1
|
Jiang Y, Xie F, Ling X, Zhang J, Yu Y, Huang Q, Zhang L, Ye L, Tao W, Hou M, Zhang C, Wang J. TCA Cycle Intermediate Mitigates Di(2-ethylhexyl) Phthalate-Induced Cholestatic Liver Injury Through Modulation of the Nrf2/NQO1 Signalling Axis. Basic Clin Pharmacol Toxicol 2025; 136:e70047. [PMID: 40370325 DOI: 10.1111/bcpt.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025]
Abstract
As a commonly used phthalate compound, di(2-ethylhexyl) phthalate (DEHP) has been shown to disrupt the tricarboxylic acid (TCA) cycle and aggravate tissue damage. However, whether the TCA cycle is involved in cholestatic liver injury (CLI) induced by DEHP and the protective effect of dimethyl fumarate (DMF), which is used to supplement TCA intermediate metabolites, remained unclear. Here, mice were randomized into five groups (n = 6/group): (1) Control, (2) DEHP (200 mg/kg/day), (3) DMF (100 mg/kg/day), (4) DEHP + DMF (30 mg/kg/day) and (5) DEHP + DMF (100 mg/kg/day). Our data demonstrated that DEHP exposure upregulated total bile acid (TBA) levels and broke the TCA cycle, resulting in reduced fumaric acid and malic acid. However, we further supplemented fumaric acid with DMF and found that DMF effectively reversed the high levels of TBA, alkaline phosphatase (ALP) and glutamyl transpeptidase (GGT) induced by DEHP in mice. Meanwhile, pathological results in the liver showed that DMF improved bile duct cell damage, inflammatory cell infiltration, collagen deposition and necrosis caused by DEHP. In addition, we found that DEHP elevated the level of interleukin (IL)-1β, IL-6, TNF-α and MDA and decreased the level of SOD in the mouse liver, which was effectively reversed by DMF treatment. Besides, DMF upregulated the expression of Nrf2 and NQO1 in the liver of DEHP-exposed mice. For in vitro validation, AML-12 cells were treated with (1) Control, (2) DEHP (250 μM), (3) DEHP + DMF (10 μM), (4) DEHP + DMF (25 μM) and (5) DEHP + DMF (50 μM). DEHP exposure increased the expression of IL-1β, IL-6 and TNF-α, which was mitigated by DMF, while ML385, an Nrf2 inhibitor, could counteract the anti-inflammatory effects of DMF. These findings indicate that DEHP broke the TCA cycle of the mouse liver, and DMF supplementation protects against DEHP-induced CLI by activating the Nrf2/NQO1 pathway.
Collapse
Affiliation(s)
- Yue Jiang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Fang Xie
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xutao Ling
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jiayi Zhang
- School of Pharmacy, Anhui Medical University, Hefei, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Yun Yu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Qianqian Huang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Lun Zhang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
| | - Lu Ye
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wenkang Tao
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Mengzhen Hou
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| | - Jianqing Wang
- School of Pharmacy, Anhui Medical University, Hefei, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Public Health Clinical Center, Hefei, China
- MOE Key Laboratory of Population Health Across Life Cycle, Hefei, China
| |
Collapse
|
2
|
Lastuvkova H, Dohnalkova E, Manna DF, Cermanova J, Mokry J, Pejchal J, Hirsova P, Nachtigal P, Pavkova I, Bajnokova M, Smutna L, Stefela A, Kamaraj R, Jandova L, Uher M, Pavek P, Micuda S, Hroch M. Dimethyl fumarate attenuates bile acid retention and liver fibrosis in a mouse model of cholestasis. Am J Physiol Gastrointest Liver Physiol 2025; 328:G558-G577. [PMID: 40210415 DOI: 10.1152/ajpgi.00262.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/14/2024] [Accepted: 03/23/2025] [Indexed: 04/12/2025]
Abstract
Cholestatic liver diseases are characterized by intrahepatic accumulation of bile acids (BAs), exacerbating liver inflammation, and fibrosis. Dimethyl fumarate (DMF) is a clinically approved anti-inflammatory drug that demonstrated protective effects in several experimental models of liver injury. Still, its effect on BA homeostasis and liver fibrosis has not been thoroughly studied. Herein, we hypothesized that DMF could improve BA homeostasis and mitigate the progression of cholestasis-induced liver fibrosis. The DMF was administered to mice with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced cholestasis for 4 wk. The content of individual BAs in the plasma, liver, bile, intestine, and feces was measured using the LC-MS method alongside the analysis of liver phenotype and related executive and regulatory pathways. The DMF slowed down the progression of DDC-induced liver fibrosis by suppressing hepatic stellate cell and macrophage activation and by reducing c-Jun N-terminal kinase phosphorylation. Notably, DMF reduced BA cumulation in the plasma and liver of cholestatic mice by increasing BA fecal excretion via their reduced Bacteroidetes phyla-mediated deconjugation in the intestine. In addition, DMF was identified as the antagonist of the mouse farnesoid X receptor in enterocytes. In conclusion, DMF alleviates DDC-induced cholestatic liver injury through pleiotropic action leading to significant anti-inflammatory and antifibrotic activity of the agent. In addition, DMF mitigates BA retention in the liver and plasma by increasing their fecal excretion in cholestatic mice. These findings suggest that DMF warrants further investigation as a potential therapeutic agent for human chronic fibrosing cholestatic liver disorders.NEW & NOTEWORTHY Chronic cholestatic cholangiopathies present a therapeutic challenge due to their complex pathophysiology, where the accumulation of bile acids plays a crucial role. In this study, we found that dimethyl fumarate attenuated cholestatic liver damage in a murine model through its significant anti-inflammatory and antifibrotic activity supported by reduced bile acid accumulation in the plasma and liver via their increased fecal excretion.
