clarified. In this study, we evaluated the protective effects of ECLM against liver damage caused by HIRI+PH. Rats were fed a normal diet (HIRI+PH) or a normal diet with 2% ECLM (HIRI+PH and ECLM) for ten days, then the liver and duodenal ligament were clamped and subjected to 15 min of hepatic ischemia. After 70% hepatectomy, the inflow occlusion was released, and liver and blood samples were collected at 3, 6, and 24 h. The effect of ECLM on mortality induced by 30 min of ischemia and hepatectomy was evaluated. The results showed that ECLM attenuated pathological liver damage, including apoptosis, in the rats treated with HIRI+PH, and decreased serum aminotransferase activity; ECLM decreased mRNA levels of the inflammation-related genes inducible nitric oxide synthase and C-X-C motif chemokine ligand 1, and increased mRNA levels of interleukin 10, an anti-inflammatory cytokine; ECLM increased hepatocyte growth factor mRNA levels and Ki-67 labeled nuclei in the liver at 24 h; ECLM significantly reduced HIRI+PH-induced mortality. In conclusion, ECLM may prevent HIRI+PH-induced liver injury in part by suppressing various inflammatory responses and promoting liver regeneration.
Collapse
Affiliation(s)
- Richi Nakatake
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Osaka, Japan; (T.O.)
| | - Tetsuya Okuyama
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Osaka, Japan; (T.O.)
| | - Morihiko Ishizaki
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Osaka, Japan; (T.O.)
| | - Hidesuke Yanagida
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Osaka, Japan; (T.O.)
| | - Hiroaki Kitade
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Osaka, Japan; (T.O.)
| | - Katsuhiko Yoshizawa
- Department of Innovative Food Sciences, School of Food Sciences and Nutrition, Mukogawa Women’s University, 6-46 Ikebiraki-cho, Nishinomiya 663-8558, Hyogo, Japan;
| | - Mikio Nishizawa
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu 525-8577, Shiga, Japan;
| | - Mitsugu Sekimoto
- Department of Surgery, Kansai Medical University, Hirakata 573-1010, Osaka, Japan; (T.O.)
| |
Collapse
|
18
|
Chen H, Ellis BW, Dinicu AT, Mojoudi M, Wilks BT, Tessier SN, Toner M, Uygun K, Uygun BE. Polyethylene Glycol and Caspase Inhibitor Emricasan Alleviates Cold Injury in Primary Rat Hepatocytes. RESEARCH SQUARE 2023:rs.3.rs-3669876. [PMID: 38076969 PMCID: PMC10705698 DOI: 10.21203/rs.3.rs-3669876/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Current methods of storing explanted donor livers at 4°C in University of Wisconsin (UW) solution result in loss of graft function and ultimately leads to less-than-ideal outcomes post transplantation. Our lab has previously shown that supplementing UW solution with 35-kilodalton polyethylene glycol (PEG) has membrane stabilizing effects for cold stored primary rat hepatocytes in suspension. Expanding on past studies, we here investigate if PEG has the same beneficial effects in an adherent primary rat hepatocyte cold storage model. In addition, we investigated the extent of cold-induced apoptosis through treating cold-stored hepatocytes with pan caspase inhibitor emricasan. In parallel to storage at the current cold storage standard of 4°C, we investigated the effects of lowering the storage temperature to -4°C, at which the storage solution remains ice-free due to the supercooling phenomenon. We show the addition of 5% PEG to the storage medium significantly reduced the release of lactate dehydrogenase (LDH) in plated rat hepatocytes and a combinatorial treatment with emricasan maintains hepatocyte viability and morphology following recovery from cold storage. These results show that cold-stored hepatocytes undergo multiple mechanisms of cold-induced injury and that PEG and emricasan treatment in combination with supercooling may improve cell and organ preservation.
Collapse
Affiliation(s)
- Huyun Chen
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital
| | - Bradley W Ellis
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital
| | - Antonia T Dinicu
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital
| | - Mohammadreza Mojoudi
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital
| | - Benjamin T Wilks
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital
| | - Shannon N Tessier
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital
| | - Korkut Uygun
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital
| | - Basak E Uygun
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital
| |
Collapse
|
19
|
Felli E, Felli E, Muttillo EM, Urade T, Laracca GG, Giannelli V, Famularo S, Geny B, Ettorre GM, Rombouts K, Pinzani M, Diana M, Gracia-Sancho J. Liver ischemia-reperfusion injury: From trigger loading to shot firing. Liver Transpl 2023; 29:1226-1233. [PMID: 37728488 DOI: 10.1097/lvt.0000000000000252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/15/2023] [Indexed: 09/21/2023]
Abstract
An ischemia-reperfusion injury (IRI) results from a prolonged ischemic insult followed by the restoration of blood perfusion, being a common cause of morbidity and mortality, especially in liver transplantation. At the maximum of the potential damage, IRI is characterized by 2 main phases. The first is the ischemic phase, where the hypoxia and vascular stasis induces cell damage and the accumulation of damage-associated molecular patterns and cytokines. The second is the reperfusion phase, where the local sterile inflammatory response driven by innate immunity leads to a massive cell death and impaired liver functionality. The ischemic time becomes crucial in patients with underlying pathophysiological conditions. It is possible to compare this process to a shooting gun, where the loading trigger is the ischemia period and the firing shot is the reperfusion phase. In this optic, this article aims at reviewing the main ischemic events following the phases of the surgical timeline, considering the consequent reperfusion damage.
Collapse
Affiliation(s)
- Eric Felli
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland
| | - Emanuele Felli
- Department of Digestive Surgery and Liver Transplantation, University Hospital of Tours, France
| | - Edoardo M Muttillo
- Department of Medical Surgical Science and Translational Medicine, Sant' Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Takeshi Urade
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Japan
| | - Giovanni G Laracca
- Department of Medical Surgical Science and Translational Medicine, Sant' Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Valerio Giannelli
- Department of Transplantation and General Surgery, San Camillo Hospital, Italy
| | - Simone Famularo
- Department of Biomedical Science, Humanitas University Pieve Emanuele, Italy
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Research Institute Against Cancer of the Digestive System (IRCAD), France
| | - Bernard Geny
- Institute of Physiology, EA3072 Mitochondria Respiration and Oxidative Stress, University of Strasbourg, France
| | - Giuseppe M Ettorre
- Department of Transplantation and General Surgery, San Camillo Hospital, Italy
| | - Krista Rombouts
- University College London - Institute for Liver and Digestive Health, Royal Free Hospital, NW3 2PF London, United Kingdom
| | - Massimo Pinzani
- University College London - Institute for Liver and Digestive Health, Royal Free Hospital, NW3 2PF London, United Kingdom
| | - Michele Diana
- Research Institute Against Cancer of the Digestive System (IRCAD), France
| | - Jordi Gracia-Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Switzerland
- Department for BioMedical Research, Visceral Surgery and Medicine, University of Bern, Switzerland
- Liver Vascular Biology Research Group, IDIBAPS Biomedical Research Institute, Hospital Clínic Barcelona, CIBEREHD, Barcelona, Spain
| |
Collapse
|
20
|
Ma Y, Zhao C, Hu H, Yin S. Liver protecting effects and molecular mechanisms of icariin and its metabolites. PHYTOCHEMISTRY 2023; 215:113841. [PMID: 37660725 DOI: 10.1016/j.phytochem.2023.113841] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/05/2023]
Abstract
As a detoxification and metabolism organ, the liver plays a vital role in human health. However, an excessive consumption of drugs and toxins, exposure to pathogenic viruses, and unhealthy living habits can lead to liver damage, which may even develop into liver cirrhosis and liver cancer. Epimedium brevicornum Maxim. is a traditional Chinese medicine and dietary supplement in which the flavonoid icariin is a main functional component. Although the protective mechanisms of icariin and its metabolites against liver injury are not yet comprehensively understood, an increasing number of studies have confirmed their liver-protective and anticancer effects. Indeed, icaritin, one of the metabolites of icariin, is currently utilized as an active component of an anti-cancer drug. This paper presents a review of the molecular mechanisms through which icariin and its metabolites actively protect against the occurrence and development of liver injury, and, thus, provides a comprehensive reference for further research and their application in liver protection.
