1
|
Hao J, Hu R, Zhao J, Li Y, Li Q, Zhang X. Metabolomics combined with network pharmacology reveals the protective effect of astragaloside IV on alcoholic liver disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156032. [PMID: 39270570 DOI: 10.1016/j.phymed.2024.156032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/13/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Alcoholic liver disease (ALD) is a significant contributor to liver damage. However, the clinical options for the treatment of ALD are limited. Astragaloside IV (AST-IV) is a saponin isolated from Astragalus membranaceus (AM). This study aimed to explore the underlying mechanisms of action of AST-IV in ALD by integrating metabolomics and network pharmacology. METHODS Sprague-Dawley (SD) rats were used to establish a rat model of ALD. AST-IV and polyene phosphatidyl choline (PPC; a positive control drug) were administered to rats with ALD for 4 weeks. We measured the body weight, liver index, ALT, AST, TC, TG, inflammatory markers (IL-1β, IL-6, and TNF-α), and oxidative stress markers (SOD, MDA) and used H&E and ORO staining to evaluate the hepatoprotective effect of both AST-IV and PPC on ALD. Subsequently, we performed untargeted metabolomics to predict the influence of AST-IV on lipid metabolism in rats with ALD. We then used a network pharmacology approach to identify the core targets through which AST-IV corrected lipid metabolism disorders and validated these targets through molecular docking, qRT-PCR and western blot analyses. Finally, we calculated the relationships between ALD-related biochemical markers, differential liver metabolites, and core targets using Spearman's correlation analysis. RESULTS AST-IV improved pathological damage and reduced lipid accumulation in the hepatocytes of rats with ALD. Furthermore, AST-IV inhibited oxidative stress and inflammatory responses in rats with ALD. The metabolomic results showed that AST-IV corrected hepatic lipid metabolism disorders by targeting linoleic acid, necrosis, sphingolipid, and glycerophospholipid metabolism. The Network pharmacology analysis revealed that the core targets of AST-IV exerting the above effects were p-RIPK3, p-MLKL, CYP1A2, CYP2C19, PPARα, PCSK9. Spearman's correlation analysis showed a strong correlation between ALD-related serum biochemical indices, core targets, and liver differential metabolites. CONCLUSION AST-IV corrects the metabolic disorders of linoleic acid, sphingolipid, and glycerophospholipid, and alleviates necrosis in rats with ALD through the core targets p-RIPK3, p-MLKL, CYP1A2, CYP2C19, PPARα, and PCSK9. This study is the first to reveal the mechanism of ALD protection through AST-IV from the perspective of metabolomics and network pharmacology. Therefore, a novel target has been identified to exert protection against ALD. This study provides a reference for ALD treatment.
Collapse
Affiliation(s)
- Jinfang Hao
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Jinzhong 030619, China
| | - Ruixian Hu
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China; Department of Nutrition and Food Hygiene, School of Public Health, Shanxi Medical University, Taiyuan 030001, China
| | - Jianming Zhao
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China
| | - Yuanhong Li
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China
| | - Qingshan Li
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, Shanxi University of Chinese Medicine, Jinzhong 030619, China.
| | - Xiaoyan Zhang
- School of Pharmaceutical Science, Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
2
|
Cheng Z, Yang L, Chu H. The role of gut microbiota, exosomes, and their interaction in the pathogenesis of ALD. J Adv Res 2024:S2090-1232(24)00268-6. [PMID: 38969094 DOI: 10.1016/j.jare.2024.07.002] [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: 05/06/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND The liver disorders caused by alcohol abuse are termed alcoholic-related liver disease (ALD), including alcoholic steatosis, alcoholic steatohepatitis, alcoholic hepatitis, and alcoholic cirrhosis, posing a significant threat to human health. Currently, ALD pathogenesis has not been completely clarified, which is likely to be related to the direct damage caused by alcohol and its metabolic products, oxidative stress, gut dysbiosis, and exosomes. AIMS The existing studies suggest that both the gut microbiota and exosomes contribute to the development of ALD. Moreover, there exists an interaction between the gut microbiota and exosomes. We discuss whether this interaction plays a role in the pathogenesis of ALD and whether it can be a potential therapeutic target for ALD treatment. KEY SCIENTIFIC CONCEPTS OF REVIEW Chronic alcohol intake alters the diversity and composition of gut microbiota, which greatly contributes to ALD's progression. Some approaches targeting the gut microbiota, including probiotics, fecal microbiota transplantation, and phage therapy, have been confirmed to effectively ameliorate ALD in many animal experiments and/or several clinical trials. In ALD, the levels of exosomes and the expression profile of microRNA have also changed, which affects the pathogenesis of ALD. Moreover, there is an interplay between exosomes and the gut microbiota, which also putatively acts as a pathogenic factor of ALD.
Collapse
Affiliation(s)
- Zilu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| |
Collapse
|
3
|
Duffus BLM, Haggerty DL, Doud EH, Mosley AL, Yamamoto BK, Atwood BK. The impact of abstinence from chronic alcohol consumption on the mouse striatal proteome: sex and subregion-specific differences. Front Pharmacol 2024; 15:1405446. [PMID: 38887549 PMCID: PMC11180734 DOI: 10.3389/fphar.2024.1405446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/13/2024] [Indexed: 06/20/2024] Open
Abstract
Alcohol misuse is the third leading preventable cause of death in the world. The World Health Organization currently estimates that 1 in 20 deaths are directly alcohol related. One of the ways in which consuming excessive levels of alcohol can both directly and indirectly affect human mortality and morbidity, is through chronic inflammation. Recently, studies have suggested a link between increased alcohol use and the incidence of neuroinflammatory-related diseases. However, the mechanism in which alcohol potentially influences neuroinflammatory processes is still being uncovered. We implemented an unbiased proteomics exploration of alcohol-induced changes in the striatum, with a specific emphasis on proteins related to inflammation. The striatum is a brain region that is critically involved with the progression of alcohol use disorder. Using mass spectrometry following voluntary alcohol self-administration in mice, we show that distinct protein abundances and signaling pathways in different subregions of the striatum are disrupted by chronic exposure to alcohol compared to water drinking control mice. Further, in mice that were allowed to experience abstinence from alcohol compared to mice that were non-abstinent, the overall proteome and signaling pathways showed additional differences, suggesting that the responses evoked by chronic alcohol exposure are dependent on alcohol use history. To our surprise we did not find that chronic alcohol drinking or abstinence altered protein abundance or pathways associated with inflammation, but rather affected proteins and pathways associated with neurodegeneration and metabolic, cellular organization, protein translation, and molecular transport processes. These outcomes suggest that in this drinking model, alcohol-induced neuroinflammation in the striatum is not a primary outcome controlling altered neurobehavioral function, but these changes are rather mediated by altered striatal neuronal structure and cellular health.
Collapse
Affiliation(s)
- Brittnie-lee M. Duffus
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - David L. Haggerty
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Emma H. Doud
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Amber L. Mosley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Bryan K. Yamamoto
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States
| | | |
Collapse
|
4
|
Cheng SY, Jiang L, Wang Y, Cai W. Emerging role of regulated cell death in intestinal failure-associated liver disease. Hepatobiliary Pancreat Dis Int 2024; 23:228-233. [PMID: 36621400 DOI: 10.1016/j.hbpd.2022.12.004] [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: 09/21/2022] [Accepted: 12/08/2022] [Indexed: 01/10/2023]
Abstract
Intestinal failure-associated liver disease (IFALD) is a common complication of long-term parenteral nutrition that is associated with significant morbidity and mortality. It is mainly characterized by cholestasis in children and steatohepatitis in adults. Unfortunately, there is no effective approach to prevent or reverse the disease. Regulated cell death (RCD) represents a fundamental biological paradigm that determines the outcome of a variety of liver diseases. Nowadays cell death is reclassified into several types, based on the mechanisms and morphological phenotypes. Emerging evidence has linked different modes of RCD, such as apoptosis, necroptosis, ferroptosis, and pyroptosis to the pathogenesis of liver diseases. Recent studies have shown that different modes of RCD are present in animal models and patients with IFALD. Understanding the pathogenic roles of cell death may help uncover the underlying mechanisms and develop novel therapeutic strategies in IFALD. In this review, we discuss the current knowledge on how RCD may link to the pathogenesis of IFALD. We highlight examples of cell death-targeted interventions aiming to attenuate the disease, and provide perspectives for future basic and translational research in the field.
Collapse
Affiliation(s)
- Si-Yang Cheng
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Lu Jiang
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Ying Wang
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Wei Cai
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China; Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China.
| |
Collapse
|
5
|
Wu X, Nagy LE, Gautheron J. Mediators of necroptosis: from cell death to metabolic regulation. EMBO Mol Med 2024; 16:219-237. [PMID: 38195700 PMCID: PMC10897313 DOI: 10.1038/s44321-023-00011-z] [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: 09/21/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 01/11/2024] Open
Abstract
Necroptosis, a programmed cell death mechanism distinct from apoptosis, has garnered attention for its role in various pathological conditions. While initially recognized for its involvement in cell death, recent research has revealed that key necroptotic mediators, including receptor-interacting protein kinases (RIPKs) and mixed lineage kinase domain-like protein (MLKL), possess additional functions that go beyond inducing cell demise. These functions encompass influencing critical aspects of metabolic regulation, such as energy metabolism, glucose homeostasis, and lipid metabolism. Dysregulated necroptosis has been implicated in metabolic diseases, including obesity, diabetes, metabolic dysfunction-associated steatotic liver disease (MASLD) and alcohol-associated liver disease (ALD), contributing to chronic inflammation and tissue damage. This review provides insight into the multifaceted role of necroptosis, encompassing both cell death and these extra-necroptotic functions, in the context of metabolic diseases. Understanding this intricate interplay is crucial for developing targeted therapeutic strategies in diseases that currently lack effective treatments.
Collapse
Affiliation(s)
- Xiaoqin Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Laura E Nagy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jérémie Gautheron
- Sorbonne Université, Inserm UMRS_938, Centre de Recherche Saint-Antoine (CRSA), Paris, 75012, France.
| |
Collapse
|
6
|
Wu X, Arya RK, Huang E, McMullen MR, Nagy LE. Receptor-interacting protein 1 and 3 kinase activity are required for high-fat diet induced liver injury in mice. Front Endocrinol (Lausanne) 2023; 14:1267996. [PMID: 38161978 PMCID: PMC10757356 DOI: 10.3389/fendo.2023.1267996] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Background The RIP1-RIP3-MLKL-mediated cell death pathway is associated with progression of non-alcohol-associated fatty liver/steatohepatitis (NAFL/NASH). Previous work identified a critical role for MLKL, the key effector regulating necroptosis, but not RIP3, in mediating high fat diet-induced liver injury in mice. RIP1 and RIP3 have active N-terminus kinase domains essential for activation of MLKL and subsequent necroptosis. However, little is known regarding domain-specific roles of RIP1/RIP3 kinase in liver diseases. Here, we hypothesized that RIP1/RIP3 kinase activity are required for the development of high fat diet-induced liver injury. Methods Rip1K45A/K45A and Rip3K51A/K51A kinase-dead mice on a C57BL/6J background and their littermate controls (WT) were allowed free access to a diet high in fat, fructose and cholesterol (FFC diet) or chow diet. Results Both Rip1K45A/K45A and Rip3K51A/K51A mice were protected against FFC diet-induced steatosis, hepatocyte injury and expression of hepatic inflammatory cytokines and chemokines. FFC diet increased phosphorylation and oligomerization of MLKL and hepatocyte death in livers of WT, but not in Rip3K51A/K51A, mice. Consistent with in vivo data, RIP3 kinase deficiency in primary hepatocytes prevented palmitic acid-induced translocation of MLKL to the cell surface and cytotoxicity. Additionally, loss of Rip1 or Rip3 kinase suppressed FFC diet-mediated formation of crown-like structures (indicators of dead adipocytes) and expression of mRNA for inflammatory response genes in epididymal adipose tissue. Moreover, FFC diet increased expression of multiple adipokines, including leptin and plasminogen activator inhibitor 1, in WT mice, which was abrogated by Rip3 kinase deficiency. Discussion The current data indicate that both RIP1 and RIP3 kinase activity contribute to FFC diet-induced liver injury. This effect of RIP1 and RIP3 kinase deficiency on injury is consistent with the protection of Mlkl-/- mice from high fat diet-induced liver injury, but not the reported lack of protection in Rip3-/- mice. Taken together with previous reports, our data suggest that other domains of RIP3 likely counteract the effect of RIP3 kinase in response to high fat diets.