Collapse
Affiliation(s)
- Hana Lastuvkova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Ester Dohnalkova
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Dina Faeq Manna
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jolana Cermanova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jaroslav Mokry
- Department of Histology and Embryology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czech Republic
| | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Ivona Pavkova
- Department of Molecular Pathology and Biology, Military Faculty of Medicine, University of Defence in Brno, Hradec Kralove, Czech Republic
| | - Maria Bajnokova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Lucie Smutna
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Alzbeta Stefela
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Rajamanikkam Kamaraj
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Lenka Jandova
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Martin Uher
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Petr Pavek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Stanislav Micuda
- Department of Pharmacology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Milos Hroch
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| |
Collapse
|
3
|
Nosaka T, Naito T, Akazawa Y, Takahashi K, Matsuda H, Ohtani M, Nishizawa T, Okamoto H, Nakamoto Y. Identification of novel antiviral host factors by functional gene expression analysis using in vitro HBV infection assay systems. PLoS One 2025; 20:e0314581. [PMID: 40048440 PMCID: PMC11884705 DOI: 10.1371/journal.pone.0314581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 11/12/2024] [Indexed: 03/09/2025] Open
Abstract
To cure hepatitis B virus (HBV) infection, it is essential to elucidate the function of hepatocyte host factors in regulating the viral life cycle. Signaling and transcription activator of transcription (STAT)1 play important roles in immune responses, but STAT1-independent pathways have also been shown to have important biological reactivity. Using an in vitro HBV infection assay system, the current study aimed to investigate the STAT1-independent host factors that contribute to the control of viral infection by comprehensive functional screening. The in vitro HBV infection system was established using primary human hepatocytes (PXB cells) infected with HBV derived from a plasmid containing the 1.3-mer HBV genome. Comprehensive functional studies were performed using small interfering RNA (siRNA) and vector transfection and analyzed using microarrays. Knockdown of STAT1 increased viral products in HBV-transfected HepG2 cells, but decreased in HBV-infected PXB cells. RNA microarray was performed using HBV-infected PXB cells with STAT1 knockdown. Fumarylacetoacetate hydrolase (FAH) was extracted by siRNA of genes in PXB cells altered by STAT1 knockdown. Transfection of FAH inhibited HBV replication. Dimethyl fumarate (DMF), the methyl ester of FAH metabolite, showed antiviral effects by inducing autophagy and anti-HBV-related genes. Independently of STAT1, FAH was identified as a host factor that contributes to the control of viral infection, and its metabolite, DMF, exhibited antiviral activity. These results suggest that the novel host factor FAH and its metabolites may be an innovative therapeutic strategy to control the HBV life cycle.
Collapse
Affiliation(s)
- Takuto Nosaka
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tatsushi Naito
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Yu Akazawa
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kazuto Takahashi
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hidetaka Matsuda
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Masahiro Ohtani
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tsutomu Nishizawa
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Hiroaki Okamoto
- Division of Virology, Department of Infection and Immunity, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Yasunari Nakamoto
- Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| |
Collapse
|
4
|
Sangineto M, Ciarnelli M, Moola A, Naik Bukke V, Cassano T, Villani R, Romano AD, Di Gioia G, Avolio C, Serviddio G. Krebs cycle derivatives, dimethyl fumarate and itaconate, control metabolic reprogramming in inflammatory human microglia cell line. Mitochondrion 2024; 79:101966. [PMID: 39276907 DOI: 10.1016/j.mito.2024.101966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/29/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
Metabolic reprogramming drives inflammatory activity in macrophages, including microglia, with Krebs cycle (KC) intermediates playing a crucial role as signaling molecules. Here, we show that the bioenergetic profile of LPS-activated human microglialclone 3 cell line (HMC3) is characterized by high levels of glycolysis and mitochondrial (mt) respiration, and the treatment with KC derivatives, namely dimethyl-fumarate (DMF) and itaconate (ITA), almost restores normal metabolism. However, despite comparable bioenergetic and anti-inflammatory effects, the mt hyper-activity was differentially modulated by DMF and ITA. DMF normalized complex I activity, while ITA dampened both complex I and II hyper-activity counteracting oxidative stress more efficiently.
Collapse
Affiliation(s)
- Moris Sangineto
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| | - Martina Ciarnelli
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Archana Moola
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Vidyasagar Naik Bukke
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Tommaso Cassano
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Rosanna Villani
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Antonino D Romano
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Giuseppe Di Gioia
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Carlo Avolio
- Neurology Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gaetano Serviddio
- Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
5
|
Ali FEM, Abdel-Reheim MA, Hassanein EHM, Abd El-Aziz MK, Althagafy HS, Badran KSA. Exploring the potential of drug repurposing for liver diseases: A comprehensive study. Life Sci 2024; 347:122642. [PMID: 38641047 DOI: 10.1016/j.lfs.2024.122642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/24/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024]
Abstract