Collapse
Affiliation(s)
- Yurong Ma
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| | - Chong Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| | - Hongbo Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| | - Shutao Yin
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
21
|
Mahmoud HM, Elsayed Abouzed DE, Abo-Youssef AM, Hemeida RAM. Zafirlukast protects against hepatic ischemia-reperfusion injury in rats via modulating Bcl-2/Bax and NF-κB/SMAD-4 pathways. Int Immunopharmacol 2023; 122:110498. [PMID: 37418987 DOI: 10.1016/j.intimp.2023.110498] [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: 02/24/2023] [Revised: 06/10/2023] [Accepted: 06/11/2023] [Indexed: 07/09/2023]
Abstract
Hepatic ischemia/reperfusion injury (IRI) is a clinical problem commonly during liver transplantation and other liver surgery. This study aimed to evaluate the protective effect of zafirlukast (ZFK) on IR-induced hepatic injury and investigate its relevant protective mechanism. Thirty-two male Wistar albino rats were randomly allocated to four groups: sham, IRI, ZFK, and ZFK + IR groups. ZFK was administered orally in a dose of 80 mg/kg/day for 10 consecutive days. Serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), total bilirubin (TBL) levels, and gamma glutamyl transferase (GGT) activity were estimated. Liver tissues were used to assess oxidative stress biomarkers including malondialdehyde (MDA), myeloperoxidase (MPO), nitric oxide (NOx), and reduced glutathione (GSH) contents. Inflammatory cytokines, tumor necrosis factor alpha (TNF-α) and interleukin-33 (IL-33), in addition to apoptosis biomarkers, BCL2 associated X protein (Bax), B-cell lymphoma 2 (Bcl2) and galactine-9 (GAL9) proteins were also assessed. Western blot analysis was performed for vascular endothelial growth factor (VEGF) and fibrinogen expressions. Immunohistochemical analysis for hepatic nuclear factor-kappa B (NF-κB) and SMAD-4 was done in addition to histopathological examination. Our study revealed that ZFK pre-treatment resulted in liver function restoration and oxidative stress correction. Moreover, inflammatory cytokines were significantly reduced and a remarkable reduction of apoptosis, angiogenesis, and clotting formation has been indicated. Additionally, a significant reduction in SMAD-4 and NF-kB protein expressions was observed. These results were supported by hepatic architecture improvement. Our findings revealed that ZFK possesses a potential protective effect against liver IR possibly through its antioxidant, anti-inflammatory and anti-apoptotic properties.
Collapse
Affiliation(s)
- Heba M Mahmoud
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Deiaa E Elsayed Abouzed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Sohag University, Sohag, 82524, Egypt.
| | - Amira M Abo-Youssef
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Ramadan A M Hemeida
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt; Department of Pharmacology & Toxicology, Faculty of Pharmacy, Deraya University, Minya, 61519, Egypt.
| |
Collapse
|
22
|
Wang L, Feng ZJ, Ma X, Li K, Li XY, Tang Y, Peng C. Mitochondrial quality control in hepatic ischemia-reperfusion injury. Heliyon 2023; 9:e17702. [PMID: 37539120 PMCID: PMC10395149 DOI: 10.1016/j.heliyon.2023.e17702] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 06/07/2023] [Accepted: 06/26/2023] [Indexed: 08/05/2023] Open
Abstract
Hepatic ischemia-reperfusion injury is a phenomenon in which exacerbating damage of liver cells due to restoration of blood flow following ischemia during liver surgery, especially those involving liver transplantation. Mitochondria, the energy-producing organelles, are crucial for cell survival and apoptosis and have evolved a range of quality control mechanisms to maintain homeostasis in the mitochondrial network in response to various stress conditions. Hepatic ischemia-reperfusion leads to disruption of mitochondrial quality control mechanisms, as evidenced by reduced mitochondrial autophagy, excessive division, reduced fusion, and inhibition of biogenesis. This leads to dysfunction of the mitochondrial network. The accumulation of damaged mitochondria ultimately results in apoptosis of hepatocytes due to the release of apoptotic proteins like cytochrome C. This worsens hepatic ischemia-reperfusion injury. Currently, hepatic ischemia-reperfusion injury protection is being studied using different approaches such as drug pretreatment, stem cells and exosomes, genetic interventions, and mechanical reperfusion, all aimed at targeting mitochondrial quality control mechanisms. This paper aims to provide direction for future research on combating HIRI by reviewing the latest studies that focus on targeting mitochondrial quality control mechanisms.
Collapse
Affiliation(s)
- LiuSong Wang
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zan Jie Feng
- Department of Biochemistry and Molecular Biology, Zunyi Medical University, Zunyi, China
| | - Xuan Ma
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Kai Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xin Yao Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi Tang
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Cijun Peng
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
23
|
Blackberry-Loaded AgNPs Attenuate Hepatic Ischemia/Reperfusion Injury via PI3K/Akt/mTOR Pathway. Metabolites 2023; 13:metabo13030419. [PMID: 36984859 PMCID: PMC10051224 DOI: 10.3390/metabo13030419] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Liver ischemia-reperfusion injury (IRI) is a pathophysiological insult that often occurs during liver surgery. Blackberry leaves are known for their anti-inflammatory and antioxidant activities. Aims: To achieve site-specific delivery of blackberry leaves extract (BBE) loaded AgNPs to the hepatocyte in IRI and to verify possible molecular mechanisms. Methods: IRI was induced in male Wister rats. Liver injury, hepatic histology, oxidative stress markers, hepatic expression of apoptosis-related proteins were evaluated. Non-targeted metabolomics for chemical characterization of blackberry leaves extract was performed. Key findings: Pre-treatment with BBE protected against the deterioration caused by I/R, depicted by a significant improvement of liver functions and structure, as well as reduction of oxidative stress with a concomitant increase in antioxidants. Additionally, BBE promoted phosphorylation of antiapoptotic proteins; PI3K, Akt and mTOR, while apoptotic proteins; Bax, Casp-9 and cleaved Casp-3 expressions were decreased. LC-HRMS-based metabolomics identified a range of metabolites, mainly flavonoids and anthocyanins. Upon comprehensive virtual screening and molecular dynamics simulation, the major annotated anthocyanins, cyanidin and pelargonidin glucosides, were suggested to act as PLA2 inhibitors. Significance: BBE can ameliorate hepatic IRI augmented by BBE-AgNPs nano-formulation via suppressing, oxidative stress and apoptosis as well as stimulation of PI3K/Akt/mTOR signaling pathway.
Collapse
|
24
|
Inhibition of γδ-TcR or IL17a Reduces T-Cell and Neutrophil Infiltration after Ischemia/Reperfusion Injury in Mouse Liver. J Clin Med 2023; 12:jcm12051751. [PMID: 36902538 PMCID: PMC10002490 DOI: 10.3390/jcm12051751] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/08/2023] [Accepted: 02/12/2023] [Indexed: 02/24/2023] Open
Abstract
Neutrophil and T-cell recruitment contribute to hepatic ischemia/reperfusion injury. The initial inflammatory response is orchestrated by Kupffer cells and liver sinusoid endothelial cells. However, other cell types, including γδ-Τ cells, seem to be key mediators in further inflammatory cell recruitment and proinflammatory cytokine release, including IL17a. In this study, we used an in vivo model of partial hepatic ischemia/reperfusion injury (IRI) to investigate the role of the γδ-Τ-cell receptor (γδTcR) and the role of IL17a in the pathogenesis of liver injury. Forty C57BL6 mice were subjected to 60 min of ischemia followed by 6 h of reperfusion (RN 6339/2/2016). Pretreatment with either anti-γδΤcR antibodies or anti-IL17a antibodies resulted in a reduction in histological and biochemical markers of liver injury as well as neutrophil and T-cell infiltration, inflammatory cytokine production and the downregulation of c-Jun and NF-κΒ. Overall, neutralizing either γδTcR or IL17a seems to have a protective role in liver IRI.
Collapse
|
25
|
Peng Q, Nowocin A, Ratnasothy K, Smith RA, Smyth LA, Lechler RI, Dorling A, Lombardi G. Inhibition of thrombin on endothelium enhances recruitment of regulatory T cells during IRI and when combined with adoptive Treg transfer, significantly protects against acute tissue injury and prolongs allograft survival. Front Immunol 2023; 13:980462. [PMID: 36793549 PMCID: PMC9924086 DOI: 10.3389/fimmu.2022.980462] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/28/2022] [Indexed: 01/31/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) amplifies T cell alloimmune responses after transplantation with thrombin playing a key pro-inflammatory role. To explore the influence of thrombin on regulatory T cell recruitment and efficacy we used a well-established model of IRI in the native murine kidney. Administration of the cytotopic thrombin inhibitor PTL060 inhibited IRI, and by skewing expression of chemokines (reducing CCL2 and CCL3 but increasing CCL17 and CCL22) increased the infiltration of M2 macrophages and Tregs. When PTL060 was combined with infusion of additional Tregs, these effects were further amplified. To test the benefits of thrombin inhibition in a transplant model, BALB/c hearts were transplanted into B6 mice with or without perfusion with PTL060 in combination with Tregs. Thrombin inhibition or Treg infusion alone led to small increments in allograft survival. However, the combined therapy led to modest graft prolongation by the same mechanisms as in renal IRI; graft survival was accompanied by increased numbers of Tregs and anti-inflammatory macrophages, and reduced expression of pro-inflammatory cytokines. While the grafts succumbed to rejection associated with the emergence of alloantibody, these data suggest that thrombin inhibition within the transplant vasculature enhances the efficacy of Treg infusion, a therapy that is currently entering the clinic to promote transplant tolerance.