Collapse
Affiliation(s)
- Xiaoqin Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Rakesh K. Arya
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Emily Huang
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Megan R. McMullen
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Laura E. Nagy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, United States
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
7
|
Chen Y, Liu M, Wei H, Guo J, Zhang S, Bu X, Chen S, Zhang D, Guan S. Alcohol induces hepatocytes necroptosis through the LC3/RIPK1/RIPK3 pathway. Food Chem Toxicol 2023; 182:114124. [PMID: 37898230 DOI: 10.1016/j.fct.2023.114124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 10/30/2023]
Abstract
Excessive alcohol consumption leads to serious liver injury. Necroptosis is a programmed cell death form, which has been confirmed to be involved in alcoholic liver injury. However, the exact mechanism remains still unclear. In this study, we found that ethanol caused hepatocytes necroptosis by activating receptor-interacting serine/threonine-protein kinase 1 and 3 (RIPK1 and RIPK3). Meanwhile, autophagy was activated in ethanol-treated hepatocytes. Accumulative studies have demonstrated a possible link between autophagy and necroptosis. Microtubule-associated protein 1 light chain 3 (LC3), an autophagy marker protein, is essential for autophagosome biogenesis/maturation. But little attention has been paid to its functional role. In this study, we explored whether LC3 was involved in ethanol-induced necroptosis. The data showed that LC3 interacted with RIPK1 and RIPK3 in ethanol-treated AML12 cells and mice liver by co-immunoprecipitation (co-IP) and colocalization assay. Ethanol-induced necrosome formation and subsequent necroptosis were alleviated in hepatocytes by knockdown of LC3 or autophagy inhibitor 3-methyladenine (3-MA). These results demonstrated that LC3 accumulation facilitated the formation of necrosome by LC3-RIPK1 and LC3-RIPK3 interactions, eventually caused hepatocytes necroptosis after acute ethanol exposure. Our current research could potentially offer a new understanding of the intricate mechanisms involved in the development of acute alcoholic liver injury.
Collapse
Affiliation(s)
- Yuelin Chen
- College of Food Science and Engineering, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Meitong Liu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Hongdi Wei
- College of Food Science and Engineering, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Jiakang Guo
- College of Food Science and Engineering, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Shengzhuo Zhang
- College of Food Science and Engineering, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Xiujuan Bu
- College of Food Science and Engineering, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Shanshan Chen
- College of Food Science and Engineering, Jilin University, Changchun, Jilin, 130062, People's Republic of China
| | - Duoduo Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Shuang Guan
- College of Food Science and Engineering, Jilin University, Changchun, Jilin, 130062, People's Republic of China; Key Laboratory of Zoonosis, Ministry of Education College of Veterinary Medicine, Jilin University, Changchun, Jilin, 130062, People's Republic of China.
| |
Collapse
|
8
|
Oh JH, Saeed WK, Kim HY, Lee SM, Lee AH, Park GR, Yoon EL, Jun DW. Hepatic stellate cells activate and avoid death under necroptosis stimuli: Hepatic fibrosis during necroptosis. J Gastroenterol Hepatol 2023; 38:2206-2214. [PMID: 37811601 DOI: 10.1111/jgh.16368] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/29/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND AND AIM Necroptosis is an emerging cell death pathway that allows cells to undergo "cellular suicide" in a caspase-independent manner. We investigated the fate of hepatic stellate cells (HSCs) under necroptotic stimuli. METHODS AND RESULTS The RNA level of mixed lineage kinase domain-like protein (MLKL) is higher in patients with non-alcoholic fatty liver disease than in healthy controls. Hepatic fibrosis was significantly lower in MLKL-KO bile duct ligation (KO-BDL) mice than in wild-type-BDL mice. Necroptotic stimuli caused the death of HT-29 and U937 cells. However, necroptotic stimuli activate HSCs instead of inducing cell death. MLKL inhibitors attenuated fibrogenic changes in HSCs during necroptosis. Unlike HT-29 and U937 cells, MLKL phosphorylation and oligomerization were not observed during necroptosis in HSCs. RNA sequencing showed that NF-κB signaling-related genes were upregulated in HSCs following necroptotic stimulation. Necroptotic stimuli in HSCs increased the nuclear expression of NF-κB, which decreased after MLKL inhibitor treatment. Induction of necroptosis in HSCs led to autophagosome activation and formation, which were attenuated by MLKL inhibitor treatment. CONCLUSION HSCs avoid necroptosis due to the absence of MLKL phosphorylation and oligomerization and are activated through autophagosome and NF-κB pathways.
Collapse
Affiliation(s)
- Ju Hee Oh
- Department of Obstetrics and Gynecology, Institute of Women's Medical Life Science, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Waqar Khalid Saeed
- Department of Biomedical Sciences, Pak-Austria Fachhochschule: Institute of Applied Sciences and Technology, Haripur, Pakistan
| | - Hye Young Kim
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Seung Min Lee
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - A Hyeon Lee
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Gye Ryeol Park
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Eileen L Yoon
- Department of Internal Medicine, College of Medicine, Hanyang University, Seoul, South Korea
| | - Dae Won Jun
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| |
Collapse
|
9
|
Wang Y, Shi C, Guo J, Zhang Y, Gong Z. Distinct Types of Cell Death and Implications in Liver Diseases: An Overview of Mechanisms and Application. J Clin Transl Hepatol 2023; 11:1413-1424. [PMID: 37719956 PMCID: PMC10500292 DOI: 10.14218/jcth.2023.00132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/17/2023] [Accepted: 07/12/2023] [Indexed: 09/19/2023] Open
Abstract
Cell death is associated with a variety of liver diseases, and hepatocyte death is a core factor in the occurrence and progression of liver diseases. In recent years, new cell death modes have been identified, and certain biomarkers have been detected in the circulation during various cell death modes that mediate liver injury. In this review, cell death modes associated with liver diseases are summarized, including some cell death modes that have emerged in recent years. We described the mechanisms associated with liver diseases and summarized recent applications of targeting cell death in liver diseases. It provides new ideas for the diagnosis and treatment of liver diseases. In addition, multiple cell death modes can contribute to the same liver disease. Different cell death modes are not isolated, and they interact with each other in liver diseases. Future studies may focus on exploring the regulation between various cell death response pathways in liver diseases.
Collapse
Affiliation(s)
- Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yanqiong Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
10
|
Kholodenko IV, Yarygin KN. Hepatic Macrophages as Targets for the MSC-Based Cell Therapy in Non-Alcoholic Steatohepatitis. Biomedicines 2023; 11:3056. [PMID: 38002056 PMCID: PMC10669188 DOI: 10.3390/biomedicines11113056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a serious public health issue associated with the obesity pandemic. Obesity is the main risk factor for the non-alcoholic fatty liver disease (NAFLD), which progresses to NASH and then to end-stage liver disease. Currently, there are no specific pharmacotherapies of NAFLD/NASH approved by the FDA or other national regulatory bodies and the treatment includes lifestyle adjustment and medicines for improving lipid metabolism, enhancing sensitivity to insulin, balancing oxidation, and counteracting fibrosis. Accordingly, further basic research and development of new therapeutic approaches are greatly needed. Mesenchymal stem cells (MSCs) and MSC-derived extracellular vesicles prevent induced hepatocyte death in vitro and attenuate NASH symptoms in animal models of the disease. They interact with hepatocytes directly, but also target other liver cells, including Kupffer cells and macrophages recruited from the blood flow. This review provides an update on the pathogenesis of NAFLD/NASH and the key role of macrophages in the development of the disease. We examine in detail the mechanisms of the cross-talk between the MSCs and the macrophages, which are likely to be among the key targets of MSCs and their derivatives in the course of NAFLD/NASH cell therapy.
Collapse
Affiliation(s)
- Irina V. Kholodenko
- Laboratory of Cell Biology, Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia;
| | | |
Collapse
|
11
|
Endo Y, Winarski KL, Sajib MS, Ju A, Wu WJ. Atezolizumab Induces Necroptosis and Contributes to Hepatotoxicity of Human Hepatocytes. Int J Mol Sci 2023; 24:11694. [PMID: 37511454 PMCID: PMC10380327 DOI: 10.3390/ijms241411694] [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: 05/04/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Atezolizumab is an immune checkpoint inhibitor (ICI) targeting PD-L1 for treatment of solid malignancies. Immune checkpoints control the immune tolerance, and the adverse events such as hepatotoxicity induced by ICIs are often considered as an immune-related adverse event (irAE). However, PD-L1 is also highly expressed in normal tissues, e.g., hepatocytes. It is still not clear whether, targeting PD-L1 on hepatocytes, the atezolizumab may cause damage to liver cells contributing to hepatotoxicity. Here, we reveal a novel mechanism by which the atezolizumab induces hepatotoxicity in human hepatocytes. We find that the atezolizumab treatment increases a release of LDH in the cell culture medium of human hepatocytes (human primary hepatocytes and THLE-2 cells), decreases cell viability, and inhibits the THLE-2 and THLE-3 cell growth. We demonstrate that both the atezolizumab and the conditioned medium (T-CM) derived from activated T cells can induce necroptosis of the THLE-2 cells, which is underscored by the fact that the atezolizumab and T-CM enhance the phosphorylation of RIP3 and MLKL proteins. Furthermore, we also show that necrostatin-1, a necrosome inhibitor, decreases the amount of phosphorylated RIP3 induced by the atezolizumab, resulting in a reduced LDH release in the culture media of the THLE-2 cells. This finding is further supported by the data that GSK872 (a RIP3 inhibitor) significantly reduced the atezolizumab-induced LDH release. Taken together, our data indicate that the atezolizumab induces PD-L1-mediated necrosome formation, contributing to hepatotoxicity in PD-L1+-human hepatocytes. This study provides the molecular basis of the atezolizumab-induced hepatotoxicity and opens a new avenue for developing a novel therapeutic approach to reducing hepatotoxicity induced by ICIs.
Collapse
Affiliation(s)
- Yukinori Endo
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Katie L Winarski
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Md Sanaullah Sajib
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Anna Ju
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Wen Jin Wu
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| |
Collapse
|
12
|
Wu J, Kim A, Wu X, Ray S, Allende DS, Welch N, Bellar A, Dasarathy J, Dasarathy S, Nagy LE. 5S rRNA pseudogene transcripts are associated with interferon production and inflammatory responses in alcohol-associated hepatitis. Hepatology 2023; 77:1983-1997. [PMID: 36645226 PMCID: PMC10192046 DOI: 10.1097/hep.0000000000000024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/15/2022] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND AIMS Interferon (IFN) signaling is critical to the pathogenesis of alcohol-associated hepatitis (AH), yet the mechanisms for activation of this system are elusive. We hypothesize that host-derived 5S rRNA pseudogene (RNA5SP) transcripts regulate IFN production and modify immunity in AH. APPROACH AND RESULTS Mining of transcriptomic datasets revealed that in patients with severe alcohol-associated hepatitis (sAH), hepatic expression of genes regulated by IFNs was perturbed and gene sets involved in IFN production were enriched. RNA5SP transcripts were also increased and correlated with expression of type I IFNs. Interestingly, inflammatory mediators upregulated in sAH, but not in other liver diseases, were positively correlated with certain RNA5SP transcripts. Real-time quantitative PCR demonstrated that RNA5SP transcripts were upregulated in peripheral blood mononuclear cells (PBMCs) from patients with sAH. In sAH livers, increased 5S rRNA and reduced nuclear MAF1 (MAF1 homolog, negative regulator of RNA polymerase III) protein suggested a higher activity of RNA polymerase III (Pol III); inhibition of Pol III reduced RNA5SP expression in monocytic THP-1 cells. Expression of several RNA5SP transcript-interacting proteins was downregulated in sAH, potentially unmasking transcripts to immunosensors. Indeed, siRNA knockdown of interacting proteins potentiated the immunostimulatory activity of RNA5SP transcripts. Molecular interaction and cell viability assays demonstrated that RNA5SP transcripts adopted Z-conformation and contributed to ZBP1-mediated caspase-independent cell death. CONCLUSIONS Increased expression and binding availability of RNA5SP transcripts was associated with hepatic IFN production and inflammation in sAH. These data identify RNA5SP transcripts as a potential target to mitigate inflammation and hepatocellular injury in AH.
Collapse
Affiliation(s)
- Jianguo Wu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
| | - Adam Kim
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Xiaoqin Wu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Semanti Ray
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Nicole Welch
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Annette Bellar
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jaividhya Dasarathy
- Department of Family Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Srinivasan Dasarathy
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Laura E. Nagy
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio, USA
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
13
|
Wu X, Fan X, Miyata T, Kim A, Cajigas-Du Ross CK, Ray S, Huang E, Taiwo M, Arya R, Wu J, Nagy LE. Recent Advances in Understanding of Pathogenesis of Alcohol-Associated Liver Disease. ANNUAL REVIEW OF PATHOLOGY 2023; 18:411-438. [PMID: 36270295 PMCID: PMC10060166 DOI: 10.1146/annurev-pathmechdis-031521-030435] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alcohol-associated liver disease (ALD) is one of the major diseases arising from chronic alcohol consumption and is one of the most common causes of liver-related morbidity and mortality. ALD includes asymptomatic liver steatosis, fibrosis, cirrhosis, and alcohol-associated hepatitis and its complications. The progression of ALD involves complex cell-cell and organ-organ interactions. We focus on the impact of alcohol on dysregulation of homeostatic mechanisms and regulation of injury and repair in the liver. In particular, we discuss recent advances in understanding the disruption of balance between programmed cell death and prosurvival pathways, such as autophagy and membrane trafficking, in the pathogenesis of ALD. We also summarize current understanding of innate immune responses, liver sinusoidal endothelial cell dysfunction and hepatic stellate cell activation, and gut-liver and adipose-liver cross talk in response to ethanol. In addition,we describe the current potential therapeutic targets and clinical trials aimed at alleviating hepatocyte injury, reducing inflammatory responses, and targeting gut microbiota, for the treatment of ALD.