Drug repurposing involves the investigation of existing drugs for new indications. It offers a great opportunity to quickly identify a new drug candidate at a lower cost than novel discovery and development. Despite the importance and potential role of drug repurposing, there is no specific definition that healthcare providers and the World Health Organization credit. Unfortunately, many similar and interchangeable concepts are being used in the literature, making it difficult to collect and analyze uniform data on repurposed drugs. This research was conducted based on understanding general criteria for drug repurposing, concentrating on liver diseases. Many drugs have been investigated for their effect on liver diseases even though they were originally approved (or on their way to being approved) for other diseases. Some of the hypotheses for drug repurposing were first captured from the literature and then processed further to test the hypothesis. Recently, with the revolution in bioinformatics techniques, scientists have started to use drug libraries and computer systems that can analyze hundreds of drugs to give a short list of candidates to be analyzed pharmacologically. However, this study revealed that drug repurposing is a potential aid that may help deal with liver diseases. It provides available or under-investigated drugs that could help treat hepatitis, liver cirrhosis, Wilson disease, liver cancer, and fatty liver. However, many further studies are needed to ensure the efficacy of these drugs on a large scale.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt; Michael Sayegh, Faculty of Pharmacy, Aqaba University of Technology, Aqaba 77110, Jordan
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Mostafa K Abd El-Aziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Khalid S A Badran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| |
Collapse
|
6
|
Sangineto M, Ciarnelli M, Colangelo T, Moola A, Bukke VN, Duda L, Villani R, Romano A, Giandomenico S, Kanwal H, Serviddio G. Monocyte bioenergetics: An immunometabolic perspective in metabolic dysfunction-associated steatohepatitis. Cell Rep Med 2024; 5:101564. [PMID: 38733988 PMCID: PMC11148801 DOI: 10.1016/j.xcrm.2024.101564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 02/18/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024]
Abstract
Monocytes (Mos) are crucial in the evolution of metabolic dysfunction-associated steatotic liver disease (MASLD) to metabolic dysfunction-associated steatohepatitis (MASH), and immunometabolism studies have recently suggested targeting leukocyte bioenergetics in inflammatory diseases. Here, we reveal a peculiar bioenergetic phenotype in circulating Mos of patients with MASH, characterized by high levels of glycolysis and mitochondrial (mt) respiration. The enhancement of mt respiratory chain activity, especially complex II (succinate dehydrogenase [SDH]), is unbalanced toward the production of reactive oxygen species (ROS) and is sustained at the transcriptional level with the involvement of the AMPK-mTOR-PGC-1α axis. The modulation of mt activity with dimethyl malonate (DMM), an SDH inhibitor, restores the metabolic profile and almost abrogates cytokine production. Analysis of a public single-cell RNA sequencing (scRNA-seq) dataset confirms that in murine models of MASH, liver Mo-derived macrophages exhibit an upregulation of mt and glycolytic energy pathways. Accordingly, the DMM injection in MASH mice contrasts Mo infiltration and macrophagic enrichment, suggesting immunometabolism as a potential target in MASH.
Collapse
Affiliation(s)
- Moris Sangineto
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy.
| | - Martina Ciarnelli
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Tommaso Colangelo
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; Cancer Cell Signalling Unit, Fondazione IRCCS "Casa Sollievo della Sofferenza," 71043 San Giovanni Rotondo (FG), Italy
| | - Archana Moola
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Vidyasagar Naik Bukke
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Loren Duda
- Pathology Unit, Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Rosanna Villani
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Antonino Romano
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Stefania Giandomenico
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Hina Kanwal
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Gaetano Serviddio
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
7
|
Xu Z, Tang W, Xie Q, Cao X, Zhang M, Zhang X, Chai J. Dimethyl fumarate attenuates cholestatic liver injury by activating the NRF2 and FXR pathways and suppressing NLRP3/GSDMD signaling in mice. Exp Cell Res 2023; 432:113781. [PMID: 37722551 DOI: 10.1016/j.yexcr.2023.113781] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/27/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
The progression of cholestasis is characterized by excessive accumulation of bile acids (BAs) in the liver, which leads to oxidative stress (OS), inflammation and liver injury. There are currently limited treatments for cholestasis. Therefore, appropriate drugs for cholestasis treatment need to be developed. Dimethyl fumarate (DMF) has been widely used in the treatment of various diseases and exerts antioxidant and anti-inflammatory effects, but its effect on cholestatic liver disease remains unclarified. We fed mice 3,5-diethoxycarbonyl-1,4-dihydrocollidine or cholic acid to induce cholestatic liver injury and treated these mice with DMF to evaluate its protective ability. Alanine aminotransferase, aspartate aminotransferase, and total liver BAs were assessed as indicators of liver function. The levels of OS, liver inflammation, transporters and metabolic enzymes were also measured. DMF markedly altered the relative ALT and AST levels and enhanced the liver antioxidant capacity. DMF regulated the MST/NRF2 signaling pathway to protect against OS and reduced liver inflammation through the NLRP3/GSDMD signaling pathway. DMF also regulated the levels of BA transporters by promoting FXR protein expression. These findings provide new strategies for the treatment of cholestatic liver disorders.
Collapse
Affiliation(s)
- Ziqian Xu
- School of Medicine, Chongqing University, Chongqing 400030, China; Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wan Tang
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qiaoling Xie
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xinyu Cao
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mengni Zhang
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoxun Zhang
- Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Jin Chai
- School of Medicine, Chongqing University, Chongqing 400030, China; Department of Gastroenterology, Institute of Digestive Diseases of PLA, Cholestatic Liver Diseases Center, and Center for Metabolic Associated Fatty Liver Disease, The First Affiliated Hospital (Southwest Hospital) to Third Military Medical University (Army Medical University), Chongqing 400038, China; The Second Affiliated Hospital, University of South China, Hengyang 421001, China.