Collapse
Affiliation(s)
- Qi Peng
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Anna Nowocin
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Kulachelvy Ratnasothy
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Richard A. Smith
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Lesley A. Smyth
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom,School of Health, Sport and Bioscience, University of East London, London, United Kingdom
| | - Robert I. Lechler
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Anthony Dorling
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom
| | - Giovanna Lombardi
- Centre for Nephrology, Urology and Transplantation, School of Immunology and Mucosal Biology, King’s College London, London, United Kingdom,*Correspondence: Giovanna Lombardi,
| |
Collapse
|
26
|
Polyzogopoulou E, Amoiridou P, Abraham TP, Ventoulis I. Acute liver injury in COVID-19 patients hospitalized in the intensive care unit: Narrative review. World J Gastroenterol 2022; 28:6662-6688. [PMID: 36620339 PMCID: PMC9813941 DOI: 10.3748/wjg.v28.i47.6662] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/14/2022] [Accepted: 12/05/2022] [Indexed: 12/19/2022] Open
Abstract
In recent years, humanity has been confronted with a global pandemic due to coronavirus disease 2019 (COVID-19), which has caused an unprecedented health and economic crisis worldwide. Apart from the respiratory symptoms, which are considered the principal manifestations of COVID-19, it has been recognized that COVID-19 constitutes a systemic inflammatory process affecting multiple organ systems. Across the spectrum of organ involvement in COVID-19, acute liver injury (ALI) has been gradually gaining increasing attention by the international scientific community. COVID-19 associated liver impairment can affect a considerable proportion of COVID-19 patients and seems to correlate with the severity of the disease course. Indeed, COVID-19 patients hospitalized in the intensive care unit (ICU) run a greater risk of developing ALI due to the severity of their clinical condition and in the context of multi-organ failure. The putative pathophysiological mechanisms of COVID-19 induced ALI in ICU patients remain poorly understood and appear to be multifactorial in nature. Several theories have been proposed to explain the occurrence of ALI in the ICU setting, such as hypoperfusion and ischemia due to hemodynamic instability, passive liver congestion as a result of congestive heart failure, ischemia-reperfusion injury, hypoxia due to respiratory failure, mechanical ventilation itself, sepsis and septic shock, cytokine storm, endotheliitis with concomitant coagulopathy, drug-induced liver injury, parenteral nutrition and direct cytopathic viral effect. It should be noted that no specific therapy for COVID-19 induced ALI exists. Therefore, the therapeutic approach lies in preventive measures and is exclusively supportive once ALI ensues. The aim of the current review is to scrutinize the existing evidence on COVID-19 associated ALI in ICU patients, explore its clinical implications, shed light on the underlying pathophysiological mechanisms and propose potential therapeutic approaches. Ongoing research on the particular scientific field will further elucidate the pathophysiology behind ALI and address unresolved issues, in the hope of mitigating the tremendous health consequences imposed by COVID-19 on ICU patients.
Collapse
Affiliation(s)
- Effie Polyzogopoulou
- Department of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens 12462, Greece
| | - Pinelopi Amoiridou
- Department of Intensive Care, AHEPA University Hospital, Thessaloniki 54621, Greece
| | - Theodore P Abraham
- Hypertrophic Cardiomyopathy Center of Excellence, University of California, San Francisco, CA 94117, United States
| | - Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, Ptolemaida 50200, Greece
| |
Collapse
|
27
|
Emara MM, Diab DG, Yassen AM, Abo-Zeid MA. Mannitol for prevention of acute kidney injury after liver transplantation: a randomized controlled trial. BMC Anesthesiol 2022; 22:393. [PMID: 36536282 PMCID: PMC9762035 DOI: 10.1186/s12871-022-01936-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is a common complication after liver transplantation, which is associated with increased morbidity and mortality. Therefore, this study investigated mannitol as an oxygen-free radical scavenger and its role in the prevention of early AKI after living donor liver transplantation (LDLT). METHODS A total of 84 adult patients who underwent LDLT were randomly assigned to two equal groups: the M group, where patients received 1 g/kg mannitol 20%, or the S group, where patients received an equal volume of saline. The primary outcome was the incidence of early AKI, defined as a 0.3 mg/dl increase in the serum creatinine 48 h postoperatively. Laboratory assessments of the graft and creatinine were recorded until 3 months after transplantation besides the post-reperfusion syndrome and the intraoperative hemodynamic measurements. RESULTS The AKI incidence was comparable between groups (relative risk ratio of 1.285, 95% CI 0.598-2.759, P = 0.518). Moreover, AKI stages and serum creatinine 3 months after transplantation, P = 0.23 and P = 0.25, respectively. The incidence of the post-reperfusion syndrome was comparable in both groups, 29/39 (74.4%) and 31/41 (75.6%) in M and S groups, respectively, P = 0.897. The intraoperative hemodynamic parameters showed no significant difference between groups using the area under the curve. CONCLUSION The current LDLT recipient sample was insufficient to demonstrate that pre-reperfusion 1 g/kg mannitol infusion would reduce the risk of early AKI or post-reperfusion syndrome. CLINICAL TRIAL REGISTRATION NUMBER Pan African Clinical Trials Registry (PACTR202203622900599); https://pactr.samrc.ac.za/TrialDisplay.aspx?TrialID=21511 .
Collapse
Affiliation(s)
- Moataz Maher Emara
- grid.10251.370000000103426662Department of Anesthesiology and Intensive Care and Pain Medicine, Mansoura University, Faculty of Medicine, Mansoura, Egypt ,grid.10251.370000000103426662Liver Transplantation program, Mansoura University, Gastrointestinal Surgery Center, Mansoura, Egypt
| | - Doaa Galal Diab
- grid.10251.370000000103426662Department of Anesthesiology and Intensive Care and Pain Medicine, Mansoura University, Faculty of Medicine, Mansoura, Egypt
| | - Amr Mohamed Yassen
- grid.10251.370000000103426662Department of Anesthesiology and Intensive Care and Pain Medicine, Mansoura University, Faculty of Medicine, Mansoura, Egypt ,grid.10251.370000000103426662Liver Transplantation program, Mansoura University, Gastrointestinal Surgery Center, Mansoura, Egypt
| | - Maha A. Abo-Zeid
- grid.10251.370000000103426662Department of Anesthesiology and Intensive Care and Pain Medicine, Mansoura University, Faculty of Medicine, Mansoura, Egypt
| |
Collapse
|
28
|
Pretzsch E, Nieß H, Khaled NB, Bösch F, Guba M, Werner J, Angele M, Chaudry IH. Molecular Mechanisms of Ischaemia-Reperfusion Injury and Regeneration in the Liver-Shock and Surgery-Associated Changes. Int J Mol Sci 2022; 23:12942. [PMID: 36361725 PMCID: PMC9657004 DOI: 10.3390/ijms232112942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/16/2022] [Accepted: 10/20/2022] [Indexed: 09/01/2023] Open
Abstract
Hepatic ischemia-reperfusion injury (IRI) represents a major challenge during liver surgery, liver preservation for transplantation, and can cause hemorrhagic shock with severe hypoxemia and trauma. The reduction of blood supply with a concomitant deficit in oxygen delivery initiates various molecular mechanisms involving the innate and adaptive immune response, alterations in gene transcription, induction of cell death programs, and changes in metabolic state and vascular function. Hepatic IRI is a major cause of morbidity and mortality, and is associated with an increased risk for tumor growth and recurrence after oncologic surgery for primary and secondary hepatobiliary malignancies. Therapeutic strategies to prevent or treat hepatic IRI have been investigated in animal models but, for the most part, have failed to provide a protective effect in a clinical setting. This review focuses on the molecular mechanisms underlying hepatic IRI and regeneration, as well as its clinical implications. A better understanding of this complex and highly dynamic process may allow for the development of innovative therapeutic approaches and optimize patient outcomes.
Collapse
Affiliation(s)
- Elise Pretzsch
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Hanno Nieß
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Najib Ben Khaled
- Department of Medicine II, University Hospital, LMU Munich, 81377 Munich, Germany
| | - Florian Bösch
- Department of General, Visceral and Pediatric Surgery, University Medical Center Goettingen, 37075 Goettingen, Germany
| | - Markus Guba
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Jens Werner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Martin Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, 81377 Munich, Germany
| | - Irshad H. Chaudry
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
29
|
Fodor M, Salcher S, Gottschling H, Mair A, Blumer M, Sopper S, Ebner S, Pircher A, Oberhuber R, Wolf D, Schneeberger S, Hautz T. The liver-resident immune cell repertoire - A boon or a bane during machine perfusion? Front Immunol 2022; 13:982018. [PMID: 36311746 PMCID: PMC9609784 DOI: 10.3389/fimmu.2022.982018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
The liver has been proposed as an important “immune organ” of the body, as it is critically involved in a variety of specific and unique immune tasks. It contains a huge resident immune cell repertoire, which determines the balance between tolerance and inflammation in the hepatic microenvironment. Liver-resident immune cells, populating the sinusoids and the space of Disse, include professional antigen-presenting cells, myeloid cells, as well as innate and adaptive lymphoid cell populations. Machine perfusion (MP) has emerged as an innovative technology to preserve organs ex vivo while testing for organ quality and function prior to transplantation. As for the liver, hypothermic and normothermic MP techniques have successfully been implemented in clinically routine, especially for the use of marginal donor livers. Although there is evidence that ischemia reperfusion injury-associated inflammation is reduced in machine-perfused livers, little is known whether MP impacts the quantity, activation state and function of the hepatic immune-cell repertoire, and how this affects the inflammatory milieu during MP. At this point, it remains even speculative if liver-resident immune cells primarily exert a pro-inflammatory and hence destructive effect on machine-perfused organs, or in part may be essential to induce liver regeneration and counteract liver damage. This review discusses the role of hepatic immune cell subtypes during inflammatory conditions and ischemia reperfusion injury in the context of liver transplantation. We further highlight the possible impact of MP on the modification of the immune cell repertoire and its potential for future applications and immune modulation of the liver.