Collapse
Affiliation(s)
- Xiaoqin Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Xiude Fan
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Tatsunori Miyata
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Adam Kim
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Christina K Cajigas-Du Ross
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Semanti Ray
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Emily Huang
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Moyinoluwa Taiwo
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Rakesh Arya
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Jianguo Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Laura E Nagy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA;
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
14
|
Preston SP, Stutz MD, Allison CC, Nachbur U, Gouil Q, Tran BM, Duvivier V, Arandjelovic P, Cooney JP, Mackiewicz L, Meng Y, Schaefer J, Bader SM, Peng H, Valaydon Z, Rajasekaran P, Jennison C, Lopaticki S, Farrell A, Ryan M, Howell J, Croagh C, Karunakaran D, Schuster-Klein C, Murphy JM, Fifis T, Christophi C, Vincan E, Blewitt ME, Thompson A, Boddey JA, Doerflinger M, Pellegrini M. Epigenetic Silencing of RIPK3 in Hepatocytes Prevents MLKL-mediated Necroptosis From Contributing to Liver Pathologies. Gastroenterology 2022; 163:1643-1657.e14. [PMID: 36037995 DOI: 10.1053/j.gastro.2022.08.040] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS Necroptosis is a highly inflammatory mode of cell death that has been implicated in causing hepatic injury including steatohepatitis/ nonalcoholic steatohepatitis (NASH); however, the evidence supporting these claims has been controversial. A comprehensive, fundamental understanding of cell death pathways involved in liver disease critically underpins rational strategies for therapeutic intervention. We sought to define the role and relevance of necroptosis in liver pathology. METHODS Several animal models of human liver pathology, including diet-induced steatohepatitis in male mice and diverse infections in both male and female mice, were used to dissect the relevance of necroptosis in liver pathobiology. We applied necroptotic stimuli to primary mouse and human hepatocytes to measure their susceptibility to necroptosis. Paired liver biospecimens from patients with NASH, before and after intervention, were analyzed. DNA methylation sequencing was also performed to investigate the epigenetic regulation of RIPK3 expression in primary human and mouse hepatocytes. RESULTS Identical infection kinetics and pathologic outcomes were observed in mice deficient in an essential necroptotic effector protein, MLKL, compared with control animals. Mice lacking MLKL were indistinguishable from wild-type mice when fed a high-fat diet to induce NASH. Under all conditions tested, we were unable to induce necroptosis in hepatocytes. We confirmed that a critical activator of necroptosis, RIPK3, was epigenetically silenced in mouse and human primary hepatocytes and rendered them unable to undergo necroptosis. CONCLUSIONS We have provided compelling evidence that necroptosis is disabled in hepatocytes during homeostasis and in the pathologic conditions tested in this study.
Collapse
Affiliation(s)
- Simon P Preston
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael D Stutz
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Cody C Allison
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ueli Nachbur
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Quentin Gouil
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Bang Manh Tran
- Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Valerie Duvivier
- Cardiovascular and Metabolic Disease Center for Therapeutic Innovation, SERVIER Group, Suresnes, France
| | - Philip Arandjelovic
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - James P Cooney
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Liana Mackiewicz
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Yanxiang Meng
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jan Schaefer
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Stefanie M Bader
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Hongke Peng
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Zina Valaydon
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Pravin Rajasekaran
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Charlie Jennison
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Sash Lopaticki
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Ann Farrell
- Department of Gastroenterology, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Marno Ryan
- Department of Gastroenterology, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jess Howell
- Department of Gastroenterology, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Catherine Croagh
- Department of Gastroenterology, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Denuja Karunakaran
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia; Monash Biomedicine Discovery Institute and Victorian Heart Institute, Monash University, Clayton, Victoria, Australia
| | - Carole Schuster-Klein
- Cardiovascular and Metabolic Disease Center for Therapeutic Innovation, SERVIER Group, Suresnes, France
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Theodora Fifis
- Department of Gastroenterology, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christopher Christophi
- Department of Gastroenterology, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Elizabeth Vincan
- Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Victorian Infectious Disease Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia; Curtin Medical School, Curtin University, Perth, Western Australia, Australia
| | - Marnie E Blewitt
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Alexander Thompson
- Department of Gastroenterology, St. Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - Justin A Boddey
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Marcel Doerflinger
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.
| | - Marc Pellegrini
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
15
|
Chaouhan HS, Vinod C, Mahapatra N, Yu SH, Wang IK, Chen KB, Yu TM, Li CY. Necroptosis: A Pathogenic Negotiator in Human Diseases. Int J Mol Sci 2022; 23:12714. [PMID: 36361505 PMCID: PMC9655262 DOI: 10.3390/ijms232112714] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/25/2022] Open
Abstract
Over the past few decades, mechanisms of programmed cell death have attracted the scientific community because they are involved in diverse human diseases. Initially, apoptosis was considered as a crucial mechanistic pathway for programmed cell death; recently, an alternative regulated mode of cell death was identified, mimicking the features of both apoptosis and necrosis. Several lines of evidence have revealed that dysregulation of necroptosis leads to pathological diseases such as cancer, cardiovascular, lung, renal, hepatic, neurodegenerative, and inflammatory diseases. Regulated forms of necrosis are executed by death receptor ligands through the activation of receptor-interacting protein kinase (RIPK)-1/3 and mixed-lineage kinase domain-like (MLKL), resulting in the formation of a necrosome complex. Many papers based on genetic and pharmacological studies have shown that RIPKs and MLKL are the key regulatory effectors during the progression of multiple pathological diseases. This review focused on illuminating the mechanisms underlying necroptosis, the functions of necroptosis-associated proteins, and their influences on disease progression. We also discuss numerous natural and chemical compounds and novel targeted therapies that elicit beneficial roles of necroptotic cell death in malignant cells to bypass apoptosis and drug resistance and to provide suggestions for further research in this field.
Collapse
Affiliation(s)
- Hitesh Singh Chaouhan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Ch Vinod
- Department of Biological Sciences, School of Applied Sciences, KIIT University, Bhubaneshwar 751024, India
| | - Nikita Mahapatra
- Department of Biological Sciences, School of Applied Sciences, KIIT University, Bhubaneshwar 751024, India
| | - Shao-Hua Yu
- Department of Emergency Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - I-Kuan Wang
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
| | - Kuen-Bao Chen
- Department of Anesthesiology, China Medical University Hospital, Taichung 40402, Taiwan
| | - Tung-Min Yu
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40402, Taiwan
| | - Chi-Yuan Li
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Anesthesiology, China Medical University Hospital, Taichung 40402, Taiwan
| |
Collapse
|
16
|
Singh V, Huang E, Pathak V, Willard BB, Allende DS, Nagy LE. Phosphoproteomics identifies pathways underlying the role of receptor-interaction protein kinase 3 in alcohol-associated liver disease and uncovers apoptosis signal-regulating kinase 1 as a target. Hepatol Commun 2022; 6:2022-2041. [PMID: 35438255 PMCID: PMC9315126 DOI: 10.1002/hep4.1956] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/14/2022] [Accepted: 03/12/2022] [Indexed: 01/21/2023] Open
Abstract
Receptor-interaction protein kinase 3 (RIP3), a critical determinant of the necroptotic pathway of programmed cell death, contributes to injury in murine models of alcohol-associated liver disease (ALD); however, the underlying mechanisms are unknown. We investigated the effect of chronic ethanol feeding on the hepatic phosphoproteome in C57BL/6 and RIP3-deficient (Rip3-/- ) mice, focusing on death receptor (DR) signaling pathways. C57BL/6 and Rip3-/- mice were fed an ethanol-containing liquid diet or pair-fed control diet. A label-free mass spectrometry-based approach identified differentially phosphorylated proteins that were mapped to pathways affected by ethanol and Rip3 genotype. Identified targets were validated in both the murine model of ALD and in liver tissue from patients with alcohol-associated hepatitis (AH) and healthy controls. Chronic ethanol dysregulated hepatic tumor necrosis factor-induced DR signaling pathways. Of particular importance, chronic ethanol feeding to C57BL/6 mice decreased the phosphorylation of apoptosis signal-regulating kinase 1 (ASK1) at serine (S)1036/S1040 (S1029/S1033 human), sites linked with the inhibition of ASK1 death-promoting activity. This decrease in phosphorylation of inhibitory sites was muted in Rip3-/- mice. Decreased phosphorylation at S1033 was also lower in liver of patients with severe AH compared to healthy controls, and phosphorylation at the ASK1 activation site (threonine [Thr]-838) was increased in patients with AH. The net impact of these changes in phosphorylation of ASK1 was associated with increased phosphorylation of p38, a downstream target of ASK1, in patients with AH and C57BL/6 but not Rip3-/- mice. Similarly, chronic ethanol feeding affected the c-Jun N-terminal kinase pathway in C57BL/6 but not Rip3-/- mice. Taken together, our data indicate that changes in inhibitory phosphorylation of ASK1 are an important target in ALD and suggest the involvement of noncanonical functions of Rip3 in ALD.
Collapse
Affiliation(s)
- Vaibhav Singh
- Department of Inflammation and Immunity, Northern Ohio Alcohol Center, Center for Liver Disease ResearchCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - Emily Huang
- Department of Inflammation and Immunity, Northern Ohio Alcohol Center, Center for Liver Disease ResearchCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - Vai Pathak
- Department of Quantitative Health SciencesCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | - Belinda B Willard
- Proteomics and Metabolomics CoreCleveland ClinicLerner Research InstituteClevelandOhioUSA
| | | | - Laura E Nagy
- Department of Inflammation and Immunity, Northern Ohio Alcohol Center, Center for Liver Disease ResearchCleveland ClinicLerner Research InstituteClevelandOhioUSA
| |
Collapse
|
17
|
Ripk3 signaling regulates HSCs during stress and represses radiation-induced leukemia in mice. Stem Cell Reports 2022; 17:1428-1441. [PMID: 35561683 PMCID: PMC9213819 DOI: 10.1016/j.stemcr.2022.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 01/03/2023] Open
Abstract
Receptor-interacting protein kinase 3 (Ripk3) is one of the critical mediators of inflammatory cytokine-stimulated signaling. Here we show that Ripk3 signaling selectively regulates both the number and the function of hematopoietic stem cells (HSCs) during stress conditions. Ripk3 signaling is not required for normal homeostatic hematopoiesis. However, in response to serial transplantation, inactivation of Ripk3 signaling prevents stress-induced HSC exhaustion and functional HSC attenuation, while in response to fractionated low doses of ionizing radiation (IR), inactivation of Ripk3 signaling accelerates leukemia/lymphoma development. In both situations, Ripk3 signaling is primarily stimulated by tumor necrosis factor-α. Activated Ripk3 signaling promotes the elimination of HSCs during serial transplantation and pre-leukemia stem cells (pre-LSCs) during fractionated IR by inducing Mlkl-dependent necroptosis. Activated Ripk3 signaling also attenuates HSC functioning and represses a pre-LSC-to-LSC transformation by promoting Mlkl-independent senescence. Furthermore, we demonstrate that Ripk3 signaling induces senescence in HSCs and pre-LSCs by attenuating ISR-mediated mitochondrial quality control. Ripk3-Mlkl signaling is not required for normal homeostatic hematopoiesis Ripk3-Mlkl signaling promotes HSC loss during serial transplantation or low-dose IR Tnf-α-Ripk3 signaling prevents leukemia development after exposure to low-dose IR Ripk3 represses pre-LSCs by inducing Mlkl necroptosis and PDC-OXPHOS-ROS senescence
Collapse
|
18
|
Zhou Y, Wu R, Wang X, Bao X, Lu C. Roles of necroptosis in alcoholic liver disease and hepatic pathogenesis. Cell Prolif 2022; 55:e13193. [PMID: 35083817 PMCID: PMC8891559 DOI: 10.1111/cpr.13193] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic alcohol consumption can cause alcoholic liver disease (ALD), leading to morbidity and mortality worldwide. Complex disease progression of ALD varies from alcoholic fatty liver to alcoholic steatohepatitis, eventually contributing to fibrosis and cirrhosis. Accumulating evidence revealed that necroptosis, a way of programmed cell death different from apoptosis and traditional necrosis, is involved in the underlying pathogenic molecular mechanism of ALD. Receptor‐interacting protein kinase 1 (RIPK1), RIPK3 and mixed‐lineage kinase domain‐like pseudokinase have been implicated as key mediators to execute necroptosis. Also, necroptosis has gained increasing attention due to its potential association with primary pathological hallmarks of ALD, including oxidative stress, hepatic steatosis and inflammation. This review summarizes the recent progress on the roles and mechanisms of necroptosis and focuses on the crosstalk between necroptosis and the other pathogenesis of ALD, providing a theoretical basis for targeting necroptosis as a novel treatment for ALD.