| |
Collapse
|
8
|
Tilg H, Adolph TE, Tacke F. Therapeutic modulation of the liver immune microenvironment. Hepatology 2023; 78:1581-1601. [PMID: 37057876 DOI: 10.1097/hep.0000000000000386] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Inflammation is a hallmark of progressive liver diseases such as chronic viral or immune-mediated hepatitis, alcohol-associated liver disease, and NAFLD. Preclinical and clinical studies have provided robust evidence that cytokines and related cellular stress sensors in innate and adaptive immunity orchestrate hepatic disease processes. Unresolved inflammation and liver injury result in hepatic scarring, fibrosis, and cirrhosis, which may culminate in HCC. Liver diseases are accompanied by gut dysbiosis and a bloom of pathobionts, fueling hepatic inflammation. Anti-inflammatory strategies are extensively used to treat human immune-mediated conditions beyond the liver, while evidence for immunomodulatory therapies and cell therapy-based strategies in liver diseases is only emerging. The development and establishment of novel immunomodulatory therapies for chronic liver diseases has been dampened by several clinical challenges, such as invasive monitoring of therapeutic efficacy with liver biopsy in clinical trials and risk of DILI in several studies. Such aspects prevented advancements of novel medical therapies for chronic inflammatory liver diseases. New concepts modulating the liver immune environment are studied and eagerly awaited to improve the management of chronic liver diseases in the future.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| |
Collapse
|
9
|
Manai F, Zanoletti L, Arfini D, Micco SGD, Gjyzeli A, Comincini S, Amadio M. Dimethyl Fumarate and Intestine: From Main Suspect to Potential Ally against Gut Disorders. Int J Mol Sci 2023; 24:9912. [PMID: 37373057 DOI: 10.3390/ijms24129912] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Dimethyl fumarate (DMF) is a well-characterized molecule that exhibits immuno-modulatory, anti-inflammatory, and antioxidant properties and that is currently approved for the treatment of psoriasis and multiple sclerosis. Due to its Nrf2-dependent and independent mechanisms of action, DMF has a therapeutic potential much broader than expected. In this comprehensive review, we discuss the state-of-the-art and future perspectives regarding the potential repurposing of DMF in the context of chronic inflammatory diseases of the intestine, such as inflammatory bowel disorders (i.e., Crohn's disease and ulcerative colitis) and celiac disease. DMF's mechanisms of action, as well as an exhaustive analysis of the in vitro/in vivo evidence of its beneficial effects on the intestine and the gut microbiota, together with observational studies on multiple sclerosis patients, are here reported. Based on the collected evidence, we highlight the new potential applications of this molecule in the context of inflammatory and immune-mediated intestinal diseases.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Lisa Zanoletti
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
- Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Davide Arfini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Simone Giorgio De Micco
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Arolda Gjyzeli
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Sergio Comincini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Marialaura Amadio
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
10
|
Torosian K, Lal E, Kavanaugh A, Loomba R, Ajmera V, Guma M. Psoriatic disease and non-alcoholic fatty liver disease shared pathogenesis review. Semin Arthritis Rheum 2023; 59:152165. [PMID: 36716599 PMCID: PMC9992353 DOI: 10.1016/j.semarthrit.2023.152165] [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: 09/30/2022] [Revised: 12/03/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023]
Abstract
Psoriatic disease (PD) and non-alcoholic fatty liver disease (NAFLD) potentially share disease pathways given the numerous inflammatory pathways involved in both diseases and a higher prevalence of NAFLD in PD patients. Metabolic syndrome and obesity are a key link between the two diseases, but even when controlling for this, associations between both diseases are still seen. Therapeutics that impact metabolic or inflammatory pathways may be impactful in both PD and NAFLD. In this review, we describe common inflammatory pathways contributing to both PD and NAFLD and critically review the potential impact of treatments for and on both diseases.
Collapse
Affiliation(s)
- Kelly Torosian
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Esha Lal
- Department of Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Arthur Kavanaugh
- Department of Rheumatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; NAFLD Research Center, Department of Medicine, University of California at San Diego, La Jolla, USA; Division of Epidemiology, Department of Family and Preventative Medicine, University of California at San Diego, La Jolla, USA
| | - Veeral Ajmera
- Division of Gastroenterology and Hepatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; NAFLD Research Center, Department of Medicine, University of California at San Diego, La Jolla, USA.
| | - Monica Guma
- Department of Rheumatology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA; Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193 Bellaterra, Barcelona, Spain; San Diego VA Healthcare Service, San Diego, CA, 92161, USA.
| |
Collapse
|
11
|
Salama RM, Abbas SS, Darwish SF, Sallam AA, Elmongy NF, El Wakeel SA. Regulation of NOX/p38 MAPK/PPARα pathways and miR-155 expression by boswellic acids reduces hepatic injury in experimentally-induced alcoholic liver disease mouse model: novel mechanistic insight. Arch Pharm Res 2023; 46:323-338. [PMID: 36959348 PMCID: PMC10123034 DOI: 10.1007/s12272-023-01441-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/28/2023] [Indexed: 03/25/2023]
Abstract
Alcoholic liver disease (ALD) refers to hepatic ailments induced by excessive alcohol intake. The pathogenesis of ALD comprises a complex interplay between various mechanistic pathways, among which inflammation and oxidative stress are key players. Boswellic acids (BAs), found in Boswellia serrata, have shown hepatoprotective effects owing to their antioxidant and anti-inflammatory activities, nevertheless, their therapeutic potential against ALD has not been previously investigated. Hence, this study was performed to depict the possible protective effect of BAs and detect their underlying mechanism of action in an experimentally-induced ALD mouse model. Male BALB/c mice were equally categorized into six groups: control, BAs-treated, ALD, and ALD that received BAs at three-dose levels (125, 250, and 500 mg/kg) by oral gavage for 14 days. Results showed that the high dose of BAs had the most protective impact against ALD according to histopathology examination, blood alcohol concentration (BAC), and liver function enzymes. Mechanistic investigations revealed that BAs (500 mg/kg) caused a significant decrease in cytochrome P450 2E1(CYP2E1), nicotine adenine dinucleotide phosphate oxidase (NOX) 1/2/4, p38 mitogen-activated protein kinase (MAPK), and sterol regulatory element-binding protein-1c (SREBP-1c) levels, and the expression of miR-155, yet increased peroxisome proliferator-activated receptor alpha (PPARα) levels. This led to an improvement in lipid profile and reduced hepatic inflammation, oxidative stress, and apoptosis indices. In summary, our study concludes that BAs can protect against ethanol-induced hepatic injury, via modulating NOX/p38 MAPK/PPARα pathways and miR-155 expression.