Collapse
Affiliation(s)
- M. Fodor
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory, Medical University of Innsbruck, Innsbruck, Austria
- Department of Visceral, Transplant and Thoracic Surgery, Daniel Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - S. Salcher
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University Innsbruck (MUI), Innsbruck, Austria
| | - H. Gottschling
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory, Medical University of Innsbruck, Innsbruck, Austria
- Department of Visceral, Transplant and Thoracic Surgery, Daniel Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - A. Mair
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University Innsbruck (MUI), Innsbruck, Austria
| | - M. Blumer
- Department of Anatomy and Embryology, Medical University of Innsbruck, Innsbruck, Austria
| | - S. Sopper
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University Innsbruck (MUI), Innsbruck, Austria
| | - S. Ebner
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory, Medical University of Innsbruck, Innsbruck, Austria
- Department of Visceral, Transplant and Thoracic Surgery, Daniel Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - A. Pircher
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University Innsbruck (MUI), Innsbruck, Austria
| | - R. Oberhuber
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory, Medical University of Innsbruck, Innsbruck, Austria
- Department of Visceral, Transplant and Thoracic Surgery, Daniel Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - D. Wolf
- Department of Internal Medicine V, Hematology and Oncology, Comprehensive Cancer Center Innsbruck (CCCI), Medical University Innsbruck (MUI), Innsbruck, Austria
| | - S. Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory, Medical University of Innsbruck, Innsbruck, Austria
- Department of Visceral, Transplant and Thoracic Surgery, Daniel Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
| | - T. Hautz
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, organLife Laboratory, Medical University of Innsbruck, Innsbruck, Austria
- Department of Visceral, Transplant and Thoracic Surgery, Daniel Swarovski Research Laboratory, Medical University of Innsbruck, Innsbruck, Austria
- *Correspondence: T. Hautz,
| |
Collapse
|
30
|
Biomolecular Pathways of Cryoinjuries in Low-Temperature Storage for Mammalian Specimens. Bioengineering (Basel) 2022; 9:bioengineering9100545. [PMID: 36290513 PMCID: PMC9598205 DOI: 10.3390/bioengineering9100545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/22/2022] Open
Abstract
Low-temperature preservation could effectively extend in vitro storage of biological materials due to delayed or suspended cellular metabolism and decaying as illustrated by the Arrhenius model. It is widely used as an enabling technology for a variety of biomedical applications such as cell therapeutics, assisted reproductive technologies, organ transplantation, and mRNA medicine. Although the technology to minimize cryoinjuries of mammalian specimens during preservation has been advanced substantially over past decades, mammalian specimens still suffer cryoinjuries under low-temperature conditions. Particularly, the molecular mechanisms underlying cryoinjuries are still evasive, hindering further improvement and development of preservation technologies. In this paper, we systematically recapitulate the molecular cascades of cellular injuries induced by cryopreservation, including apoptosis, necroptosis, ischemia-reperfusion injury (IRI). Therefore, this study not only summarizes the impact of low-temperature preservations on preserved cells and organs on the molecular level, but also provides a molecular basis to reduce cryoinjuries for future exploration of biopreservation methods, materials, and devices.
Collapse
|
31
|
Nakatake R, Schulz M, Kalvelage C, Benstoem C, Tolba RH. Effects of iNOS in Hepatic Warm Ischaemia and Reperfusion Models in Mice and Rats: A Systematic Review and Meta-Analysis. Int J Mol Sci 2022; 23:ijms231911916. [PMID: 36233220 PMCID: PMC9569681 DOI: 10.3390/ijms231911916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 12/09/2022] Open
Abstract
Warm ischaemia is usually induced by the Pringle manoeuver (PM) during hepatectomy. Currently, there is no widely accepted standard protocol to minimise ischaemia-related injury, so reducing ischaemia-reperfusion damage is an active area of research. This systematic review and meta-analysis focused on inducible nitric oxide synthase (iNOS) as an early inflammatory response to hepatic ischaemia reperfusion injury (HIRI) in mouse- and rat-liver models. A systematic search of studies was performed within three databases. Studies meeting the inclusion criteria were subjected to qualitative and quantitative synthesis of results. We performed a meta-analysis of studies grouped by different HIRI models and ischaemia times. Additionally, we investigated a possible correlation of endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) regulation with iNOS expression. Of 124 included studies, 49 were eligible for the meta-analysis, revealing that iNOS was upregulated in almost all HIRIs. We were able to show an increase of iNOS regardless of ischemia or reperfusion time. Additionally, we found no direct associations of eNOS or NO with iNOS. A sex gap of primarily male experimental animals used was observed, leading to a higher risk of outcomes not being translatable to humans of all sexes.
Collapse
Affiliation(s)
- Richi Nakatake
- Institute for Laboratory Animal Science and Experimental Surgery, RWTH Aachen University, 52074 Aachen, Germany
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka 573-1010, Japan
| | - Mareike Schulz
- Institute for Laboratory Animal Science and Experimental Surgery, RWTH Aachen University, 52074 Aachen, Germany
| | - Christina Kalvelage
- Department of Intensive Care Medicine, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Carina Benstoem
- Department of Intensive Care Medicine, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - René H. Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, RWTH Aachen University, 52074 Aachen, Germany
- Correspondence:
| |
Collapse
|
32
|
Liu Y, Li S, Zhang G, Cai J. NOD1 induces pyroptotic cell death to aggravate liver ischemia-reperfusion injury in mice. MedComm (Beijing) 2022; 3:e170. [PMID: 36092860 PMCID: PMC9433815 DOI: 10.1002/mco2.170] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022] Open
Abstract
Nucleotide-binding oligomerization domain 1 (NOD1) can direct the release of inflammatory factors and influence autophagy and apoptosis in hepatic ischemia-reperfusion injury (IRI) in mice. As pyroptosis is involved in a number of inflammatory reactions, in this report, we investigated the potential for NOD1 to affect pyroptosis. We found that an increased expression of NOD1 during IRI was related to activation of the pyroptotic signaling pathway. With NOD1 activation, cleavage fragments of Caspase-1, gasdermin D (GSDMD), and interleukin (IL)-1β were all increased. Moreover, downregulation of NOD1 expression in AML12 cells exerted an opposite effect. Expression levels of cleaved-Caspase-1 and cleaved-GSDMD decreased after exposure to IRI and the number of cell membrane pores and apoptotic or pyroptotic cells decreased, along with the contents of inflammatory factors and lactate dehydrogenase in the supernatants of AML12 cells. Based on these findings, we conclude that NOD1 aggravates the pyroptotic cell death associated with hepatic ischemia-reperfusion injury in a mouse model via the Caspase-1/GSDMD axis. These findings help to alleviate pyroptotic cell death during liver transplantation or resection, providing new insights into novel protective therapies for liver IRI.
Collapse
Affiliation(s)
- Yu Liu
- Department of GastroenterologyTianjin First Central HospitalThe First Central Clinical CollegeTianjin Medical UniversityTianjinChina
- Department of internal medicineWangdingdi HospitalNankai DistrictTianjinChina
| | - Shipeng Li
- Department of General SurgeryJiaozuo People's HospitalXinxiang Medical UniversityJiaozuoChina
| | - Guoliang Zhang
- Department of GastroenterologyTianjin First Central HospitalThe First Central Clinical CollegeTianjin Medical UniversityTianjinChina
| | - Jinzhen Cai
- Department of organ transplantationOrgan Transplant CenterAffiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
33
|
Kaltenmeier C, Wang R, Popp B, Geller D, Tohme S, Yazdani HO. Role of Immuno-Inflammatory Signals in Liver Ischemia-Reperfusion Injury. Cells 2022; 11:cells11142222. [PMID: 35883665 PMCID: PMC9323912 DOI: 10.3390/cells11142222] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/09/2022] [Accepted: 07/13/2022] [Indexed: 02/01/2023] Open
Abstract
Ischemia reperfusion injury (IRI) is a major obstacle in liver resection and liver transplantation. The initial step of IRI is mediated through ischemia which promotes the production of reactive oxygen species in Kupffer cells. This furthermore promotes the activation of pro-inflammatory signaling cascades, including tumor necrosis factor-alpha, IL-6, interferon, inducible nitric oxide synthase, TLR9/nuclear-factor kappa B pathway, and the production of damage-associated molecular patterns (DAMPs), such as ATP, histone, high mobility group box 1 (HMGB1), urate, mitochondrial formyl peptides and S100 proteins. With ongoing cell death of hepatocytes during the ischemic phase, DAMPs are built up and released into the circulation upon reperfusion. This promotes a cytokines/chemokine storm that attracts neutrophils and other immune cells to the site of tissue injury. The effect of IRI is further aggravated by the release of cytokines and chemokines, such as epithelial neutrophil activating protein (CXCL5), KC (CXCL1) and MIP-2 (CXCL2), the complement proteins C3a and C5a, mitochondrial-derived formyl peptides, leukotriene B4 and neutrophil extracellular traps (NETs) from migrating neutrophils. These NETs can also activate platelets and form Neutrophil-platelet microthrombi to further worsen ischemia in the liver. In this review we aim to summarize the current knowledge of mediators that promote liver IRI, and we will discuss the role of neutrophils and neutrophil extracellular traps in mediating IRI.