Collapse
Affiliation(s)
- Ying Zhou
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Ruoman Wu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xinqi Wang
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
19
|
Chen H, McKeen T, Chao X, Chen A, Deng F, Jaeschke H, Ding WX, Ni HM. The role of MLKL in Hepatic Ischemia-Reperfusion Injury of Alcoholic Steatotic Livers. Int J Biol Sci 2022; 18:1096-1106. [PMID: 35173541 PMCID: PMC8771841 DOI: 10.7150/ijbs.67533] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022] Open
Abstract
Alcohol-related liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) are the primary causes of chronic liver disease in western countries. Liver transplantation is currently one of the most efficient approaches to save patients with liver failure, which is often associated with hepatic ischemia-reperfusion (IR) injury. IR injury is exacerbated by hepatic steatosis, yet the mechanism remains elusive. Necroptosis is a form of regulated cell death mediated by receptor-interacting protein kinase 1 (RIP1), RIP3 and mixed lineage kinase domain-like (MLKL) protein, which has been implicated in the pathogenesis of ALD and NAFLD. Though necroptosis plays an important role in IR injury of high fat diet - induced steatotic livers, the role of necroptosis in IR injury of ethanol - induced steototic livers has not been investigated. In the present study, we used chronic plus binge alcohol (Gao-binge) feeding followed by IR surgery to investigate IR liver injury with ethanol-associated steatosis. We found that the levels of key necroptotic proteins MLKL and RIP3 increased in alcohol-fed mouse livers. Moreover, we observed increased liver injury after IR in control diet-fed mice, which was further exacerbated by alcohol feeding based on serum alanine aminotransferase (ALT) levels and TUNEL staining of necrotic cells. Hepatic neutrophil infiltration also increased in alcohol-fed mice after IR surgery. However, deletion of Mlkl did not protect against IR liver injury in alcohol-fed mice compared with matched wild-type mice. In conclusion, alcoholic steatosis promotes IR injury, which seems to be independent of MLKL-mediated necroptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hong-Min Ni
- ✉ Corresponding author: Hong-Min Ni, MD., Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, MS 1018, 3901 Rainbow Blvd., Kansas City, Kansas 66160, Phone: 913-588-8999;
| |
Collapse
|
20
|
Huang MY, Wan DW, Deng J, Guo WJ, Huang Y, Chen H, Xu DL, Jiang ZG, Xue Y, He YH. Downregulation of RIP3 Improves the Protective Effect of ATF6 in an Acute Liver Injury Model. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8717565. [PMID: 34778458 PMCID: PMC8589516 DOI: 10.1155/2021/8717565] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND Activating transcription factor 6 (ATF6) and receptor-interacting protein 3 (RIP3) are important signaling proteins in endoplasmic reticulum (ER) stress and necroptosis, respectively. However, their regulatory relationship and clinical significance are unknown. We investigate the impact of ATF6 on RIP3 expression, and its role in hepatocyte necroptosis in an acute liver injury model. METHODS In vivo and in vitro experiments were carried out. LO2 cells were treated with thapsigargin (TG). In vivo, male BALB/c mice were treated with carbon tetrachloride (CCl4, 1 mL/kg) or tunicamycin (TM, 2 mg/kg). Then, the impact of ATF6 or RIP3 silencing on liver injury, hepatocyte necroptosis, and ER stress-related protein expression was examined. RESULTS TG induced ER stress and necroptosis and ATF6 and RIP3 expression in LO2 cells. The knockdown of ATF6 significantly decreased RIP3 expression (p < 0.05) and increased ER stress and necroptosis. The downregulation of RIP3 significantly reduced necroptosis and ER stress (p < 0.05). Similar results were observed in CCl4 or the TM-induced mouse model. The knockdown of ATF6 significantly decreased CCl4-induced RIP3 expression and increased liver injury, necroptosis, and ER stress in mice livers (p < 0.05). In contrast, the downregulation of RIP3 significantly reduced liver injury, hepatocyte necroptosis, and ER stress. CONCLUSIONS Hepatocyte ATF6 has multiple roles in acute liver injury. It reduces hepatocyte necroptosis via negative feedback regulation of ER stress. In addition, ATF6 can upregulate the expression of RIP3, which is not helpful to the recovery process. However, downregulating RIP3 reduces hepatocyte necroptosis by promoting the alleviation of ER stress. The findings suggest that RIP3 could be a plausible target for the treatment of liver injury.
Collapse
Affiliation(s)
- Mei-Ying Huang
- Department of Pediatrics, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou, China
| | - Dian-Wei Wan
- Department of Infectious Diseases, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou, China
| | - Jie Deng
- Department of Infectious Diseases, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou, China
| | - Wen-Jie Guo
- Department of Infectious Diseases, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou, China
| | - Yue Huang
- Department of Infectious Diseases, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou, China
| | - Huan Chen
- Department of Infectious Diseases, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou, China
| | - De-Lin Xu
- Department of Cell Biology, Zunyi Medical University, Zunyi, 563099 Guizhou, China
| | - Zhi-Gang Jiang
- School of Public Health, Zunyi Medical University, Zunyi, 563099 Guizhou, China
| | - Yuan Xue
- Department of Liver Diseases, The Third People's Hospital of Changzhou, Changzhou, 213000 Jiangsu Province, China
| | - Yi-Huai He
- Department of Infectious Diseases, The Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 Guizhou, China
| |
Collapse
|
21
|
Wang S, Chao X, Jiang X, Wang T, Rodriguez Y, Yang L, Pacher P, Ni HM, Ding WX. Loss of acinar cell VMP1 triggers spontaneous pancreatitis in mice. Autophagy 2021; 18:1572-1582. [PMID: 34709991 DOI: 10.1080/15548627.2021.1990672] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The pathogenesis of pancreatitis has been linked to disruption of organelle homeostasis including macroautophagy/autophagy dysfunction and endoplasmic reticulum (ER) stress. However, the direct impact of aberrant organelle function on pancreatitis initiation and progression is largely unknown. Recently an ER membrane protein, VMP1 (vacuole membrane protein 1), has been reported to play a crucial role in autophagosome formation. Notably, we found that VMP1 is downregulated in both human chronic pancreatitis (CP) and experimental mouse acute pancreatitis (AP). Pancreatic acinar cell-specific vmp1 deletion promotes inflammation, acinar-to-ductal metaplasia, and fibrosis in mice, sharing histological similarities with human CP. Mechanistically, loss of pancreatic VMP1 leads to defective autophagic degradation and ER stress as well as activation of the NFE2L2/Nrf2 pathway. Genetic ablation of NFE2L2 attenuated pancreatitis in VMP1-deficient mice. Our data highlight the importance of VMP1 in modulating an integrated organelle stress response and its functional role in maintaining pancreas homeostasis in the context of CP.Abbreviations: AMY: amylase; ADM: acinar-to-ductal metaplasia; AP: acute pancreatitis; CASP3: caspase 3; CP: chronic pancreatitis; DDIT3/CHOP: DNA damage inducible transcript 3; DKO, double knockout; ER: endoplasmic reticulum; GCLC: glutamate-cysteine ligase catalytic subunit; GCLM: glutamate-cysteine ligase modifier subunit; HSPA5/BIP: heat shock protein family A (Hsp70) member 5; KO: knockout; KRT19/CK19: keratin 19; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MPO: myeloperoxidase; NFE2L2/NRF2: nuclear factor, erythroid 2 like 2; ND: normal donor; NQO1: NAD(P)H quinone dehydrogenase 1; PCNA: proliferating cell nuclear antigen; RIPA: radio-immunoprecipitation; SQSTM1/p62: sequestosome 1; SOX9: SRY-box transcription factor 9; TAP: trypsinogen activation peptide; TFEB: transcription factor EB; TUNEL: terminal deoxynucleotidyl transferase dUTP nick end labeling; UB: ubiquitin; VMP1: vacuole membrane protein 1; XBP1: X-box binding protein 1; YAP1, Yes1 associated transcriptional regulator; ZG: zymogen granule.
Collapse
Affiliation(s)
- Shaogui Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.,International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Xiaoxiao Jiang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tiantian Wang
- International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yssa Rodriguez
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, Pappajohn Biomedical Institute, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
22
|
Construction, Identification and Analysis of the Interaction Network of African Swine Fever Virus MGF360-9L with Host Proteins. Viruses 2021; 13:v13091804. [PMID: 34578385 PMCID: PMC8473002 DOI: 10.3390/v13091804] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 12/24/2022] Open
Abstract
African swine fever virus (ASFV) is prevalent in many countries and is a contagious and lethal virus that infects pigs, posing a threat to the global pig industry and public health. The interaction between the virus and the host is key to unlocking the mystery behind viral pathogenesis. A comprehensive understanding of the viral and host protein interaction may provide clues for developing new antiviral strategies. Here, we show a network of ASFV MGF360-9L protein interactions in porcine kidney (PK-15) cells. Overall, 268 proteins that interact with MGF360-9L are identified using immunoprecipitation and liquid chromatography–mass spectrometry (LC-MS). Accordingly, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted, and the protein–protein interaction (PPI) network was created. It was speculated that the cellular proteins interacting with MGF360-9L are involved in protein binding, metabolism, and the innate immune response. Proteasome subunit alpha type (PSMA3), 26S protease regulatory subunit 4 (PSMC1), autophagy and beclin 1 regulator 1 (AMBRA1), and DEAD-box helicase 20 (DDX20) could interact with MGF360-9L in vitro. PSMA3 and PSMC1 overexpression significantly promoted ASFV replication, and MGF360-9L maintained the transcriptional level of PSMA3 and PSMC1. Here, we show the interaction between ASFV MGF360-9L and cellular proteins and elucidate the virus–host interaction network, which effectively provides useful protein-related information that can enable further study of the potential mechanism and pathogenesis of ASFV infection.
Collapse
|
23
|
Chao X, Wang S, Yang L, Ni HM, Ding WX. Trehalose activates hepatic transcription factor EB (TFEB) but fails to ameliorate alcohol-impaired TFEB and liver injury in mice. Alcohol Clin Exp Res 2021; 45:1950-1964. [PMID: 34486131 DOI: 10.1111/acer.14695] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/27/2021] [Accepted: 08/04/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Recent evidence demonstrates that alcohol activates the mechanistic target of rapamycin (mTOR) and impairs hepatic transcription factor EB (TFEB) reducing autophagy and contributing to alcohol-induced liver injury. Trehalose, a disaccharide, activates TFEB and protects against diet-induced nonalcoholic fatty liver disease in mice. The aim of the present study was to investigate whether trehalose would reverse the impairment of TFEB induced by alcohol and protect against alcohol-induced liver injury. METHODS Male C57BL/6J mice were subjected to chronic-plus-binge (Gao-binge) alcohol feeding with and without trehalose supplementation. Some mice were also administrered Alda-1, an aldehyde dehydrogenase 2 agonist. RESULTS We found that Alda-1 did not affect Gao-binge alcohol-induced mTOR activation and impaired TFEB in mouse livers. Trehalose increased TFEB nuclear translocation, elevated levels of LC3-II and lysosomal proteins in mouse livers and cultured AML12 cells, confirming the activation of TFEB by trehalose. However, trehalose did not improve the impairment in TFEB induced by Gao-binge alcohol. Both Alda-1 and trehalose failed to protect against Gao-binge alcohol-induced steatosis and liver injury, based on the serum levels of alanine aminotransferase (ALT), histological analysis, and levels of hepatic triglyceride. Interestingly, trehalose increased expression of pro-inflammatory genes in mouse macrophage RAW264.7 cells and slightly increased the infiltration of hepatic neutrophils and inflammatory cytokine gene expression in Gao-binge alcohol-fed mice livers. CONCLUSIONS Trehalose fails to improve the impaired TFEB induced by Gao-binge alcohol and does not protect against alcohol-induced liver injury.
Collapse
Affiliation(s)
- Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Shaogui Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ling Yang
- Department of Anatomy and Cell Biology, Fraternal Order of Eagles Diabetes Research Center, The Pappajohn Biomedical Institute, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
24
|
Fan X, Wu J, Poulsen KL, Kim A, Wu X, Huang E, Miyata T, Sanz‐Garcia C, Nagy LE. Identification of a MicroRNA-E3 Ubiquitin Ligase Regulatory Network for Hepatocyte Death in Alcohol-Associated Hepatitis. Hepatol Commun 2021; 5:830-845. [PMID: 34027272 PMCID: PMC8122386 DOI: 10.1002/hep4.1677] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 01/09/2023] Open
Abstract
We aimed to identify a microRNA (miRNA)-E3 ubiquitin ligase regulatory network for protein substrates enriched in cell death pathways and investigate the underlying molecular mechanisms in alcohol-associated hepatitis (AH). An miRNA-E3 ubiquitin ligase regulatory network for protein substrates enriched in cell death pathways was constructed using integrated bioinformatics analysis. Differentially expressed hub miRNAs (GSE59492) and their validated miRNA target genes (GSE28619) were identified in the liver of patients with AH compared with healthy controls. Liver samples from patients with AH and healthy individuals and mice exposed to Gao-binge (acute on chronic) ethanol were used for experimental validation. Using hub miRNAs identified by weighted correlation network analysis, a miRNA-E3 ubiquitin ligase regulatory network was established based on 17 miRNAs and 7 E3 ligase genes targeted by these miRNAs that were down-regulated in AH. Among the miRNAs in this regulatory network, miR-150-5p was the only miRNA regulating the E3 ligase cytokine-inducible SH2 containing protein (CISH), the E3 ligase that regulates the largest number of substrates among all E3 ligase family members. Therefore, the CISH regulatory pathway for ubiquitinated substrates was selected for subsequent experimental validation. Consistent with the bioinformatics analysis results, expression of miR-150-5p was markedly increased, while CISH was decreased, in the livers of patients with AH and mice exposed to Gao-binge ethanol. Moreover, ubiquitination of Fas-associated protein with death domain, a predicted CISH substrate involved in the regulation of programmed cell death, was reduced in livers from mice after Gao-binge ethanol. Conclusion: Identification of the miRNA-E3 ubiquitin ligase regulatory network for protein substrates enriched in the cell death pathways provides insights into the molecular mechanisms contributing to hepatocyte death in AH.