Collapse
Affiliation(s)
- Rania M Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), KM 28, Cairo-Ismailia Road, Ahmed Orabi District, Cairo, Egypt.
| | - Samah S Abbas
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), KM 28, Cairo-Ismailia Road, Ahmed Orabi District, Cairo, Egypt
| | - Samar F Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt
| | - Al Aliaa Sallam
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Noura F Elmongy
- Physiology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | - Sara A El Wakeel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), KM 28, Cairo-Ismailia Road, Ahmed Orabi District, Cairo, Egypt
| |
Collapse
|
12
|
Wang J, Sun Z, Xie J, Ji W, Cui Y, Ai Z, Liang G. Inflammasome and pyroptosis in autoimmune liver diseases. Front Immunol 2023; 14:1150879. [PMID: 36969233 PMCID: PMC10030845 DOI: 10.3389/fimmu.2023.1150879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
Autoimmune hepatitis (AIH), primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), and IgG4-related sclerosing cholangitis (IgG4-SC) are the four main forms of autoimmune liver diseases (AILDs), which are all defined by an aberrant immune system attack on the liver. Most previous studies have shown that apoptosis and necrosis are the two major modes of hepatocyte death in AILDs. Recent studies have reported that inflammasome-mediated pyroptosis is critical for the inflammatory response and severity of liver injury in AILDs. This review summarizes our present understanding of inflammasome activation and function, as well as the connections among inflammasomes, pyroptosis, and AILDs, thus highlighting the shared features across the four disease models and gaps in our knowledge. In addition, we summarize the correlation among NLRP3 inflammasome activation in the liver-gut axis, liver injury, and intestinal barrier disruption in PBC and PSC. We summarize the differences in microbial and metabolic characteristics between PSC and IgG4-SC, and highlight the uniqueness of IgG4-SC. We explore the different roles of NLRP3 in acute and chronic cholestatic liver injury, as well as the complex and controversial crosstalk between various types of cell death in AILDs. We also discuss the most up-to-date developments in inflammasome- and pyroptosis-targeted medicines for autoimmune liver disorders.
Collapse
Affiliation(s)
- Jixuan Wang
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiwen Sun
- Department of Liver, Spleen and Stomach Diseases, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jingri Xie
- Department of Liver, Spleen and Stomach Diseases, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wanli Ji
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yang Cui
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zongxiong Ai
- School of First Clinical Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Guoying Liang, ; Zongxiong Ai,
| | - Guoying Liang
- Department of Liver, Spleen and Stomach Diseases, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Guoying Liang, ; Zongxiong Ai,
| |
Collapse
|
13
|
Lactobacillus plantarum J26 Alleviating Alcohol-Induced Liver Inflammation by Maintaining the Intestinal Barrier and Regulating MAPK Signaling Pathways. Nutrients 2022; 15:nu15010190. [PMID: 36615846 PMCID: PMC9824527 DOI: 10.3390/nu15010190] [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: 11/26/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
Alcoholic liver disease (ALD), as a global health problem, is mainly caused by liver inflammation. Meanwhile, probiotics have been considered as a potential and promising strategy to prevent and alleviate ALD. This study aimed to investigate the ameliorative effect of pre-intaking with Lactobacillus plantarum J26 (L. plantarum J26) on alcohol-induced liver inflammation, with emphasis on the underlying mechanism for alleviating ALD. The results indicated that L. plantarum J26 could reduce the abundance of Gram-negative pathogenic bacteria by regulating the gut microbiota in mice with alcoholic liver injury, thereby reducing the lipopolysaccharide (LPS) content in the intestine. In addition, L. plantarum J26 could also maintain the intestinal barrier, prevent LPS from crossing the intestinal barrier to correct disorders of the gut-liver axis and then inhibit the activation of Toll-like receptor 4 (TLR4)-mediated MAPK signaling pathway, reducing liver inflammation and restoring liver functions. In conclusion, pre-intake of L. plantarum J26 could alleviate alcohol-induced liver inflammation, which may be closely related to the role of intestinal microbiota in regulating and maintaining the intestinal barrier and then regulating the MAPK signaling pathway.
Collapse
|
14
|
Bukke VN, Moola A, Serviddio G, Vendemiale G, Bellanti F. Nuclear factor erythroid 2-related factor 2-mediated signaling and metabolic associated fatty liver disease. World J Gastroenterol 2022; 28:6909-6921. [PMID: 36632321 PMCID: PMC9827579 DOI: 10.3748/wjg.v28.i48.6909] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/05/2022] [Accepted: 11/22/2022] [Indexed: 12/26/2022] Open
Abstract
Oxidative stress is a key driver in the development and progression of several diseases, including metabolic associated fatty liver disease (MAFLD). This condition includes a wide spectrum of pathological injuries, extending from simple steatosis to inflammation, fibrosis, cirrhosis, and hepatocellular carcinoma. Excessive buildup of lipids in the liver is strictly related to oxidative stress in MAFLD, progressing to liver fibrosis and cirrhosis. The nuclear factor erythroid 2-related factor 2 (NRF2) is a master regulator of redox homeostasis. NRF2 plays an important role for cellular protection by inducing the expression of genes related to antioxidant, anti-inflammatory, and cytoprotective response. Consistent evidence demonstrates that NRF2 is involved in every step of MAFLD deve-lopment, from simple steatosis to inflammation, advanced fibrosis, and ini-tiation/progression of hepatocellular carcinoma. NRF2 activators regulate lipid metabolism and oxidative stress alleviating the fatty liver disease by inducing the expression of cytoprotective genes. Thus, modulating NRF2 activation is crucial not only in understanding specific mechanisms underlying MAFLD progression but also to characterize effective therapeutic strategies. This review outlined the current knowledge on the effects of NRF2 pathway, modulators, and mechanisms involved in the therapeutic implications of liver steatosis, inflammation, and fibrosis in MAFLD.