Collapse
Affiliation(s)
- Christof Kaltenmeier
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (C.K.); (R.W.); (D.G.); (S.T.)
| | - Ronghua Wang
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (C.K.); (R.W.); (D.G.); (S.T.)
| | - Brandon Popp
- Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA;
| | - David Geller
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (C.K.); (R.W.); (D.G.); (S.T.)
| | - Samer Tohme
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (C.K.); (R.W.); (D.G.); (S.T.)
| | - Hamza O. Yazdani
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; (C.K.); (R.W.); (D.G.); (S.T.)
- Correspondence:
| |
Collapse
|
34
|
Li Y, Palmer A, Lupu L, Huber-Lang M. Inflammatory response to the ischaemia-reperfusion insult in the liver after major tissue trauma. Eur J Trauma Emerg Surg 2022; 48:4431-4444. [PMID: 35831749 DOI: 10.1007/s00068-022-02026-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/28/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Polytrauma is often accompanied by ischaemia-reperfusion injury to tissues and organs, and the resulting series of immune inflammatory reactions are a major cause of death in patients. The liver is one of the largest organs in the body, a characteristic that makes it the most vulnerable organ after multiple injuries. In addition, the liver is an important digestive organ that secretes a variety of inflammatory mediators involved in local as well as systemic immune inflammatory responses. Therefore, this review considers the main features of post-traumatic liver injury, focusing on the immuno-pathophysiological changes, the interactions between liver organs, and the principles of treatment deduced. METHODS We focus on the local as well as systemic immune response involving the liver after multiple injuries, with emphasis on the pathophysiological mechanisms. RESULTS An overview of the mechanisms underlying the pathophysiology of local as well as systemic immune responses involving the liver after multiple injuries, the latest research findings, and the current mainstream therapeutic approaches. CONCLUSION Cross-reactivity between various organs and cascade amplification effects are among the main causes of systemic immune inflammatory responses after multiple injuries. For the time being, the pathophysiological mechanisms underlying this interaction remain unclear. Future work will continue to focus on identifying potential signalling pathways as well as target genes and intervening at the right time points to prevent more severe immune inflammatory responses and promote better and faster recovery of the patient.
Collapse
Affiliation(s)
- Yang Li
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Annette Palmer
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Ludmila Lupu
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Markus Huber-Lang
- Institute for Clinical and Experimental Trauma Immunology (ITI), University Hospital Ulm, Helmholtzstr. 8/1, 89081, Ulm, Germany.
| |
Collapse
|
35
|
Ding M, Fang H, Zhang J, Shi J, Yu X, Wen P, Wang Z, Cao S, Zhang Y, Shi X, Zhang H, He Y, Yan B, Tang H, Guo D, Gao J, Liu Z, Zhang L, Zhang S, Zhang X, Guo W. E3 ubiquitin ligase ring finger protein 5 protects against hepatic ischemia reperfusion injury by mediating phosphoglycerate mutase family member 5 ubiquitination. Hepatology 2022; 76:94-111. [PMID: 34735734 PMCID: PMC9303746 DOI: 10.1002/hep.32226] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Hepatic ischemia-reperfusion (HIR) injury, a common clinical complication of liver transplantation and resection, affects patient prognosis. Ring finger protein 5 (RNF5) is an E3 ubiquitin ligase that plays important roles in endoplasmic reticulum stress, unfolded protein reactions, and inflammatory responses; however, its role in HIR is unclear. APPROACH AND RESULTS RNF5 expression was significantly down-regulated during HIR in mice and hepatocytes. Subsequently, RNF5 knockdown and overexpression of cell lines were subjected to hypoxia-reoxygenation challenge. Results showed that RNF5 knockdown significantly increased hepatocyte inflammation and apoptosis, whereas RNF5 overexpression had the opposite effect. Furthermore, hepatocyte-specific RNF5 knockout and transgenic mice were established and subjected to HIR, and RNF5 deficiency markedly aggravated liver damage and cell apoptosis and activated hepatic inflammatory responses, whereas hepatic RNF5 transgenic mice had the opposite effect compared with RNF5 knockout mice. Mechanistically, RNF5 interacted with phosphoglycerate mutase family member 5 (PGAM5) and mediated the degradation of PGAM5 through K48-linked ubiquitination, thereby inhibiting the activation of apoptosis-regulating kinase 1 (ASK1) and its downstream c-Jun N-terminal kinase (JNK)/p38. This eventually suppresses the inflammatory response and cell apoptosis in HIR. CONCLUSIONS We revealed that RNF5 protected against HIR through its interaction with PGAM5 to inhibit the activation of ASK1 and the downstream JNK/p38 signaling cascade. Our findings indicate that the RNF5-PGAM5 axis may be a promising therapeutic target for HIR.
Collapse
Affiliation(s)
- Ming‐Jie Ding
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Hao‐Ran Fang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Jia‐Kai Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Ji‐Hua Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Xiao Yu
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Pei‐Hao Wen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Zhi‐Hui Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Sheng‐Li Cao
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Yi Zhang
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xiao‐Yi Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Hua‐Peng Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Yu‐Ting He
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Bing Yan
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Hong‐Wei Tang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Dan‐Feng Guo
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Jie Gao
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | - Zhen Liu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Li Zhang
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Shui‐Jun Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| | | | - Wen‐Zhi Guo
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina,Henan Engineering Technology Research Center for Organ TransplantationZhengzhouChina,Zhengzhou Engineering Laboratory for Organ Transplantation Technique and ApplicationZhengzhouChina,Henan Research Centre for Organ TransplantationZhengzhouChina
| |
Collapse
|
36
|
Chen Q, Song Y, Yang N, Ai X, Pu L, Kong L. Aging deteriorated liver Ischemia and reperfusion injury by suppressing Tribble's proteins 1 mediated macrophage polarization. Bioengineered 2022; 13:14519-14533. [PMID: 36694470 PMCID: PMC9995131 DOI: 10.1080/21655979.2022.2090218] [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: 01/26/2023] Open
Abstract
Aggravated liver injury has been reported in aged ischemia/reperfusion-stressed livers; however, the mechanism of aged macrophage inflammatory regulation is not well understood. Here, we found that the adaptor protein TRIB1 plays a critical role in the differentiation of macrophages and the inflammatory response in the liver after ischemia/reperfusion injury. In the present study, we determined that aging promoted macrophage-mediated liver injury and that inflammation was mainly responsible for lower M2 polarization in liver transplantation-exposed humans post I/R. Young and aged mice were subjected to hepatic I/R modeling and showed that aging aggravated liver injury and suppressed macrophage TRIB1 protein expression and anti-inflammatory function in I/R-stressed livers. Restoration of TRIB1 is mediated by lentiviral infection-induced macrophage anti-inflammatory M2 polarization and alleviated hepatic I/R injury. Moreover, TRIB1 overexpression in macrophages facilitates M2 polarization and anti-inflammation by activating MEK1-ERK1/2 signaling under IL-4 stimulation. Taken together, our results demonstrated that aging promoted hepatic I/R injury by suppressing TRIB1-mediated MEK1-induced macrophage M2 polarization and anti-inflammatory function.