Collapse
Affiliation(s)
- Xiude Fan
- Department of Inflammation and ImmunityCleveland ClinicClevelandOHUSA
- Department of Infectious DiseasesFirst Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxi ProvinceChina
| | - Jianguo Wu
- Department of Inflammation and ImmunityCleveland ClinicClevelandOHUSA
| | - Kyle L. Poulsen
- Department of Inflammation and ImmunityCleveland ClinicClevelandOHUSA
| | - Adam Kim
- Department of Inflammation and ImmunityCleveland ClinicClevelandOHUSA
| | - Xiaoqin Wu
- Department of Inflammation and ImmunityCleveland ClinicClevelandOHUSA
| | - Emily Huang
- Department of Inflammation and ImmunityCleveland ClinicClevelandOHUSA
| | - Tatsunori Miyata
- Department of Inflammation and ImmunityCleveland ClinicClevelandOHUSA
| | | | - Laura E. Nagy
- Department of Inflammation and ImmunityCleveland ClinicClevelandOHUSA
- Department of Gastroenterology and HepatologyCleveland ClinicClevelandOHUSA
- Department of Molecular MedicineCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
25
|
Dai W, Cheng J, Leng X, Hu X, Ao Y. The potential role of necroptosis in clinical diseases (Review). Int J Mol Med 2021; 47:89. [PMID: 33786617 PMCID: PMC8012024 DOI: 10.3892/ijmm.2021.4922] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
As an important type of programmed cell death in addition to apoptosis, necroptosis occurs in a variety of pathophysiological processes, including infections, liver diseases, kidney injury, neurodegenerative diseases, cardiovascular diseases, and human tumors. It can be triggered by a variety of factors, such as tumor necrosis factor receptor and Toll‑like receptor families, intracellular DNA and RNA sensors, and interferon, and is mainly mediated by receptor‑interacting protein kinase 1 (RIP1), RIP3, and mixed lineage kinase domain‑like protein. A better understanding of the mechanism of necroptosis may be useful in the development of novel drugs for necroptosis‑related diseases. In this review, the focus is on the molecular mechanisms of necroptosis, exploring the role of necroptosis in different pathologies, discussing their potential as a novel therapeutic target for disease therapy, and providing suggestions for further study in this area.
Collapse
Affiliation(s)
- Wenli Dai
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Jin Cheng
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Xi Leng
- Medical Imaging Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, P.R. China
| | - Xiaoqing Hu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Yingfang Ao
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing 100191, P.R. China
| |
Collapse
|
26
|
Miyata T, Wu X, Fan X, Huang E, Sanz-Garcia C, Ross CKCD, Roychowdhury S, Bellar A, McMullen MR, Dasarathy J, Allende DS, Caballeria J, Sancho-Bru P, McClain CJ, Mitchell M, McCullough AJ, Radaeva S, Barton B, Szabo G, Dasarathy S, Nagy LE. Differential role of MLKL in alcohol-associated and non-alcohol-associated fatty liver diseases in mice and humans. JCI Insight 2021; 6:140180. [PMID: 33616081 PMCID: PMC7934930 DOI: 10.1172/jci.insight.140180] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 01/07/2021] [Indexed: 01/12/2023] Open
Abstract
Hepatocellular death contributes to progression of alcohol-associated (ALD-associated) and non-alcohol-associated (NAFL/NASH) liver diseases. However, receptor-interaction protein kinase 3 (RIP3), an intermediate in necroptotic cell death, contributes to injury in murine models of ALD but not NAFL/NASH. We show here that a differential role for mixed-lineage kinase domain-like protein (MLKL), the downstream effector of RIP3, in murine models of ALD versus NAFL/NASH and that RIP1-RIP3-MLKL can be used as biomarkers to distinguish alcohol-associated hepatitis (AH) from NASH. Phospho-MLKL was higher in livers of patients with NASH compared with AH or healthy controls (HCs). MLKL expression, phosphorylation, oligomerization, and translocation to plasma membrane were induced in WT mice fed diets high in fat, fructose, and cholesterol but not in response to Gao-binge (acute on chronic) ethanol exposure. Mlkl-/- mice were not protected from ethanol-induced hepatocellular injury, which was associated with increased expression of chemokines and neutrophil recruitment. Circulating concentrations of RIP1 and RIP3, but not MLKL, distinguished patients with AH from HCs or patients with NASH. Taken together, these data indicate that MLKL is differentially activated in ALD/AH compared with NAFL/NASH in both murine models and patients. Furthermore, plasma RIP1 and RIP3 may be promising biomarkers for distinguishing AH and NASH.
Collapse
Affiliation(s)
- Tatsunori Miyata
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Gastroenterological Surgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Xiaoqin Wu
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Xiude Fan
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Emily Huang
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Carlos Sanz-Garcia
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Sanjoy Roychowdhury
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine and
| | - Annette Bellar
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Megan R. McMullen
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jaividhya Dasarathy
- Department of Family Medicine, Metro Health Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Joan Caballeria
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Pau Sancho-Bru
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Craig J. McClain
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Mack Mitchell
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Arthur J. McCullough
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Svetlana Radaeva
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Bruce Barton
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Srinivasan Dasarathy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine and
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Laura E. Nagy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine and
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
27
|
Samson AL, Fitzgibbon C, Patel KM, Hildebrand JM, Whitehead LW, Rimes JS, Jacobsen AV, Horne CR, Gavin XJ, Young SN, Rogers KL, Hawkins ED, Murphy JM. A toolbox for imaging RIPK1, RIPK3, and MLKL in mouse and human cells. Cell Death Differ 2021; 28:2126-2144. [PMID: 33589776 DOI: 10.1038/s41418-021-00742-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/20/2021] [Accepted: 01/25/2021] [Indexed: 12/18/2022] Open
Abstract
Necroptosis is a lytic, inflammatory cell death pathway that is dysregulated in many human pathologies. The pathway is executed by a core machinery comprising the RIPK1 and RIPK3 kinases, which assemble into necrosomes in the cytoplasm, and the terminal effector pseudokinase, MLKL. RIPK3-mediated phosphorylation of MLKL induces oligomerization and translocation to the plasma membrane where MLKL accumulates as hotspots and perturbs the lipid bilayer to cause death. The precise choreography of events in the pathway, where they occur within cells, and pathway differences between species, are of immense interest. However, they have been poorly characterized due to a dearth of validated antibodies for microscopy studies. Here, we describe a toolbox of antibodies for immunofluorescent detection of the core necroptosis effectors, RIPK1, RIPK3, and MLKL, and their phosphorylated forms, in human and mouse cells. By comparing reactivity with endogenous proteins in wild-type cells and knockout controls in basal and necroptosis-inducing conditions, we characterise the specificity of frequently-used commercial and recently-developed antibodies for detection of necroptosis signaling events. Importantly, our findings demonstrate that not all frequently-used antibodies are suitable for monitoring necroptosis by immunofluorescence microscopy, and methanol- is preferable to paraformaldehyde-fixation for robust detection of specific RIPK1, RIPK3, and MLKL signals.
Collapse
Affiliation(s)
- André L Samson
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| | - Cheree Fitzgibbon
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Komal M Patel
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Joanne M Hildebrand
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Lachlan W Whitehead
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Joel S Rimes
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Annette V Jacobsen
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Christopher R Horne
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Xavier J Gavin
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Samuel N Young
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Kelly L Rogers
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Edwin D Hawkins
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
28
|
Tian RD, Chen YQ, He YH, Tang YJ, Chen GM, Yang FW, Li Y, Huang WG, Chen H, Liu X, Lin SD. Phosphorylation of eIF2α mitigates endoplasmic reticulum stress and hepatocyte necroptosis in acute liver injury. Ann Hepatol 2021; 19:79-87. [PMID: 31548168 DOI: 10.1016/j.aohep.2019.05.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/26/2019] [Accepted: 05/27/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION AND OBJECTIVES Necroptosis and endoplasmic reticulum (ER) stress has been implicated in acute and chronic liver injury. Activated eukaryotic initiation factor 2 alpha (eIF2α) attenuates protein synthesis and relieves the load of protein folding in the ER. In this study, we aimed to analyze the impact of eIF2α phosphorylation on hepatocyte necroptosis in acute liver injury. MATERIALS AND METHODS Male BALB/c mice were injected with tunicamycin or d-galactosamine, and LO2 cells were incubated with tunicamycin to induce acute liver injury. 4-Phenylbutyric acid (PBA) and salubrinal were used to inhibit ER stress and eIF2α dephosphorylation, respectively. We analyzed the eIF2α phosphorylation, ER stress, and hepatocyte necroptosis in mice and cells model. RESULTS Tunicamycin or d-galactosamine significantly induced ER stress and necroptosis, as well as eIF2α phosphorylation, in mice and LO2 cells (p<0.05). ER stress aggravated tunicamycin-induced hepatocyte necroptosis in mice and LO2 cells (p<0.05). Elevated eIF2α phosphorylation significantly mitigated hepatocyte ER stress (p<0.05) and hepatocyte necroptosis in mice (34.37±3.39% vs 22.53±2.18%; p<0.05) and LO2 cells (1±0.11 vs 0.33±0.05; p<0.05). Interestingly, tumor necrosis factor receptor (TNFR) 1 protein levels were not completely synchronized with necroptosis. TNFR1 expression was reduced in d-galactosamine-treated mice (p<0.05) and cells incubated with tunicamycin for 12 and 24h (p<0.05). ER stress partially restored TNFR1 expression and increased necroptosis in tunicamycin-incubated cells (p<0.05). CONCLUSIONS These results imply that ER stress can mediate hepatocyte necroptosis independent of TNFR1 signaling and elevated eIF2α phosphorylation can mitigate ER stress during acute liver injury.
Collapse
Affiliation(s)
- Ren-Dong Tian
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| | - Yi-Qun Chen
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| | - Yi-Huai He
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China.
| | - Yong-Jing Tang
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| | - Gui-Mei Chen
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| | - Fang-Wan Yang
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| | - Ying Li
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| | - Wen-Ge Huang
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| | - Huan Chen
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| | - Xia Liu
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| | - Shi-De Lin
- Department of Infectious Diseases, the Affiliated Hospital of Zunyi Medical College, ZunyiGuizhou, China
| |
Collapse
|
29
|
Inhibition of MLKL Attenuates Necroptotic Cell Death in a Murine Cell Model of Ischaemia Injury. J Clin Med 2021; 10:jcm10020212. [PMID: 33435617 PMCID: PMC7826539 DOI: 10.3390/jcm10020212] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/01/2021] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Steatosis in donor livers poses a major risk of organ dysfunction due to their susceptibility to ischaemia-reperfusion (I/R) injury during transplant. Necroptosis, a novel form of programmed cell death, is orchestrated by receptor-interacting protein kinase 1 (RIPK1), receptor-interacting protein kinase 3 (RIPK3) and mixed-lineage kinase domain-like pseudokinase (MLKL), has been implicated in I/R injury. Here we investigated the mechanisms of cell death pathways in an in vitro model of hepato-steatotic ischaemia. METHODS Free fatty acid (FFA) treated alpha mouse liver 12 (AML-12) cells were incubated in oxygen-glucose-deprivation (OGD) conditions as seen during ischaemia. RESULTS We found that OGD triggered upregulation of insoluble fraction of RIPK3 and MLKL in FFA + OGD cells compared to FFA control cells. We report that intervention with small interfering (si) MLKL and siRIPK3 significantly attenuated cell death in FFA + OGD cells. Absence of activated CASPASE8 and cleaved-CASPASE3, no change in the expression of CASPASE1 and prostaglandin-endoperoxide synthase 2 (Ptgs2) in FFA + OGD treated cells compared to FFA control cells indicated that apoptosis, pyroptosis and ferroptosis, respectively, are unlikely to be active in this model. CONCLUSION Our findings indicate that RIPK3-MLKL dependent necroptosis contributed to cell death in our in vitro model. Both MLKL and RIPK3 are promising therapeutic targets to inhibit necroptosis during ischaemic injury in fatty liver.
Collapse
|
30
|
Shojaie L, Iorga A, Dara L. Cell Death in Liver Diseases: A Review. Int J Mol Sci 2020; 21:ijms21249682. [PMID: 33353156 PMCID: PMC7766597 DOI: 10.3390/ijms21249682] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Regulated cell death (RCD) is pivotal in directing the severity and outcome of liver injury. Hepatocyte cell death is a critical event in the progression of liver disease due to resultant inflammation leading to fibrosis. Apoptosis, necrosis, necroptosis, autophagy, and recently, pyroptosis and ferroptosis, have all been investigated in the pathogenesis of various liver diseases. These cell death subroutines display distinct features, while sharing many similar characteristics with considerable overlap and crosstalk. Multiple types of cell death modes can likely coexist, and the death of different liver cell populations may contribute to liver injury in each type of disease. This review addresses the known signaling cascades in each cell death pathway and its implications in liver disease. In this review, we describe the common findings in each disease model, as well as the controversies and the limitations of current data with a particular focus on cell death-related research in humans and in rodent models of alcoholic liver disease, non-alcoholic fatty liver disease and steatohepatitis (NASH/NAFLD), acetaminophen (APAP)-induced hepatotoxicity, autoimmune hepatitis, cholestatic liver disease, and viral hepatitis.
Collapse
Affiliation(s)
- Layla Shojaie
- Division of Gastrointestinal & Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (L.S.); (A.I.)