Collapse
Affiliation(s)
- Vidyasagar Naik Bukke
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Archana Moola
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Gianluigi Vendemiale
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| | - Francesco Bellanti
- Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy
| |
Collapse
|
15
|
Patel V, Joharapurkar A, Kshirsagar S, Patel M, Savsani H, Patel A, Ranvir R, Jain M. Repurposing dimethyl fumarate for gastric ulcer and ulcerative colitis: Evidence of local efficacy without systemic side effect. MEDICINE IN DRUG DISCOVERY 2022; 16:100142. [DOI: 10.1016/j.medidd.2022.100142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
16
|
Lucas-Ruiz F, Peñín-Franch A, Pons JA, Ramírez P, Pelegrín P, Cuevas S, Baroja-Mazo A. Emerging Role of NLRP3 Inflammasome and Pyroptosis in Liver Transplantation. Int J Mol Sci 2022; 23:ijms232214396. [PMID: 36430874 PMCID: PMC9698208 DOI: 10.3390/ijms232214396] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
The nucleotide-binding domain leucine-rich repeat-receptor, pyrin domain-containing-3 (NLRP3) inflammasome contributes to the inflammatory response by activating caspase-1, which in turn participates in the maturation of interleukin (IL)-1β and IL-18, which are mainly secreted via pyroptosis. Pyroptosis is a lytic type of cell death that is controlled by caspase-1 processing gasdermin D. The amino-terminal fragment of gasdermin D inserts into the plasma membrane, creating stable pores and enabling the release of several proinflammatory factors. The activation of NLRP3 inflammasome and pyroptosis has been involved in the progression of liver fibrosis and its end-stage cirrhosis, which is among the main etiologies for liver transplantation (LT). Moreover, the NLRP3 inflammasome is involved in ischemia-reperfusion injury and early inflammation and rejection after LT. In this review, we summarize the recent literature addressing the role of the NLRP3 inflammasome and pyroptosis in all stages involved in LT and argue the potential targeting of this pathway as a future therapeutic strategy to improve LT outcomes. Likewise, we also discuss the impact of graft quality influenced by donation after circulatory death and the expected role of machine perfusion technology to modify the injury response related to inflammasome activation.
Collapse
Affiliation(s)
- Fernando Lucas-Ruiz
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Alejandro Peñín-Franch
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - José Antonio Pons
- Hepatology and Liver Transplant Unit, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Pablo Ramírez
- General Surgery and Abdominal Solid Organ Transplantation Unit, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Pablo Pelegrín
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
- Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, 30120 Murcia, Spain
- Correspondence: (P.P.); (A.B.-M.); Tel.: +34-86-8885-031 (P.P.); Tel.: +34-86-8885-039 (A.B.-M.)
| | - Santiago Cuevas
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
| | - Alberto Baroja-Mazo
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Pascual Parrilla), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain
- Correspondence: (P.P.); (A.B.-M.); Tel.: +34-86-8885-031 (P.P.); Tel.: +34-86-8885-039 (A.B.-M.)
| |
Collapse
|
17
|
Hughey CC, Puchalska P, Crawford PA. Integrating the contributions of mitochondrial oxidative metabolism to lipotoxicity and inflammation in NAFLD pathogenesis. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159209. [DOI: 10.1016/j.bbalip.2022.159209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/25/2022] [Accepted: 07/27/2022] [Indexed: 11/28/2022]
|
18
|
Tan J, Zhang J, Wang M, Wang Y, Dong M, Ma X, Sun B, Liu S, Zhao Z, Chen L, Jin W, Liu K, Xin Y, Zhuang L. DRAM1 increases the secretion of PKM2-enriched EVs from hepatocytes to promote macrophage activation and disease progression in ALD. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:375-389. [PMID: 35036051 PMCID: PMC8728309 DOI: 10.1016/j.omtn.2021.12.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/09/2021] [Indexed: 11/24/2022]
Abstract
DNA damage-regulated autophagy modulator 1 (DRAM1) could play important roles in inflammation and hepatic apoptosis, while its roles in alcohol-related liver disease (ALD), which is characterized by hepatic inflammation and apoptosis, are still unclear. In this study, we explored the expression, role, and mechanism of DRAM1 in ALD. Firstly, our results showed that DRAM1 was significantly increased in liver tissues of mice at the early stage of alcohol treatment. In addition, DRAM1 knockout reduced, and liver-specific overexpression of DRAM1 aggravated, alcohol-induced hepatic steatosis, injury, and expressions of M1 macrophage markers in mice. Furthermore, ethanol-induced DRAM1 of hepatic cells increased pyruvate kinase M2 (PKM2)-enriched extracellular vesicles (EVs), and ectosomes derived from hepatic cells with DRAM1 overexpression promoted macrophage activation. Mechanistic investigations showed that DRAM1 interacted with PKM2 and increased the PKM2 level in plasma membrane. At last, DRAM1 was significantly increased in liver tissues of ALD patients, and it was positively correlated with M1 macrophage markers. Taken together, this study revealed that ethanol-induced DRAM1 of hepatic cells could increase the PKM2-enriched EVs, promote macrophage activation, and aggravate the disease progression of ALD. These findings suggested that DRAM1 might be a potentially promising target for the therapy of ALD.