Collapse
Affiliation(s)
- Qi Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Department of General Surgery, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yating Song
- Department of Bariatric and Metabolic Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ningli Yang
- Department of Bariatric and Metabolic Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoming Ai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liyong Pu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lianbao Kong
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
37
|
Mobed A, Hasanzadeh M. Environmental protection based on the nanobiosensing of bacterial lipopolysaccharides (LPSs): material and method overview. RSC Adv 2022; 12:9704-9724. [PMID: 35424904 PMCID: PMC8959448 DOI: 10.1039/d1ra09393b] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
Lipopolysaccharide (LPS) or endotoxin control is critical for environmental and healthcare issues. LPSs are responsible for several infections, including septic and shock sepsis, and are found in water samples. Accurate and specific diagnosis of endotoxin is one of the most challenging issues in medical bacteriology. Enzyme-linked immunosorbent assay (ELISA), plating and culture-based methods, and Limulus amebocyte lysate (LAL) assay are the conventional techniques in quantifying LPS in research and medical laboratories. However, these methods have been restricted due to their disadvantages, such as low sensitivity and time-consuming and complicated procedures. Therefore, the development of new and advanced methods is demanding, particularly in the biological and medical fields. Biosensor technology is an innovative method that developed extensively in the past decade. Biosensors are classified based on the type of transducer and bioreceptor. So in this review, various types of biosensors, such as optical (fluorescence, SERS, FRET, and SPR), electrochemical, photoelectrochemical, and electrochemiluminescence, on the biosensing of LPs were investigated. Also, the critical role of advanced nanomaterials on the performance of the above-mentioned biosensors is discussed. In addition, the application of different labels on the efficient usage of biosensors for LPS is surveyed comprehensively. Also, various bio-elements (aptamer, DNA, miRNA, peptide, enzyme, antibody, etc.) on the structure of the LPS biosensor are investigated. Finally, bio-analytical parameters that affect the performance of LPS biosensors are surveyed.
Collapse
Affiliation(s)
- Ahmad Mobed
- Aging Research Institute, Faculty of Medicine, Tabriz University of Medical Sciences Iran
- Physical Medicine and Rehabilitation Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences Tabriz 51664 Iran
| | - Mohammad Hasanzadeh
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences Tabriz 51664 Iran
- Nutrition Research Center, Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
38
|
Shaping of Hepatic Ischemia/Reperfusion Events: The Crucial Role of Mitochondria. Cells 2022; 11:cells11040688. [PMID: 35203337 PMCID: PMC8870414 DOI: 10.3390/cells11040688] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 12/10/2022] Open
Abstract
Hepatic ischemia reperfusion injury (HIRI) is a major hurdle in many clinical scenarios, including liver resection and transplantation. Various studies and countless surgical events have led to the observation of a strong correlation between HIRI induced by liver transplantation and early allograft-dysfunction development. The detrimental impact of HIRI has driven the pursuit of new ways to alleviate its adverse effects. At the core of HIRI lies mitochondrial dysfunction. Various studies, from both animal models and in clinical settings, have clearly shown that mitochondrial function is severely hampered by HIRI and that its preservation or restoration is a key indicator of successful organ recovery. Several strategies have been thus implemented throughout the years, targeting mitochondrial function. This work briefly discusses some the most utilized approaches, ranging from surgical practices to pharmacological interventions and highlights how novel strategies can be investigated and implemented by intricately discussing the way mitochondrial function is affected by HIRI.
Collapse
|
39
|
Othman MA, Mubarak HA, Sayed MM. Ameliorative role of alpha-lipoic acid in renal cortical structural damage, induced by limb ischemia-reperfusion injury in the rat. Ultrastruct Pathol 2022; 46:110-121. [PMID: 35135431 DOI: 10.1080/01913123.2022.2035875] [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: 10/19/2022]
Abstract
Ischemia-reperfusion injury is related to kidney dysfunction due to bilateral lower limb ischemia. This kidney injury may lead to acute kidney failure and mortality. Alpha-Lipoic Acid, a known antioxidant, can ameliorate kidney dysfunction and histopathology related to several etiologies. Ischemia was performed in adult male rats by bilateral femoral artery occlusion, then ischemia-reperfusion was done for 1 day and 7 days. Lipoic acid was administered to rats that had undergone ischemia-reperfusion for 7 days. The renal cortices of the kidneys of the tested groups were processed for light and electron microscopic examination. Immunohistochemical evaluation was performed using the following markers: cleaved caspase 3, inducible nitric oxide synthase, and tumor necrosis factor-alpha. There was damage to the renal cortical tubules and degeneration of podocytes and thickening of the glomerular basement membrane. Additionally, there was an increase in apoptosis and the inflammatory markers' immunoreactivity. Administration of alpha-lipoic acid resulted in improvement of the structural and immunohistochemical changes of the renal cortex. This may suggest a therapeutic rule of it and promising application for variable kidney injuries.
Collapse
Affiliation(s)
- Manal A Othman
- Department of Anatomy, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain.,Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assuit, Egypt
| | - Heba A Mubarak
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assuit, Egypt
| | - Manal M Sayed
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assuit, Egypt
| |
Collapse
|
40
|
Aliyeva D, Amanvermez R, Karabulut K, Gün S. The effects of silymarin plus glutathione on the prevention of liver ischemia-reperfusion injury. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e20561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
| | | | | | - Seda Gün
- Ondokuz Mayıs University, Turkey
| |
Collapse
|
41
|
Torres RR, Tannuri ACA, Serafini S, Belon A, Gonçalves JO, Loreto CD, Tannuri U. Does Arterialization of Portal Vein Have Any Effects in Large-for-Size Liver Transplantation? Hemodynamic, Histological, and Biomolecular Experimental Studies. J INVEST SURG 2021; 35:1197-1207. [PMID: 34965813 DOI: 10.1080/08941939.2021.2021333] [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: 10/19/2022]
Abstract
BACKGROUND In pediatric liver transplantation, the optimal size of the transplanted liver ranges between 0.8% and 4.0% of the recipient's weight. Sometimes, the graft weight exceeds this upper limit, characterizing the large-for-size condition potentially associated with reduced blood flow and worsening of ischemia-reperfusion injury. Therefore, it would be beneficial to increase the portal flow through arterialization of the portal vein. Materials and methods: Fifteen pigs underwent large-for-size liver transplants. They were divided into two groups: control (CTRL 6 animals - conventional technique) and arterialization - a shunt was established between the portal vein and the splenic artery (ART 9 animals). Hemodynamic, biochemical, histological, and molecular variables were compared. Results: Arterialization resulted in a significant increase in portal vein pressure but no changes in other hemodynamic variables, as shown in the analysis of variance. It was observed lower ALT values (p = 0.007), with no differences regarding the values of blood pH and lactate (p = 0.54 and p = 0.699 respectively) or histological variables (edema, steatosis, inflammation, necrosis, and IRI - p = 1.0, p = 0.943, p = 0.174, p = 0.832, p = 0.662, respectively). The molecular studies showed significantly increased expression of IL6 after 3 hours of reperfusion (p = 0.048) and decreased expression of ICAM immediately after reperfusion (p = 0.03). The regression analysis suggested a positive influence of portal flow and pressure on biochemical parameters. Conclusion: Arterialization of the portal vein showed no histological, biochemical, or molecular benefits in large-for-size transplantation.
Collapse
Affiliation(s)
- Rafael Rodrigues Torres
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Ana Cristina Aoun Tannuri
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Suellen Serafini
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Alessandro Belon
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Josiane Oliveira Gonçalves
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Celso di Loreto
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Uenis Tannuri
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| |
Collapse
|
42
|
de Souza JR, Yokoyama AP, Magnus MM, Boin I, de Ataide EC, Munhoz DC, Pereira FB, Luzo A, Orsi FA. Association of acidosis with coagulopathy and transfusion requirements in liver transplantation. J Thromb Thrombolysis 2021; 53:887-897. [PMID: 34800258 DOI: 10.1007/s11239-021-02609-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/09/2021] [Indexed: 11/26/2022]
Abstract
The relationship between acidosis and coagulopathy has long been described in vitro and in trauma patients, but not yet in orthotopic liver transplantation (OLT). The association of metabolic acidosis with coagulopathy and with transfusion requirements was evaluated in patients submitted to OLT. Changes in acid-base and coagulation parameters were analyzed by repeated measures. Regression analyses [adjusted for sex, age, model for end stage liver disease (MELD) score, and baseline values of hemoglobin, fibrinogen, international normalized ratio, platelets] determined the association of acid-base parameters with coagulation markers and transfusion requirement. We included 95 patients, 66% were male, 49.5% of the patients had hepatocellular carcinoma and the mean MELD score was 20.4 (SD 8.9). The values of all the coagulation and acid-base parameters significantly changed during OLT, particularly in the reperfusion phase. After adjustments for baseline parameters, the decrease in pH and base excess (BE) values were associated with a decrease in fibrinogen levels (mean decrease of fibrinogen level = 14.88 mg/dL per 0.1 unit reduction of pH values and 3.6 mg/dL per 1 mmol/L reduction of BE levels) and an increase in red blood cells transfusion (2.16 units of RBC per 0.1 unit reduction of pH and 0.38 units of RBC per 1 mmol/L reduction of BE levels). Among multiple factors potentially associated with adverse outcomes, decreasing pH levels were independently associated with the length of hospitalization but not with in-hospital mortality. Metabolic acidosis is independently associated with decreased fibrinogen levels and increased intraoperative transfusion requirement during OLT. Awareness of that association may improve treatment strategies to reduce intraoperative bleeding risk in OLT.