- Research Center for Liver Disease, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Andrea Iorga
- Division of Gastrointestinal & Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (L.S.); (A.I.)
- Research Center for Liver Disease, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Lily Dara
- Division of Gastrointestinal & Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (L.S.); (A.I.)
- Research Center for Liver Disease, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Correspondence:
| |
Collapse
|
31
|
Immunological mechanisms and therapeutic targets of fatty liver diseases. Cell Mol Immunol 2020; 18:73-91. [PMID: 33268887 PMCID: PMC7852578 DOI: 10.1038/s41423-020-00579-3] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD) are the two major types of chronic liver disease worldwide. Inflammatory processes play key roles in the pathogeneses of fatty liver diseases, and continuous inflammation promotes the progression of alcoholic steatohepatitis (ASH) and nonalcoholic steatohepatitis (NASH). Although both ALD and NAFLD are closely related to inflammation, their respective developmental mechanisms differ to some extent. Here, we review the roles of multiple immunological mechanisms and therapeutic targets related to the inflammation associated with fatty liver diseases and the differences in the progression of ASH and NASH. Multiple cell types in the liver, including macrophages, neutrophils, other immune cell types and hepatocytes, are involved in fatty liver disease inflammation. In addition, microRNAs (miRNAs), extracellular vesicles (EVs), and complement also contribute to the inflammatory process, as does intertissue crosstalk between the liver and the intestine, adipose tissue, and the nervous system. We point out that inflammation also plays important roles in promoting liver repair and controlling bacterial infections. Understanding the complex regulatory process of disrupted homeostasis during the development of fatty liver diseases may lead to the development of improved targeted therapeutic intervention strategies.
Collapse
|
32
|
Wang M, Chen WY, Zhang J, Gobejishvili L, Barve SS, McClain CJ, Joshi-Barve S. Elevated Fructose and Uric Acid Through Aldose Reductase Contribute to Experimental and Human Alcoholic Liver Disease. Hepatology 2020; 72:1617-1637. [PMID: 32086945 DOI: 10.1002/hep.31197] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Alcohol-associated liver disease (ALD) is a common chronic liver disease worldwide with high morbidity and mortality, and no Food and Drug Administration-approved therapies. Fructose (dietary or endogenous), its metabolite uric acid, and aldose reductase (AR, the only endogenous enzyme that produces fructose) are strongly associated with the development of nonalcoholic fatty liver disease. However, the role of AR or its metabolites in ALD remains understudied and was examined using human specimens, cultured cells, and mouse model systems. APPROACH AND RESULTS We demonstrated in liver specimens from patients with alcoholic hepatitis, the AR up-regulation and elevated AR metabolites (sorbitol, fructose, and uric acid), which correlated significantly with (1) increased lipid peroxidation byproducts and endoplasmic reticulum (ER) stress, (2) decreased protective ER chaperones, and (3) greater cell death and liver injury. Furthermore, we established a causal role for AR in ALD by showing that the genetic deficiency of AR (knockout mice) prevented alcohol-induced increase in harmful AR metabolites, toxic aldehydes, steatosis, ER stress, apoptosis, and liver injury. Finally, we demonstrated the therapeutic potential of pharmacological AR inhibition against alcohol-induced hepatic injury in experimental ALD. CONCLUSIONS Our data demonstrate that hepatic AR up-regulation, and consequent elevation in fructose, sorbitol and/or uric acid, are important factors contributing to alcohol-induced steatosis, ER stress, apoptosis, and liver injury in both experimental and human ALD. Our study provides a strong rationale to evaluate AR as a potential therapeutic target and to test AR inhibitors to ameliorate alcohol-induced liver injury.
Collapse
Affiliation(s)
- Min Wang
- Department of Medicine, University of Louisville, Louisville, KY.,Alcohol Research Center, University of Louisville, Louisville, KY.,Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY
| | - Wei-Yang Chen
- Alcohol Research Center, University of Louisville, Louisville, KY.,Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY
| | - Jingwen Zhang
- Department of Medicine, University of Louisville, Louisville, KY.,Alcohol Research Center, University of Louisville, Louisville, KY.,Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY
| | - Leila Gobejishvili
- Department of Medicine, University of Louisville, Louisville, KY.,Alcohol Research Center, University of Louisville, Louisville, KY.,Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY.,Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY
| | - Shirish S Barve
- Department of Medicine, University of Louisville, Louisville, KY.,Alcohol Research Center, University of Louisville, Louisville, KY.,Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY.,Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY
| | - Craig J McClain
- Department of Medicine, University of Louisville, Louisville, KY.,Alcohol Research Center, University of Louisville, Louisville, KY.,Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY.,Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY.,Robley Rex VAMC, Louisville, KY
| | - Swati Joshi-Barve
- Department of Medicine, University of Louisville, Louisville, KY.,Alcohol Research Center, University of Louisville, Louisville, KY.,Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY.,Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY
| |
Collapse
|
33
|
Miyata T, Nagy LE. Programmed cell death in alcohol-associated liver disease. Clin Mol Hepatol 2020; 26:618-625. [PMID: 32951412 PMCID: PMC7641549 DOI: 10.3350/cmh.2020.0142] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Alcohol-associated liver disease (ALD), which ranges from mild disease to alcohol-associated hepatitis and cirrhosis, is the most prevalent type of chronic liver disease and a leading cause of morbidity and mortality worldwide. Accumulating evidence reveals that programmed cell death (PCD) plays a crucial role in progression of ALD involving crosstalk between hepatocytes and immune cells. Multiple pathways of PCD, including apoptosis, necroptosis, autophagy, pyroptosis and ferroptosis, are reported in ALD. Interestingly, PCD pathways are intimately linked and interdependent, making it difficult to therapeutically target a single pathway. This review clarifies the multiple types of PCD occurring in liver and focuses on crosstalk between hepatocytes and innate immune cells in ALD.
Collapse
Affiliation(s)
- Tatsunori Miyata
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA.,Department of Gastroenterological Surgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Laura E Nagy
- Northern Ohio Alcohol Center, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA.,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, USA.,Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
34
|
Wu X, Poulsen KL, Sanz-Garcia C, Huang E, McMullen MR, Roychowdhury S, Dasarathy S, Nagy LE. MLKL-dependent signaling regulates autophagic flux in a murine model of non-alcohol-associated fatty liver and steatohepatitis. J Hepatol 2020; 73:616-627. [PMID: 32220583 PMCID: PMC7438259 DOI: 10.1016/j.jhep.2020.03.023] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS Autophagy maintains cellular homeostasis and plays a critical role in the development of non-alcoholic fatty liver and steatohepatitis. The pseudokinase mixed lineage kinase domain-like (MLKL) is a key downstream effector of receptor interacting protein kinase 3 (RIP3) in the necroptotic pathway of programmed cell death. However, recent data reveal that MLKL also regulates autophagy. Herein, we tested the hypothesis that MLKL contributes to the progression of Western diet-induced liver injury in mice by regulating autophagy. METHODS Rip3+/+, Rip3-/-, Mlkl+/+ and Mlkl-/- mice were fed a Western diet (FFC diet, high in fat, fructose and cholesterol) or chow for 12 weeks. AML12 and primary mouse hepatocytes were exposed to palmitic acid (PA). RESULTS The FFC diet increased expression, phosphorylation and oligomerization of MLKL in the liver. Mlkl, but not Rip3, deficiency protected mice from FFC diet-induced liver injury. The FFC diet also induced accumulation of p62 and LC3-II, as well as markers of endoplasmic reticulum stress, in Mlkl+/+ but not Mlkl-/- mice. Mlkl deficiency in mice also prevented the inhibition of autophagy by a protease inhibitor, leupeptin. Using an mRFP-GFP-LC3 reporter in cultured hepatocytes revealed that PA blocked the fusion of autophagosomes with lysosomes. PA triggered MLKL expression and translocation, first to autophagosomes and then to the plasma membrane, independently of Rip3. Mlkl, but not Rip3, deficiency prevented inhibition of autophagy in PA-treated hepatocytes. Overexpression of Mlkl blocked autophagy independently of PA. Additionally, pharmacologic inhibition of autophagy induced MLKL expression and translocation to the plasma membrane in hepatocytes. CONCLUSIONS Taken together, these data indicate that MLKL-dependent, but RIP3-independent, signaling contributes to FFC diet-induced liver injury by inhibiting autophagy. LAY SUMMARY Autophagy is a regulated process that maintains cellular homeostasis. Impaired autophagy contributes to cell injury and death, thus playing a critical role in the pathogenesis of a number of diseases, including non-alcohol-associated fatty liver and steatohepatitis. Herein, we show that Mlkl-dependent, but Rip3-independent, signaling contributed to diet-induced liver injury and inflammatory responses by inhibiting autophagy. These data identify a novel co-regulatory mechanism between necroptotic and autophagic signaling pathways in non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Xiaoqin Wu
- Center for Liver Disease Research, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Kyle L. Poulsen
- Center for Liver Disease Research, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Carlos Sanz-Garcia
- Center for Liver Disease Research, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Emily Huang
- Center for Liver Disease Research, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Megan R. McMullen
- Center for Liver Disease Research, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States
| | - Sanjoy Roychowdhury
- Center for Liver Disease Research, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States;,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Srinivasan Dasarathy
- Center for Liver Disease Research, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States;,Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, United States;,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Laura E. Nagy
- Center for Liver Disease Research, Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, United States;,Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, United States;,Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States,Corresponding author. Address: Cleveland Clinic, Lerner Research Institute/NE40 9500 Euclid Ave, Cleveland, OH 44195. Tel.: 216-444-4021; Fax: 216-636-1493. (L.E. Nagy)
| |
Collapse
|
35
|
Baidya R, Crawford DHG, Gautheron J, Wang H, Bridle KR. Necroptosis in Hepatosteatotic Ischaemia-Reperfusion Injury. Int J Mol Sci 2020; 21:ijms21165931. [PMID: 32824744 PMCID: PMC7460692 DOI: 10.3390/ijms21165931] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
While liver transplantation remains the sole treatment option for patients with end-stage liver disease, there are numerous limitations to liver transplantation including the scarcity of donor livers and a rise in livers that are unsuitable to transplant such as those with excess steatosis. Fatty livers are susceptible to ischaemia-reperfusion (IR) injury during transplantation and IR injury results in primary graft non-function, graft failure and mortality. Recent studies have described new cell death pathways which differ from the traditional apoptotic pathway. Necroptosis, a regulated form of cell death, has been associated with hepatic IR injury. Receptor-interacting protein kinase 3 (RIPK3) and mixed-lineage kinase domain-like pseudokinase (MLKL) are thought to be instrumental in the execution of necroptosis. The study of hepatic necroptosis and potential therapeutic approaches to attenuate IR injury will be a key factor in improving our knowledge regarding liver transplantation with fatty donor livers. In this review, we focus on the effect of hepatic steatosis during liver transplantation as well as molecular mechanisms of necroptosis and its involvement during liver IR injury. We also discuss the immune responses triggered during necroptosis and examine the utility of necroptosis inhibitors as potential therapeutic approaches to alleviate IR injury.
Collapse
Affiliation(s)
- Raji Baidya
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland QLD 4006, Australia; (R.B.); (D.H.G.C.)
- Gallipoli Medical Research Institute, Brisbane, Queensland QLD 4120, Australia;
| | - Darrell H. G. Crawford
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland QLD 4006, Australia; (R.B.); (D.H.G.C.)
- Gallipoli Medical Research Institute, Brisbane, Queensland QLD 4120, Australia;
| | - Jérémie Gautheron
- Sorbonne University, Inserm, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France;
- Institute of Cardiometabolism and Nutrition (ICAN), 75013 Paris, France
| | - Haolu Wang
- Gallipoli Medical Research Institute, Brisbane, Queensland QLD 4120, Australia;
- Diamantina Institute, The University of Queensland, Brisbane, Queensland QLD 4102, Australia
| | - Kim R. Bridle
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland QLD 4006, Australia; (R.B.); (D.H.G.C.)
- Gallipoli Medical Research Institute, Brisbane, Queensland QLD 4120, Australia;
- Correspondence: ; Tel.: +61-7-3346-0698
| |
Collapse
|
36
|
Gautheron J, Gores GJ, Rodrigues CMP. Lytic cell death in metabolic liver disease. J Hepatol 2020; 73:394-408. [PMID: 32298766 PMCID: PMC7371520 DOI: 10.1016/j.jhep.2020.04.001] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 03/02/2020] [Accepted: 04/01/2020] [Indexed: 12/11/2022]
Abstract
Regulated cell death is intrinsically associated with inflammatory liver disease and is pivotal in governing outcomes of metabolic liver disease. Different types of cell death may coexist as metabolic liver disease progresses to inflammation, fibrosis, and ultimately cirrhosis. In addition to apoptosis, lytic forms of hepatocellular death, such as necroptosis, pyroptosis and ferroptosis elicit strong inflammatory responses due to cell membrane permeabilisation and release of cellular components, contributing to the recruitment of immune cells and activation of hepatic stellate cells. The control of liver cell death is of fundamental importance and presents novel opportunities for potential therapeutic intervention. This review summarises the underlying mechanism of distinct lytic cell death modes and their commonalities, discusses their relevance to metabolic liver diseases of different aetiologies, and acknowledges the limitations of current knowledge in the field. We focus on the role of hepatocyte necroptosis, pyroptosis and ferroptosis in non-alcoholic fatty liver disease, alcohol-associated liver disease and other metabolic liver disorders, as well as potential therapeutic implications.