Collapse
Affiliation(s)
- Jie Tan
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Jie Zhang
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Mengke Wang
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Yifen Wang
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Mengzhen Dong
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Xuefeng Ma
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Baokai Sun
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Shousheng Liu
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China
| | - Zhenzhen Zhao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China
| | - Lizhen Chen
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Wenwen Jin
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Kai Liu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Yongning Xin
- Department of Infectious Diseases, Qingdao Municipal Hospital, Qingdao University, Qingdao 266011, China
| | - Likun Zhuang
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao 266071, China
| |
Collapse
|
19
|
Sangineto M, Grander C, Grabherr F, Mayr L, Enrich B, Schwärzler J, Dallio M, Bukke VN, Moola A, Moschetta A, Adolph TE, Sabbà C, Serviddio G, Tilg H. Recovery of Bacteroides thetaiotaomicron ameliorates hepatic steatosis in experimental alcohol-related liver disease. Gut Microbes 2022; 14:2089006. [PMID: 35786161 PMCID: PMC9255095 DOI: 10.1080/19490976.2022.2089006] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 06/06/2022] [Indexed: 02/04/2023] Open
Abstract
Alcohol-related liver disease (ALD) is a major cause of liver disease and represents a global burden, as treatment options are scarce. Whereas 90% of ethanol abusers develop alcoholic fatty liver disease (AFLD), only a minority evolves to steatohepatitis and cirrhosis. Alcohol increases lipogenesis and suppresses lipid-oxidation implying steatosis, although the key role of intestinal barrier integrity and microbiota in ALD has recently emerged. Bacteroides thetaiotaomicron (Bt) is a prominent member of human and murine intestinal microbiota, and plays important functions in metabolism, gut immunity, and mucosal barrier. We aimed to investigate the role of Bt in the genesis of ethanol-induced liver steatosis. Bt DNA was measured in feces of wild-type mice receiving a Lieber-DeCarli diet supplemented with an increase in alcohol concentration. In a second step, ethanol-fed mice were orally treated with living Bt, followed by analysis of intestinal homeostasis and histological and biochemical alterations in the liver. Alcohol feeding reduced Bt abundance, which was preserved by Bt oral supplementation. Bt-treated mice displayed lower hepatic steatosis and triglyceride content. Bt restored mucosal barrier and reduced LPS translocation by enhancing mucus thickness and production of Mucin2. Furthermore, Bt up-regulated Glucagon-like peptide-1 (GLP-1) expression and restored ethanol-induced Fibroblast growth factor 15 (FGF15) down-regulation. Lipid metabolism was consequently affected as Bt administration reduced fatty acid synthesis (FA) and improved FA oxidation and lipid exportation. Moreover, treatment with Bt preserved the mitochondrial fitness and redox state in alcohol-fed mice. In conclusion, recovery of ethanol-induced Bt depletion by oral supplementation was associated with restored intestinal homeostasis and ameliorated experimental ALD. Bt could serve as a novel probiotic to treat ALD in the future.
Collapse
Affiliation(s)
- Moris Sangineto
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Barbara Enrich
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Marcello Dallio
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Vidyasagar Naik Bukke
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Archana Moola
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari, Bari, Italy
| | - Timon E. Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Carlo Sabbà
- Department of Interdisciplinary Medicine, University of Bari, Bari, Italy
| | - Gaetano Serviddio
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
20
|
Tong H, Ding Y, Gui X, Sun Z, Wang G, Zhang S, Xu Z, Wang X, Xu X, Ju W, Li Y, Li Z, Zeng L, Xu K, Qiao J. Dimethyl fumarate inhibits antibody-induced platelet destruction in immune thrombocytopenia mouse. Thromb J 2021; 19:61. [PMID: 34454532 PMCID: PMC8403390 DOI: 10.1186/s12959-021-00314-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 08/19/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Immune thrombocytopenia (ITP) is an autoimmune disease characterized as a low platelet count resulting from immune-mediated platelet destruction. Dimethyl fumarate (DMF) is widely applied for the treatment of several autoimmune diseases with immunosuppressive effect. However, whether it ameliorates ITP is unclear. This study aims to evaluate whether DMF has a preventive effect on ITP in mice. METHODS DMF (30, 60 or 90 mg/kg body weight) was intraperitoneally injected into mice followed by injection of rat anti-mouse integrin GPIIb/CD41antibody to induce ITP. Peripheral blood was isolated to measure platelet count and spleen mononuclear cells were extracted to measure Th1 and Treg cells along with detecting the levels of IFN-γ, and TGFβ-1 in plasma and CD68 expression in spleen by immuohistochemical staining. Additionally, macrophage cell line RAW264.7 was cultured and treated with DMF followed by analysis of cell apoptosis and cycle, and the expression of FcγRI, FcγRIIb and FcγRIV mRNA. RESULTS DMF significantly inhibited antiplatelet antibody-induced platelet destruction, decreased Th1 cells and the expression of T-bet and IFN-γ, upregulated Treg cells and the expression of Foxp3 and TGF-β1 as well as reduced CD68 expression in the spleen of ITP mouse. DMF-treated RAW264.7 cells showed S-phase arrest, increased apoptosis and downregulated expression of FcγRI and FcγRIV. Meanwhile, in vitro treatment of DMF also decreased the expression of cyclin D1 and E2, reduced Bcl-2 level and increased Bax expression and caspase-3 activation. CONCLUSIONS In conclusion, DMF prevents antibody-mediated platelet destruction in ITP mice possibly through promoting apoptosis, indicating that it might be used as a new approach for the treatment of ITP.
Collapse
Affiliation(s)
- Huan Tong
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Yangyang Ding
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Xiang Gui
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Zengtian Sun
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Guozhang Wang
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Sixuan Zhang
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Zhengqing Xu
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Xiamin Wang
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Xiaoqi Xu
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Yue Li
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China. .,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China. .,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China. .,School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China.
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China. .,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China. .,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China.
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, 84 West Huaihai Road, Quanshan District, Xuzhou, 221002, Jiangsu, China. .,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China. .,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, 221002, China.