Collapse
Affiliation(s)
- Júlia Ruete de Souza
- Faculty of Medicine, Pontifical Catholic University of Campinas, Campinas, Brazil
| | - Ana Paula Yokoyama
- School of Medical Sciences, University of Campinas, Campinas, Brazil
- Hemotherapy and Cell Therapy Department, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Ilka Boin
- Department of Surgery, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Derli Conceição Munhoz
- Department of Anestiology, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | | | - Angela Luzo
- Hematology and Hemotherapy Center, University of Campinas, Campinas, Brazil
| | - Fernanda Andrade Orsi
- Department of Pathology, School of Medical Sciences, University of Campinas, Campinas R. Tessália Vieira de Camargo, 126 Cidade Universitária, Campinas, 13083-887, Brazil.
| |
Collapse
|
43
|
Kollaras V, Valsami G, Lambropoulou M, Konstandi O, Kostomistsopoulos N, Pikoulis E, Simopoulos C, Tsaroucha A. Effect of silibinin on the expression of MMP2, MMP3, MMP9 and TIMP2 in kidney and lung after hepatic ischemia/reperfusion injury in an experimental rat model. Acta Cir Bras 2021; 36:e360904. [PMID: 34755764 PMCID: PMC8580512 DOI: 10.1590/acb360904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/12/2021] [Indexed: 11/22/2022] Open
Abstract
PURPOSE The protective effect of silibinin on kidney and lung parenchyma during hepatic ischemia/reperfusion injury (IRI) is explored. METHODS Sixty-three Wistar rats were separated into three groups: sham; control (45 min IRI); and silibinin (200 μL silibinin administration after 45 min of ischemia and before reperfusion). Immunohistochemistry and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to evaluate the expression levels of MMP2, MMP3, MMP9, and TIMP2 on kidney and lung. RESULTS Comparing sham vs. control groups, confirmed that hepatic IRI increased both renal and lung MMP2, MMP3, MMP9 and TIMP2 expressions starting at 180 min (p<0.001). Comparison of the control vs. silibinin groups showed a statistically significant decrease in the expression levels of MMP2, MMP3, and MMP9 and increase of TIMP2 in kidney and lung parenchyma. The starting point of this decrease was at 120 min after reperfusion, both for kidney and lung parameters, and it was statistically significant at 240 min (p<0.001) for kidney, while silibinin showed a peak of lung protection at 180 min after hepatic reperfusion (p<0.001). CONCLUSIONS Hepatic IRI causes distant kidney and lung damage, while a statistically significant protective action, both on kidney and lung parenchyma, is conveyed by the intravenous administration of silibinin.
Collapse
|
44
|
Ji H, Li H, Zhang H, Cheng Z. Role of microRNA‑218‑5p in sevoflurane‑induced protective effects in hepatic ischemia/reperfusion injury mice by regulating GAB2/PI3K/AKT pathway. Mol Med Rep 2021; 25:1. [PMID: 34726254 PMCID: PMC8600399 DOI: 10.3892/mmr.2021.12517] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 05/20/2021] [Indexed: 12/11/2022] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury (HIRI) often occurs following tissue resection, hemorrhagic shock or transplantation surgery. Previous investigations showed that sevoflurane (Sevo), an inhalation anesthetic, had protective properties against different organ damage in animal models including HIRI. This study is aimed to investigate the underlying mechanisms involved in the protective effects of Sevo on HIRI. The present study results showed that treatment with Sevo improved histologic damage, inflammatory response, oxidative stress and apoptosis after hepatic I/R, indicating the protective role of Sevo against liver I/R injury. Importantly, in order to determine the molecular mechanism of Sevo in HIRI, the focus of the study was on microRNA (miR) regulation. By retrieving the microarray data in the Gene Expression Omnibus dataset (GSE72315), miR-218-5p was found to be significantly downregulated by Sevo. Moreover, miR-218-5p overexpression using agomiR-218-5p reversed the protective roles of Sevo against HIRI. Furthermore, GAB2, a positive regulator of PI3K/AKT signaling pathway, was found as a target gene of miR-218-5p. It was also found that the Sevo-mediated protective effects may be dependent on the activation of GAB2/PI3K/AKT. Collectively, these data revealed that Sevo alleviated HIRI in mice by restraining apoptosis, relieving oxidative stress and inflammatory response through the miR-218-5p/GAB2/PI3K/AKT pathway, which helps in understanding the novel mechanism of the hepatic-protection of Sevo.
Collapse
Affiliation(s)
- Hui Ji
- Department of Anesthesiology, Xinhua Hospital, Chongming Branch, Shanghai 202150, P.R. China
| | - Hui Li
- Department of Anesthesiology, Xinhua Hospital, Chongming Branch, Shanghai 202150, P.R. China
| | - Haixia Zhang
- Department of Anesthesiology, Xinhua Hospital, Chongming Branch, Shanghai 202150, P.R. China
| | - Zhijun Cheng
- Department of Anesthesiology, Xinhua Hospital, Chongming Branch, Shanghai 202150, P.R. China
| |
Collapse
|
45
|
Song J, He Z, Yang M, Yu T, Wang X, Liu B, Li J. HepaticIschemia/Reperfusion Injuryinvolves functional tryptase/PAR-2 signaling in liver sinusoidal endothelial cell population. Int Immunopharmacol 2021; 100:108052. [PMID: 34454294 DOI: 10.1016/j.intimp.2021.108052] [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: 04/04/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
Mast cells (MCs) are tissue-resident effector cells that could be the earliest responder to release a unique, stimulus-specific set of mediators in hepatic ischemia-reperfusion (IR) injury However, how MCs function in the hepatic IR has remained a formidable challenge due to the substantial redundancy and functional diverse of these mediators. Tryptase is the main protease for degranulation of MCs and its receptor-protease-activated receptor 2 (PAR-2) is widely expressed in endothelial cells. It is unclear whether and how tryptase/PAR-2 axis participates in hepatic IR. We employed an experimental warm 70% liver IR model in mice and found that tryptase was accumulated in the circulation during hepatic IR and positively correlated with liver injury. Tryptase inhibition by protamine can significantly down-regulate the expression of adhesion molecules and reduce neutrophil infiltration within the liver. The level of inflammatory factors and chemokines were also consistent with the pathological change of the liver. In addition, the treatment with exogeneous tryptase in MC-deficient mice can induce the damage observed in wild type mice in the context of liver IR. In vitro, neutrophil infiltration and inflammatory factor secretion were regulated by Tryptase/PAR-2, involving the adhesion molecule expression to regulate neutrophil adhesion dependent on NF-κB pathway. Conclusion: tryptase/PAR-2 participates in liver injury through the activation of LSECs in the early phase of liver IR.
Collapse
Affiliation(s)
- Jian Song
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China; Department of General Surgery, Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Zhigang He
- Department of Plastic and Constructive Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Muqing Yang
- Department of General Surgery, Shanghai Tenth People's Hospital School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Tianyu Yu
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China
| | - Xiaodong Wang
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China
| | - Bin Liu
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China
| | - Jiyu Li
- Geriatric Cancer Center, Huadong Hospital, Fudan University, West 221 Yan-an Road, Shanghai 200040, China.
| |
Collapse
|
46
|
Methods of Attenuating Ischemia-Reperfusion Injury in Liver Transplantation for Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:ijms22158229. [PMID: 34360995 PMCID: PMC8347959 DOI: 10.3390/ijms22158229] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/18/2021] [Accepted: 07/29/2021] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most frequent indications for liver transplantation. However, the transplantation is ultimately associated with the occurrence of ischemia-reperfusion injury (IRI). It affects not only the function of the graft but also significantly worsens the oncological results. Various methods have been used so far to manage IRI. These include the non-invasive approach (pharmacotherapy) and more advanced options encompassing various types of liver conditioning and machine perfusion. Strategies aimed at shortening ischemic times and better organ allocation pathways are still under development as well. This article presents the mechanisms responsible for IRI, its impact on treatment outcomes, and strategies to mitigate it. An extensive review of the relevant literature using MEDLINE (PubMed) and Scopus databases until September 2020 was conducted. Only full-text articles written in English were included. The following search terms were used: “ischemia reperfusion injury”, “liver transplantation”, “hepatocellular carcinoma”, “preconditioning”, “machine perfusion”.
Collapse
|
47
|
Abdulredha A, Abosaooda M, Al-Amran F, Hadi NR. Berberine Protests the Heart from Ischemic Reperfusion Injury via Interference with Oxidative and Inflammatory Pathways. Med Arch 2021; 75:174-179. [PMID: 34483445 PMCID: PMC8385727 DOI: 10.5455/medarh.2021.75.174-179] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/10/2021] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Ischemia and reperfusion (I/R) is a pathological condition characterized by an initial restriction of blood supply to an organ followed by the subsequent restoration of perfusion and concomitant reoxygenation. OBJECTIVE The aim of the study is to assess the possible cardioprotective potential effect of berberine in myocardial ischemia reperfusion injury induced by ligation of coronary artery in a male rat model. METHODS Total amount of 28 adult male albino rats were randomized into 4 equal groups: 1) Sham group, rats underwent the same anesthetic and surgical procedure as the control group except for LAD ligation; 2), Active control group, rats subjected to regional ischemia for 30 min by ligation of LAD coronary artery and reperfusion for 2 hours, 3), Control vehicle group, rats received dimethyl sulphoxide (DMSO) (vehicle of berberine) via IP route and subjected to ischemia for 30 minutes before ligation of LAD coronary artery & reperfusion for 2 hr; 4), Berberine treated group, rats pretreated with berberine10 mg/kg via IP injection 30minutes before ligation of LAD coronary artery & then subjected to reperfusion for 2 hr. RESULTS In the control group, as compared with sham, tissue TNF-α, IL-6, IL-10, caspase-3 and BAX, plasma cTn-T and serum MDA significantly increased (P<0.05), while serum GSH significantly decreased (P<0.05). The histopathological control group showed a significant cardiac injury (P<0.05) compared with the sham group. Berberine significantly counteracted (P<0.05) the increase of TNF-α, IL-6, caspase-3 and BAX and counteracted the increase in plasma cTn-T and serum MDA. Berberine produces a significant elevation (P<0.05) in cardiac IL-10 and serum GSH with a significant reduction in (P<0.05) cardiac injury. CONCLUSION Berberine attenuates myocardial I/R injury in male rats via interfering with inflammatory reactions and apoptosis which were induced by I/R injury.