Collapse
Affiliation(s)
- Jérémie Gautheron
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Paris, France; Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
37
|
Samir P, Malireddi RKS, Kanneganti TD. The PANoptosome: A Deadly Protein Complex Driving Pyroptosis, Apoptosis, and Necroptosis (PANoptosis). Front Cell Infect Microbiol 2020; 10:238. [PMID: 32582562 PMCID: PMC7283380 DOI: 10.3389/fcimb.2020.00238] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/24/2020] [Indexed: 01/05/2023] Open
Abstract
Programmed cell death is regulated by evolutionarily conserved pathways that play critical roles in development and the immune response. A newly recognized pathway for proinflammatory programmed cell death called PANoptosis is controlled by a recently identified cytoplasmic multimeric protein complex named the PANoptosome. The PANoptosome can engage, in parallel, three key modes of programmed cell death—pyroptosis, apoptosis, and necroptosis. The PANoptosome components have been implicated in a wide array of human diseases including autoinflammatory diseases, neurodegenerative diseases, cancer, microbial infections, and metabolic diseases. Here, we review putative components of the PANoptosome and present a phylogenetic analysis of their molecular domains and interaction motifs that support complex assembly. We also discuss genetic data that suggest PANoptosis is coordinated by scaffolding and catalytic functions of the complex components and propose mechanistic models for PANoptosome assembly. Overall, this review presents potential mechanisms governing PANoptosis based on evolutionary analysis of the PANoptosome components.
Collapse
Affiliation(s)
- Parimal Samir
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | | |
Collapse
|
38
|
Sarcognato S, de Jong IEM, Fabris L, Cadamuro M, Guido M. Necroptosis in Cholangiocarcinoma. Cells 2020; 9:cells9040982. [PMID: 32326539 PMCID: PMC7226990 DOI: 10.3390/cells9040982] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/19/2022] Open
Abstract
Necroptosis is a type of regulated cell death that is increasingly being recognized as a relevant pathway in different pathological conditions. Necroptosis can occur in response to multiple stimuli, is triggered by the activation of death receptors, and is regulated by receptor-interacting protein kinases 1 and 3 and mixed-lineage kinase domain-like, which form a regulatory complex called the necrosome. Accumulating evidence suggests that necroptosis plays a complex role in cancer, which is likely context-dependent and can vary among different types of neoplasms. Necroptosis serves as an alternative mode of programmed cell death overcoming apoptosis and, as a pro-inflammatory death type, it may inhibit tumor progression by releasing damage-associated molecular patterns to elicit robust cross-priming of anti-tumor CD8+ T cells. The development of therapeutic strategies triggering necroptosis shows great potential for anti-cancer therapy. In this review, we summarize the current knowledge on necroptosis and its role in liver biliary neoplasms, underlying the potential of targeting necroptosis components for cancer treatment.
Collapse
Affiliation(s)
- Samantha Sarcognato
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, 31100 Treviso, Italy
| | - Iris E. M. de Jong
- Department of Surgery, Section of Hepatobiliary Surgery and Liver Transplantation, University Medical Center Groningen, 9700 Groningen, The Netherlands
| | - Luca Fabris
- Department of Molecular Medicine—DMM, University of Padova, 35121 Padova, Italy
| | | | - Maria Guido
- Department of Pathology, Azienda ULSS2 Marca Trevigiana, 31100 Treviso, Italy
- Department of Medicine—DIMED, University of Padova, 35121 Padova, Italy
- Correspondence: ; Tel.: +39-0422-322750
| |
Collapse
|
39
|
Fahim M, Rafiee Zadeh A, Shoureshi P, Ghadimi K, Cheshmavar M, Sheikhinia N, Afzali M. Alcohol and multiple sclerosis: an immune system-based review. INTERNATIONAL JOURNAL OF PHYSIOLOGY, PATHOPHYSIOLOGY AND PHARMACOLOGY 2020; 12:58-69. [PMID: 32419901 PMCID: PMC7218739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/02/2020] [Indexed: 06/11/2023]
Abstract
Multiple sclerosis is a chronic inflammatory disease of the central nervous system (CNS). Although the exact etiology of multiple sclerosis is unknown, researchers suggest that genetic, environmental, and microbial factors play a central role in causing multiple sclerosis. Pathology of multiple sclerosis is based on inflammation as T cells enter the brain via disruptions in the blood-brain barrier, recognizing myelin as foreign antigen; and as a result, the T cells attack myelin and start the inflammatory processes, enhancing inflammatory cytokines and antibodies. Since previous studies show ethanol can suppress the immune system such as innate, humoral, and cellular immunity and increases the production of anti-inflammatory cytokines, we hypothesized maybe ethanol also have ameliorating effects on multiple sclerosis symptoms. Although alcohol induces apoptosis in oligodendrocytes and neurons, causing demyelination and affects CNS directly, in this study we will investigate ethanol's effects on some aspects of the immune system in multiple sclerosis.
Collapse
Affiliation(s)
- Maryam Fahim
- School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
| | | | - Pouria Shoureshi
- Department of Internal Medicine, Orange Park Medical CenterFlorida, USA
| | - Keyvan Ghadimi
- School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
| | - Masoumeh Cheshmavar
- Department of Neurology, School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
| | - Neda Sheikhinia
- Department of Neurology, School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
| | - Mahdieh Afzali
- Department of Neurology, School of Medicine, Isfahan University of Medical SciencesIsfahan, Iran
| |
Collapse
|
40
|
Heib M, Rose-John S, Adam D. Necroptosis, ADAM proteases and intestinal (dys)function. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 353:83-152. [PMID: 32381179 DOI: 10.1016/bs.ircmb.2020.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Recently, an unexpected connection between necroptosis and members of the a disintegrin and metalloproteinase (ADAM) protease family has been reported. Necroptosis represents an important cell death routine which helps to protect from viral, bacterial, fungal and parasitic infections, maintains adult T cell homeostasis and contributes to the elimination of potentially defective organisms before parturition. Equally important for organismal homeostasis, ADAM proteases control cellular processes such as development and differentiation, immune responses or tissue regeneration. Notably, necroptosis as well as ADAM proteases have been implicated in the control of inflammatory responses in the intestine. In this review, we therefore provide an overview of the physiology and pathophysiology of necroptosis, ADAM proteases and intestinal (dys)function, discuss the contribution of necroptosis and ADAMs to intestinal (dys)function, and review the current knowledge on the role of ADAMs in necroptotic signaling.
Collapse
Affiliation(s)
- Michelle Heib
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Stefan Rose-John
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Dieter Adam
- Institut für Immunologie, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| |
Collapse
|
41
|
Contreras A, Morales L, Del Olmo N, Pérez-García C. Effects of Intermittent versus Chronic-Moderate Ethanol Administration during Adolescence in the Adult Hippocampal Phosphoproteome. Chem Res Toxicol 2020; 33:448-460. [PMID: 31944673 DOI: 10.1021/acs.chemrestox.9b00359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Alcohol consumption during adolescence is known to cause different impairments in the hippocampus that could lead to persistent deficits in adulthood. A common pattern of alcohol use in adolescents consists of excessive and intermittent alcohol consumption over a very short period of time (binge drinking). Protein phosphorylation is a mechanism underlying memory processes and we have previously demonstrated changes in the rat hippocampal phosphoproteome after a single dose of ethanol; however, studies showing the phosphoprotein alterations in the hippocampus after repeated exposition to alcohol are limited. This study focuses on the identification of the phosphoproteins differentially regulated in the adolescent rat hippocampus after repeated ethanol administration by comparing different patterns of alcohol treatments according to dose and frequency of administration ((i) moderate dose-chronic use, (ii) low dose-intermittent use, and (iii) high dose-intermittent use). We have used a proteomic approach, including phosphoprotein enrichment by immobilized metal affinity chromatography, which revealed 21 proteins differentially affected depending on the pattern of alcohol treatment used. Many of these proteins are included in glycolysis and glucagon signaling pathways and are also involved in neurodegeneration, which could reinforce the role of metabolic alterations in the neural damage induced by repeated alcohol exposure during adolescence.
Collapse
Affiliation(s)
- Ana Contreras
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia , Universidad CEU-San Pablo , Madrid 28003 , Spain
| | - Lidia Morales
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia , Universidad CEU-San Pablo , Madrid 28003 , Spain
| | - Nuria Del Olmo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia , Universidad CEU-San Pablo , Madrid 28003 , Spain
| | - Carmen Pérez-García
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia , Universidad CEU-San Pablo , Madrid 28003 , Spain
| |
Collapse
|
42
|
Khoury MK, Gupta K, Franco SR, Liu B. Necroptosis in the Pathophysiology of Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:272-285. [PMID: 31783008 PMCID: PMC6983729 DOI: 10.1016/j.ajpath.2019.10.012] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/17/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022]
Abstract
Over the past 15 years, elegant studies have demonstrated that in certain conditions, programed cell death resembles necrosis and depends on a unique molecular pathway with no overlap with apoptosis. This form of regulated necrosis is represented by necroptosis, in which the receptor-interacting protein kinase-3 and its substrate mixed-lineage kinase domain-like protein play a crucial role. With the development of knockout mouse models and molecular inhibitors unique to necroptotic proteins, this cell death has been found to occur in virtually all tissues and diseases evaluated. There are different immunologic consequences depending on whether cells die through apoptosis or necroptosis. Therefore, distinguishing between these two forms of cell death may be crucial during pathologic evaluations. In this review, we provide an understanding of necroptotic cell-death and highlight diseases in which necroptosis has been found to play a role. We also discuss the inhibitors of necroptosis and the ways these inhibitors have been used in preclinical models of diseases. These two discussions offer an understanding of the role of necroptosis in diseases and will foster efforts to pharmacologically target this unique yet pervasive form of programed cell death in the clinic.
Collapse
Affiliation(s)
- Mitri K Khoury
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kartik Gupta
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sarah R Franco
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin
| | - Bo Liu
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
43
|
Li Y, Chao X, Wang S, Williams JA, Ni HM, Ding WX. Role of Mechanistic Target of Rapamycin and Autophagy in Alcohol-Induced Adipose Atrophy and Liver Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:158-175. [PMID: 31733185 PMCID: PMC6940593 DOI: 10.1016/j.ajpath.2019.09.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 09/19/2019] [Accepted: 09/26/2019] [Indexed: 01/07/2023]
Abstract
Chronic alcohol consumption induces adipose tissue atrophy. However, the mechanisms for how alcohol induces lipodystrophy and its impact on liver steatosis and injury are not fully elucidated. Autophagy is a highly conserved lysosomal degradation pathway, which regulates cellular homeostasis. Mice with autophagy deficiency in adipose tissue have impaired adipogenesis. However, whether autophagy plays a role in alcohol-induced adipose atrophy and how altered adipocyte autophagy contributes to alcohol-induced liver injury remain unclear. To determine the role of adipose autophagy and mechanistic target of rapamycin (mTOR) in alcohol-induced adipose and liver pathogenesis, we generated adipocyte-specific Atg5 knockout (KO), adipocyte-specific mTOR KO, adipocyte-specific Raptor KO, and adipocyte-specific tuberous sclerosis complex 1 KO mice by crossing floxed mice with Adipoq-Cre. The KO mice and their matched wild-type mice were challenged with chronic-plus-binge alcohol mouse model. Chronic-plus-binge alcohol induced adipose atrophy with increased autophagy and decreased Akt/mTOR signaling in epididymal adipose tissue in wild-type mice. Adipocyte-specific Raptor KO mice experienced exacerbated alcohol-induced steatosis, but neither adipocyte-specific mTOR nor adipocyte-specific tuberous sclerosis complex 1 KO mice exhibited similar detrimental effects. Adipocyte-specific Atg5 KO mice had increased circulating levels of fibroblast growth factor 21 and adiponectin and were resistant to alcohol-induced adipose atrophy and liver injury. In conclusion, autophagy deficiency in adipose tissue leads to reduced sensitivity to alcohol-induced adipose atrophy, which ameliorates alcohol-induced liver injury in mice.