| |
Collapse
|
21
|
Wang X, Chang X, Zhan H, Li C, Zhang Q, Li S, Sun Y. Curcumin combined with Baicalin attenuated ethanol-induced hepatitis by suppressing p38 MAPK and TSC1/ eIF-2α/ATF4 pathways in rats. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2020.100851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
Sangineto M, Bukke VN, Bellanti F, Tamborra R, Moola A, Duda L, Villani R, Romano AD, Serviddio G. A Novel Nutraceuticals Mixture Improves Liver Steatosis by Preventing Oxidative Stress and Mitochondrial Dysfunction in a NAFLD Model. Nutrients 2021; 13:652. [PMID: 33671262 PMCID: PMC7923152 DOI: 10.3390/nu13020652] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/13/2021] [Accepted: 02/14/2021] [Indexed: 02/05/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the leading cause of liver disease globally, and represents a health care burden as treatment options are very scarce. The reason behind the NAFLD progression to non-alcoholic steatohepatitis (NASH) is not completely understood. Recently, the deficiency of micronutrients (e.g., vitamins, minerals, and other elements) has been suggested as crucial in NAFLD progression, such that recent studies reported the potential hepatic antioxidant properties of micronutrients supplementation. However, very little is known. Here we have explored the potential beneficial effects of dietary supplementation with FLINAX, a novel mixture of nutraceuticals (i.e., vitamin E, vitamin D3, olive dry-extract, cinnamon dry-extract and fish oil) in a NAFLD model characterized by oxidative stress and mitochondrial function impairment. Steatosis was firstly induced in Wistar rats by feeding with a high-fat/high-cholesterol diet for 4 weeks, and following this the rats were divided into two groups. One group (n = 8) was treated for 2 weeks with a normal chow-diet, while a second group (n = 8) was fed with a chow-diet supplemented with 2% FLINAX. Along with the entire experiment (6 weeks), a third group of rats was fed with a chow-diet only as control. Statistical analysis was performed with Student's T test or one-way ANOVA followed by post-hoc Bonferroni test when appropriate. Steatosis, oxidative stress and mitochondrial respiratory chain (RC) complexes activity were analyzed in liver tissues. The dietary supplementation with FLINAX significantly improved hepatic steatosis and lipid accumulation compared to untreated rats. The mRNA and protein levels analysis showed that CPT1A and CPT2 were up-regulated by FLINAX, suggesting the enhancement of fatty acids oxidation (FAO). Important lipoperoxidation markers (i.e., HNE- and MDA-protein adducts) and the quantity of total mitochondrial oxidized proteins were significantly lower in FLINAX-treated rats. Intriguingly, FLINAX restored the mitochondrial function, stimulating the activity of mitochondrial RC complexes (i.e., I, II, III and ATP-synthase) and counteracting the peroxide production from pyruvate/malate (complex I) and succinate (complex II). Therefore, the supplementation with FLINAX reprogrammed the cellular energy homeostasis by restoring the efficiency of mitochondrial function, with a consequent improvement in steatosis.
Collapse
Affiliation(s)
- Moris Sangineto
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.S.); (V.N.B.); (R.T.); (A.M.); (R.V.); (A.D.R.)
| | - Vidyasagar Naik Bukke
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.S.); (V.N.B.); (R.T.); (A.M.); (R.V.); (A.D.R.)
| | - Francesco Bellanti
- Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Rosanna Tamborra
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.S.); (V.N.B.); (R.T.); (A.M.); (R.V.); (A.D.R.)
| | - Archana Moola
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.S.); (V.N.B.); (R.T.); (A.M.); (R.V.); (A.D.R.)
| | - Loren Duda
- Pathology Unit, Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Rosanna Villani
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.S.); (V.N.B.); (R.T.); (A.M.); (R.V.); (A.D.R.)
| | - Antonino Davide Romano
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.S.); (V.N.B.); (R.T.); (A.M.); (R.V.); (A.D.R.)
| | - Gaetano Serviddio
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.S.); (V.N.B.); (R.T.); (A.M.); (R.V.); (A.D.R.)
| |
Collapse
|
23
|
Immunity as Cornerstone of Non-Alcoholic Fatty Liver Disease: The Contribution of Oxidative Stress in the Disease Progression. Int J Mol Sci 2021; 22:ijms22010436. [PMID: 33406763 PMCID: PMC7795122 DOI: 10.3390/ijms22010436] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the hepatic manifestation of metabolic syndrome and has become the major cause of chronic liver disease, especially in western countries. NAFLD encompasses a wide spectrum of hepatic histological alterations, from simple steatosis to steatohepatitis and cirrhosis with a potential development of hepatocellular carcinoma. Non-alcoholic steatohepatitis (NASH) is characterized by lobular inflammation and fibrosis. Several studies reported that insulin resistance, redox unbalance, inflammation, and lipid metabolism dysregulation are involved in NAFLD progression. However, the mechanisms beyond the evolution of simple steatosis to NASH are not clearly understood yet. Recent findings suggest that different oxidized products, such as lipids, cholesterol, aldehydes and other macromolecules could drive the inflammation onset. On the other hand, new evidence indicates innate and adaptive immunity activation as the driving force in establishing liver inflammation and fibrosis. In this review, we discuss how immunity, triggered by oxidative products and promoting in turn oxidative stress in a vicious cycle, fuels NAFLD progression. Furthermore, we explored the emerging importance of immune cell metabolism in determining inflammation, describing the potential application of trained immune discoveries in the NASH pathological context.
Collapse
|
24
|
Timpani CA, Rybalka E. Calming the (Cytokine) Storm: Dimethyl Fumarate as a Therapeutic Candidate for COVID-19. Pharmaceuticals (Basel) 2020; 14:15. [PMID: 33375288 PMCID: PMC7824470 DOI: 10.3390/ph14010015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
COVID-19 has rapidly spread worldwide and incidences of hospitalisation from respiratory distress are significant. While a vaccine is in the pipeline, there is urgency for therapeutic options to address the immune dysregulation, hyperinflammation and oxidative stress that can lead to death. Given the shared pathogenesis of severe cases of COVID-19 with aspects of multiple sclerosis and psoriasis, we propose dimethyl fumarate as a viable treatment option. Currently approved for multiple sclerosis and psoriasis, dimethyl fumarate is an immunomodulatory, anti-inflammatory and anti-oxidative drug that could be rapidly implemented into the clinic to calm the cytokine storm which drives severe COVID-19.
Collapse
Affiliation(s)
- Cara A. Timpani
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science, St Albans, VIC 3021, Australia
| | - Emma Rybalka
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science, St Albans, VIC 3021, Australia
| |
Collapse
|