Collapse
Affiliation(s)
- Abeer Abdulredha
- Al-Sadar Teaching Medical City, Al-Najaf Open Heart and Interventional Cardiac Center. Najaf, Iraq
| | | | - Fadhil Al-Amran
- Department of Thoracic Surgery, College of Medicine, Kufa University, Kufa, Iraq
| | - Najah R. Hadi
- Al-Sadar Teaching Medical City, Al-Najaf Open Heart and Interventional Cardiac Center. Najaf, Iraq
| |
Collapse
|
48
|
Wang L, Qu P, Yin W, Sun J. Lnc-NEAT1 induces cell apoptosis and inflammation but inhibits proliferation in a cellular model of hepatic ischemia/reperfusion injury. J Int Med Res 2021; 49:300060519887251. [PMID: 33682508 PMCID: PMC7944539 DOI: 10.1177/0300060519887251] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE We aimed to investigate the effect of long non-coding RNA nuclear-enriched abundant transcript 1 (lnc-NEAT1) on regulating hepatocyte proliferation, apoptosis, and inflammation during hepatic ischemia/reperfusion (I/R) injury. METHODS Human liver cells (HL-7702) were cultured under glucose-free and oxygen-free conditions to construct the I/R injury model. Expression of lnc-NEAT1 was detected in this model and in normal cells. Plasmids of control overexpression [NC(+)], lnc-NEAT1 overexpression [NEAT1(+)], control short hairpin (sh)RNA [NC(-)], and lnc-NEAT1 shRNA [NEAT1(-)] were transfected into HL-7702 cells and subsequently subjected to I/R treatment. Cell proliferation, apoptosis, apoptosis-related proteins, and inflammatory cytokines were assessed. RESULTS Lnc-NEAT1 expression was elevated in the I/R group compared with the normal group. Cell proliferation was decreased in the NEAT1(+) group compared with the NC(+) group but increased in NEAT1(-) compared with NC(-). The apoptosis rate increased in the NEAT1(+) group compared with the NC(+) group but decreased in NEAT1(-) compared with NC(-). Western blot assay (detection of apoptosis-related proteins) showed similar results. Expression of interleukin-1β, interleukin-6, and tumor necrosis factor-α increased in the NEAT1(+) group compared with NC(+) but decreased in NEAT1(-) compared with NC(-). CONCLUSION Lnc-NEAT1 is overexpressed, induces cell apoptosis and inflammation, and inhibits proliferation during hepatic I/R injury.
Collapse
Affiliation(s)
- Liu Wang
- General Department 2, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Houhu General Department, Wuhan, China
| | - Pan Qu
- General Department 3, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanling Yin
- Department of Geratology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Sun
- General Department 2, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Houhu General Department, Wuhan, China
| |
Collapse
|
49
|
Tang D, Fu G, Li W, Sun P, Loughran PA, Deng M, Scott MJ, Billiar TR. Maresin 1 protects the liver against ischemia/reperfusion injury via the ALXR/Akt signaling pathway. Mol Med 2021; 27:18. [PMID: 33632134 PMCID: PMC7905895 DOI: 10.1186/s10020-021-00280-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/27/2021] [Accepted: 02/10/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Hepatic ischemia/reperfusion (I/R) injury can be a major complication following liver surgery contributing to post-operative liver dysfunction. Maresin 1 (MaR1), a pro-resolving lipid mediator, has been shown to suppress I/R injury. However, the mechanisms that account for the protective effects of MaR1 in I/R injury remain unknown. METHODS WT (C57BL/6J) mice were subjected to partial hepatic warm ischemia for 60mins followed by reperfusion. Mice were treated with MaR1 (5-20 ng/mouse), Boc2 (Lipoxin A4 receptor antagonist), LY294002 (Akt inhibitor) or corresponding controls just prior to liver I/R or at the beginning of reperfusion. Blood and liver samples were collected at 6 h post-reperfusion. Serum aminotransferase, histopathologic changes, inflammatory cytokines, and oxidative stress were analyzed to evaluate liver injury. Signaling pathways were also investigated in vitro using primary mouse hepatocyte (HC) cultures to identify underlying mechanisms for MaR1 in liver I/R injury. RESULTS MaR1 treatment significantly reduced ALT and AST levels, diminished necrotic areas, suppressed inflammatory responses, attenuated oxidative stress and decreased hepatocyte apoptosis in liver after I/R. Akt signaling was significantly increased in the MaR1-treated liver I/R group compared with controls. The protective effect of MaR1 was abrogated by pretreatment with Boc2, which together with MaR1-induced Akt activation. MaR1-mediated liver protection was reversed by inhibition of Akt. CONCLUSIONS MaR1 protects the liver against hepatic I/R injury via an ALXR/Akt signaling pathway. MaR1 may represent a novel therapeutic agent to mitigate the detrimental effects of I/R-induced liver injury.
Collapse
Affiliation(s)
- Da Tang
- Department of General Surgery, The Third Xiangya Hospital, Central South University, 410000, Changsha, People's Republic of China
| | - Guang Fu
- Department of General Surgery, The Third Xiangya Hospital, Central South University, 410000, Changsha, People's Republic of China
| | - Wenbo Li
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital, Central South University, 410000, Changsha, People's Republic of China
| | - Ping Sun
- Department of Hepatobiliary Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | - Meihong Deng
- Department of Surgery, Ohio State University Medical School, OH, Columbus, USA
| | - Melanie J Scott
- Department of Surgery, University of Pittsburgh, PA, 15213, Pittsburgh, USA
- Pittsburgh Trauma Research Center, University of Pittsburgh, 15213, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, 15213, Pittsburgh, PA, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, PA, 15213, Pittsburgh, USA.
- Pittsburgh Trauma Research Center, University of Pittsburgh, 15213, Pittsburgh, PA, USA.
- Pittsburgh Liver Research Center, University of Pittsburgh, 15213, Pittsburgh, PA, USA.
| |
Collapse
|
50
|
Complement 5 Inhibition Ameliorates Hepatic Ischemia/reperfusion Injury in Mice, Dominantly via the C5a-mediated Cascade. Transplantation 2021; 104:2065-2077. [PMID: 32384381 DOI: 10.1097/tp.0000000000003302] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Hepatic ischemia/reperfusion injury (IRI) is a serious complication in liver surgeries, including transplantation. Complement activation seems to be closely involved in hepatic IRI; however, no complement-targeted intervention has been clinically applied. We investigated the therapeutic potential of Complement 5 (C5)-targeted regulation in hepatic IRI. METHODS C5-knockout (B10D2/oSn) and their corresponding wild-type mice (WT, B10D2/nSn) were exposed to 90-minute partial (70%) hepatic ischemia/reperfusion with either anti-mouse-C5 monoclonal antibody (BB5.1) or corresponding control immunoglobulin administration 30 minutes before ischemia. C5a receptor 1 antagonist was also given to WT to identify which cascade, C5a or C5b-9, is dominant. RESULTS C5-knockout and anti-C5-Ab administration to WT both significantly reduced serum transaminase release and histopathological damages from 2 hours after reperfusion. This improvement was characterized by significantly reduced CD41+ platelet aggregation, maintained F4/80+ cells, and decreased high-mobility group box 1 release. After 6 hours of reperfusion, the infiltration of CD11+ and Ly6-G+ cells, cytokine/chemokine expression, single-stranded DNA+ cells, and cleaved caspase-3 expression were all significantly alleviated by anti-C5-Ab. C5a receptor 1 antagonist was as effective as anti-C5-Ab for reducing transaminases. CONCLUSIONS Anti-C5 antibody significantly ameliorated hepatic IRI, predominantly via the C5a-mediated cascade, not only by inhibiting platelet aggregation during the early phase but also by attenuating the activation of infiltrating macrophages/neutrophils and hepatocyte apoptosis in the late phase of reperfusion. Given its efficacy, clinical availability, and controllability, C5-targeted intervention may provide a novel therapeutic strategy against hepatic IRI.
Collapse
|