Collapse
Affiliation(s)
- Yuan Li
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Shaogui Wang
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Jessica A Williams
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
44
|
Sun H, McKeen T, Wang H, Ni HM. Necroptosis in ischemia-reperfusion injury of lean and steatotic livers. LIVER RESEARCH 2019. [DOI: 10.1016/j.livres.2019.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
45
|
Rodriguez WE, Wahlang B, Wang Y, Zhang J, Vadhanam MV, Joshi-Barve S, Bauer P, Cannon R, Ahmadi AR, Sun Z, Cameron A, Barve S, Maldonado C, McClain C, Gobejishvili L. Phosphodiesterase 4 Inhibition as a Therapeutic Target for Alcoholic Liver Disease: From Bedside to Bench. Hepatology 2019; 70:1958-1971. [PMID: 31081957 PMCID: PMC6851418 DOI: 10.1002/hep.30761] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/30/2019] [Indexed: 12/11/2022]
Abstract
Alcoholic liver disease (ALD) is a major cause of liver-related mortality. There is still no US Food and Drug Administration-approved therapy for ALD, and therefore, identifying therapeutic targets is needed. Our previous work demonstrated that ethanol exposure leads to up-regulation of cAMP-degrading phosphodiesterase 4 (PDE4) expression, which compromises normal cAMP signaling in monocytes/macrophages and hepatocytes. This effect of ethanol on cAMP signaling contributes to dysregulated inflammatory response and altered lipid metabolism. It is unknown whether chronic alcohol consumption in humans alters hepatic PDE4 expression and cAMP signaling and whether inadequate cAMP signaling plays a pathogenic role in alcohol-induced liver injury. Our present work shows that expression of the PDE4 subfamily of enzymes is significantly up-regulated and cAMP levels are markedly decreased in hepatic tissues of patients with severe ALD. We also demonstrate the anti-inflammatory efficacy of roflumilast, a clinically available PDE4 inhibitor, on endotoxin-inducible proinflammatory cytokine production ex vivo in whole blood of patients with alcoholic hepatitis. Moreover, we demonstrate that ethanol-mediated changes in hepatic PDE4 and cAMP levels play a causal role in liver injury in in vivo and in vitro models of ALD. This study employs a drug delivery system that specifically delivers the PDE4 inhibitor rolipram to the liver to avoid central nervous system side effects associated with this drug. Our results show that PDE4 inhibition significantly attenuates ethanol-induced hepatic steatosis and injury through multiple mechanisms, including reduced oxidative and endoplasmic reticulum stress both in vivo and in vitro. Conclusion: Increased PDE4 plays a pathogenic role in the development of ALD; hence, directed interventions aimed at inhibiting PDE4 might be an effective treatment for ALD.
Collapse
Affiliation(s)
- Walter E. Rodriguez
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA,,Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA,,Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - Banrida Wahlang
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA,,Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA,,Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - Yali Wang
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA,,Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA,,Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - Jingwen Zhang
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA,,Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA,,Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - Manicka V. Vadhanam
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA,,Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA,,Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA
| | - Swati Joshi-Barve
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA,,Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA,,Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA,,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Kentucky, USA
| | - Philip Bauer
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA,,EndoProtech, Inc., Louisville, Kentucky, USA
| | - Robert Cannon
- Department of Surgery, School of Medicine, University of Louisville, Kentucky, USA
| | - Ali Reza Ahmadi
- Department of Surgery and Transplant Biology Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhaoli Sun
- Department of Surgery and Transplant Biology Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Cameron
- Department of Surgery and Transplant Biology Research Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Shirish Barve
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA,,Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA,,Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA,,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Kentucky, USA
| | - Claudio Maldonado
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, USA,,EndoProtech, Inc., Louisville, Kentucky, USA
| | - Craig McClain
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA,,Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA,,Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA,,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Kentucky, USA,,Robley Rex VA Medical Center, Louisville, Kentucky, USA
| | - Leila Gobejishvili
- University of Louisville Alcohol Research Center, University of Louisville, Kentucky, USA,,Hepatobiology and Toxicology Center, University of Louisville, Kentucky, USA,,Department of Medicine, School of Medicine, University of Louisville, Kentucky, USA,,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Kentucky, USA
| |
Collapse
|
46
|
Necroptosis signaling in liver diseases: An update. Pharmacol Res 2019; 148:104439. [PMID: 31476369 DOI: 10.1016/j.phrs.2019.104439] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/11/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
The apoptosis alternate cell death pathways are extensively studied in recent years and their significance has been well recognized. With identification of newer cell death pathways, the therapeutic opportunities to modulate cell death have indeed further extended. Necroptosis, among other apoptosis alternate pathways, has been immensely studied recently in different hepatic disease models. Receptor-interacting protein 1 (RIPK1), RIPK3 and mixed lineage kinase domain like (MLKL) seemed to be the key players to mediate necroptosis pathway. Initially, necroptosis seemed to be following the typical pathway. But recently diverse pathways and outcomes have been observed. With recent studies reporting diverse outcomes, the necroptosis signalling has become a lot more interesting and intricate. The typical RIPK1 signalling followed by RIPK3 and MLKL might not always be strictly followed. Although, necroptosis signalling has been intensively investigated in various disease conditions; however, there is still a need to further elaborate and understand the unique scaffolding and kinase properties and other signalling interactions of necroptosis signalling molecules.
Collapse
|
47
|
Liu Y, Liu T, Lei T, Zhang D, Du S, Girani L, Qi D, Lin C, Tong R, Wang Y. RIP1/RIP3-regulated necroptosis as a target for multifaceted disease therapy (Review). Int J Mol Med 2019; 44:771-786. [PMID: 31198981 PMCID: PMC6658002 DOI: 10.3892/ijmm.2019.4244] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 06/11/2019] [Indexed: 12/27/2022] Open
Abstract
Necroptosis is a type of programmed cell death with necrotic morphology, occurring in a variety of biological processes, including inflammation, immune response, embryonic development and metabolic abnormalities. The current nomenclature defines necroptosis as cell death mediated by signal transduction from receptor‑interacting serine/threonine kinase (RIP) 1 to RIP3 (hereafter called RIP1/RIP3). However, RIP3‑dependent cell death would be a more precise definition of necroptosis. RIP3 is indispensable for necroptosis, while RIP1 is not consistently involved in the signal transduction. Notably, deletion of RIP1 even promotes RIP3‑mediated necroptosis under certain conditions. Necroptosis was previously thought as an alternate process of cell death in case of apoptosis inhibition. Currently, necroptosis is recognized to serve a pivotal role in regulating various physiological processes. Of note, it mediates a variety of human diseases, such as ischemic brain injury, immune system disorders and cancer. Targeting and inhibiting necroptosis, therefore, has the potential to be used for therapeutic purposes. To date, research has elucidated the suppression of RIP1/RIP3 via effective inhibitors and highlighted their potential application in disease therapy. The present review focused on the molecular mechanisms of RIP1/RIP3‑mediated necroptosis, explored the functions of RIP1/RIP3 in necroptosis, discussed their potential as a novel therapeutic target for disease therapy, and provided valuable suggestions for further study in this field.
Collapse
Affiliation(s)
- Yuping Liu
- Health Management Center, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Ting Liu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Tiantian Lei
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Dingding Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Suya Du
- Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Lea Girani
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Dandan Qi
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Chen Lin
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, P.R. China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| | - Yi Wang
- Health Management Center, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
48
|
Place DE, Kanneganti TD. Cell death-mediated cytokine release and its therapeutic implications. J Exp Med 2019; 216:1474-1486. [PMID: 31186281 PMCID: PMC6605758 DOI: 10.1084/jem.20181892] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/19/2022] Open
Abstract
Targeting apoptosis to treat diseases has seen tremendous success over the past decades. More recently, alternative forms of regulated cell death, including pyroptosis and necroptosis, have been described. Understanding the molecular cascades regulating both pyroptosis and necroptosis will yield even more targets to treat diseases. These lytic forms of cell death are distinct from apoptosis due to their characteristic lysis and release of cellular components that promote disease or direct a beneficial immune response. In this review, we focus on how pyroptosis and necroptosis, which release potent immune cytokines such as IL-1 and IL-18, contribute to various diseases. We also consider the important role that the executioners of these cell death pathways, GSDMD and MLKL, play in the progression of inflammatory diseases. Crosstalk between the different cell death pathways likely plays a major role physiologically. New therapeutic strategies targeting these specific molecules hold enormous potential for managing inflammatory diseases.
Collapse
Affiliation(s)
- David E Place
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN
| | | |
Collapse
|
49
|
Sanz-Garcia C, Poulsen KL, Bellos D, Wang H, McMullen MR, Li X, Chattopadhyay S, Sen G, Nagy LE. The non-transcriptional activity of IRF3 modulates hepatic immune cell populations in acute-on-chronic ethanol administration in mice. J Hepatol 2019; 70:974-984. [PMID: 30710579 PMCID: PMC6462245 DOI: 10.1016/j.jhep.2019.01.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 01/10/2019] [Accepted: 01/16/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Interferon regulatory factor 3 (IRF3) is a transcription factor mediating antiviral responses, yet recent evidence indicates that IRF3 also has critical non-transcriptional functions, including activating RIG-I-like receptors-induced IRF-3-mediated pathway of apoptosis (RIPA) and restricting activity of NF-κB. Using a novel murine model expressing only non-transcriptional IRF3 activity (Irf3S1/S1), we tested the hypothesis that non-transcriptional functions of IRF3 modulate innate immune responses in the Gao-binge (acute-on-chronic) model of alcohol-related liver disease. METHODS IRF3 and IRF3-mediated signals were analysed in liver samples from 5 patients transplanted for alcoholic hepatitis and 5 healthy controls. C57BL/6, Irf3-/- and Irf3S1/S1 mice were exposed to Gao-binge ethanol-induced liver injury. IRF3-mediated RIPA was investigated in cultured macrophages. RESULTS Phospho-IRF3 and IRF3-mediated signals were elevated in livers of patients with alcoholic hepatitis. In C57BL/6 mice, Gao-binge ethanol exposure activated IRF3 signaling and resulted in hepatocellular injury. Indicators of liver injury were differentially impacted by Irf3 genotype. Irf3-/-, but not Irf3S1/S1, mice were protected from steatosis, elevated alanine/aspartate aminotransferase levels and inflammatory cytokine expression. In contrast, neutrophil accumulation and endoplasmic reticulum stress were independent of genotype. Protection from Gao-binge injury in Irf3-/- mice was associated with an increased ratio of Ly6Clow (restorative) to Ly6Chigh (inflammatory) cells compared to C57BL/6 and Irf3S1/S1 mice. Reduced ratios of Ly6Clow/Ly6Chigh in C57BL/6 and Irf3S1/S1 mice were associated with increased apoptosis in the Ly6Clow population in response to Gao-binge. Activation of primary macrophage cultures with Poly (I:C) induced translocation of IRF3 to the mitochondria, where it associated with Bax and activated caspases 3 and 9, processes indicative of activation of the RIPA pathway. CONCLUSIONS Taken together, these data identify that the non-transcriptional function of IRF3 plays an important role in modulating the innate immune environment in response to Gao-binge ethanol exposure, via regulation of immune cell apoptosis. LAY SUMMARY Activation of the innate immune system contributes to inflammation in the progression of alcohol-related liver disease, as well as to the resolution of injury. Here we show that the protein IRF3 modulates the innate immune environment of the liver in a mouse model of alcoholic hepatitis. It does this by increasing the apoptotic cell death of immune cells that promote the resolution of injury.
Collapse
Affiliation(s)
- Carlos Sanz-Garcia
- Departments of Inflammation and Immunity, Case Western Reserve University, Cleveland, Ohio
| | - Kyle L. Poulsen
- Departments of Inflammation and Immunity, Case Western Reserve University, Cleveland, Ohio
| | - Damien Bellos
- Departments of Inflammation and Immunity, Case Western Reserve University, Cleveland, Ohio,,Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Han Wang
- Departments of Inflammation and Immunity, Case Western Reserve University, Cleveland, Ohio
| | - Megan R. McMullen
- Departments of Inflammation and Immunity, Case Western Reserve University, Cleveland, Ohio
| | - Xiaoxia Li
- Departments of Inflammation and Immunity, Case Western Reserve University, Cleveland, Ohio,,Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Saurabh Chattopadhyay
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Ganes Sen
- Departments of Inflammation and Immunity, Case Western Reserve University, Cleveland, Ohio,,Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Laura E. Nagy
- Departments of Inflammation and Immunity, Case Western Reserve University, Cleveland, Ohio,,Gastroenterology and Hepatology, Cleveland Clinic, Case Western Reserve University, Cleveland, Ohio,,Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
50
|
Ni HM, Chao X, Kaseff J, Deng F, Wang S, Shi YH, Li T, Ding WX, Jaeschke H. Receptor-Interacting Serine/Threonine-Protein Kinase 3 (RIPK3)-Mixed Lineage Kinase Domain-Like Protein (MLKL)-Mediated Necroptosis Contributes to Ischemia-Reperfusion Injury of Steatotic Livers. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1363-1374. [PMID: 31026418 DOI: 10.1016/j.ajpath.2019.03.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/12/2019] [Accepted: 03/21/2019] [Indexed: 02/07/2023]
Abstract
Increased hepatic ischemia-reperfusion (IR) injury in steatotic livers is a major reason for rejecting the use of fatty livers for liver transplantation. Necroptosis is implicated in the pathogenesis of fatty liver diseases. Necroptosis is regulated by three key proteins: receptor-interacting serine/threonine-protein kinase (RIPK)-1, RIPK3, and mixed-lineage kinase domain-like protein (MLKL). Here, we found that marked steatosis of the liver was induced when a Western diet was given in mice; steatosis was associated with the inhibition of hepatic proteasome activities and with increased levels of key necroptosis-related proteins. Mice fed a Western diet had more severe liver injury, as demonstrated by increases in serum alanine aminotransferase and necrotic areas of liver, after IR than did mice fed a control diet. Although hepatic steatosis was not different between Mlkl knockout mice and wild-type mice, Mlkl knockout mice had decreased hepatic neutrophil infiltration and inflammation and were protected from hepatic IR injury, irrespective of diet. Intriguingly, Ripk3 knockout or Ripk3 kinase-dead knock-in mice were protected against IR injury at the late phase but not the early phase, irrespective of diet. Overall, our findings indicate that liver steatosis exacerbates hepatic IR injury via increased MLKL-mediated necroptosis. Targeting MLKL-mediated necroptosis may help to improve outcomes in steatotic liver transplantation.
Collapse
Affiliation(s)
- Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas.
| | - Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Joshua Kaseff
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Fengyan Deng
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Shaogui Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Ying-Hong Shi
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Tiangang Li
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|