1
|
Feng D, Hwang S, Guillot A, Wang Y, Guan Y, Chen C, Maccioni L, Gao B. Inflammation in Alcohol-Associated Hepatitis: Pathogenesis and Therapeutic Targets. Cell Mol Gastroenterol Hepatol 2024; 18:101352. [PMID: 38697358 PMCID: PMC11234022 DOI: 10.1016/j.jcmgh.2024.04.009] [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: 01/09/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
Alcohol-associated hepatitis (AH) is an acute-on-chronic liver injury that occurs in patients with chronic alcohol-associated liver disease (ALD). Patients with severe AH have high short-term mortality and lack effective pharmacologic therapies. Inflammation is believed to be one of the key factors promoting AH progression and has been actively investigated as therapeutic targets over the last several decades, but no effective inflammatory targets have been identified so far. In this review, we discuss how inflammatory cells and the inflammatory mediators produced by these cells contribute to the development and progression of AH, with focus on neutrophils and macrophages. The crosstalk between inflammatory cells and liver nonparenchymal cells in the pathogenesis of AH is elaborated. We also deliberate the application of recent cutting-edge technologies in characterizing liver inflammation in AH. Finally, the potential therapeutic targets of inflammatory mediators for AH are briefly summarized.
Collapse
Affiliation(s)
- Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland.
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yang Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Yukun Guan
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Cheng Chen
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Luca Maccioni
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
2
|
Lu H. Inflammatory liver diseases and susceptibility to sepsis. Clin Sci (Lond) 2024; 138:435-487. [PMID: 38571396 DOI: 10.1042/cs20230522] [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: 09/03/2023] [Revised: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/05/2024]
Abstract
Patients with inflammatory liver diseases, particularly alcohol-associated liver disease and metabolic dysfunction-associated fatty liver disease (MAFLD), have higher incidence of infections and mortality rate due to sepsis. The current focus in the development of drugs for MAFLD is the resolution of non-alcoholic steatohepatitis and prevention of progression to cirrhosis. In patients with cirrhosis or alcoholic hepatitis, sepsis is a major cause of death. As the metabolic center and a key immune tissue, liver is the guardian, modifier, and target of sepsis. Septic patients with liver dysfunction have the highest mortality rate compared with other organ dysfunctions. In addition to maintaining metabolic homeostasis, the liver produces and secretes hepatokines and acute phase proteins (APPs) essential in tissue protection, immunomodulation, and coagulation. Inflammatory liver diseases cause profound metabolic disorder and impairment of energy metabolism, liver regeneration, and production/secretion of APPs and hepatokines. Herein, the author reviews the roles of (1) disorders in the metabolism of glucose, fatty acids, ketone bodies, and amino acids as well as the clearance of ammonia and lactate in the pathogenesis of inflammatory liver diseases and sepsis; (2) cytokines/chemokines in inflammatory liver diseases and sepsis; (3) APPs and hepatokines in the protection against tissue injury and infections; and (4) major nuclear receptors/signaling pathways underlying the metabolic disorders and tissue injuries as well as the major drug targets for inflammatory liver diseases and sepsis. Approaches that focus on the liver dysfunction and regeneration will not only treat inflammatory liver diseases but also prevent the development of severe infections and sepsis.
Collapse
Affiliation(s)
- Hong Lu
- Department of Pharmacology, SUNY Upstate Medical University, Syracuse, NY 13210, U.S.A
| |
Collapse
|
3
|
Zhou S, Zhong H, Wang Y, Wang X, Pan H, Liu X, Hu L. JNK/MAPK pathway regulation by BEX2 gene silencing in alcoholic hepatitis mice: Effects on oxidative stress. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:1869-1882. [PMID: 37864534 DOI: 10.1111/acer.15178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Alcoholic hepatitis (AH) is a severe alcoholic-related liver disease that is a leading cause of morbidity and mortality, for which effective treatments are lacking. Brain-expressed X-linked gene 2 (BEX2) has been implicated in various diseases, but its association with AH has received limited attention. Thus, this study investigated BEX2's impact on the progression of AH by affecting the c-Jun NH2-terminal kinase/mitogen-activated protein kinase (JNK/MAPK) pathway. METHODS Microarray dataset GSE28619 from the Gene Expression Omnibus database was used to identify differentially expressed genes in AH. Immunohistochemistry, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL), Western blot analysis, and flow cytometry were used to measure various factors in the liver tissue of AH mice. RESULTS BEX2 expression was significantly upregulated in the model. BEX2 gene silencing increased the levels of glutathione peroxidase and superoxide dismutase while decreasing malondialdehyde content; phosphorylation of JNK, c-JUN, and p38MAPK; apoptosis rate; and the extent of JNK/MAPK pathway activation. CONCLUSIONS These findings provide valuable insights into the mechanisms underlying AH development and highlight the potential role of BEX2 gene expression as a promising therapeutic target for AH.
Collapse
Affiliation(s)
- Shuai Zhou
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Anhui Medical University, Hefei, China
| | - Hai Zhong
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yong Wang
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Anhui Medical University, Hefei, China
| | - Xiaoguang Wang
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Hongtao Pan
- Department of General Surgery, Anhui No. 2 Provincial People's Hospital, Anhui Medical University, Hefei, China
| | - Xiaolin Liu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lingyu Hu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
4
|
Liu N, Bauer M, Press AT. The immunological function of CXCR2 in the liver during sepsis. J Inflamm (Lond) 2022; 19:23. [DOI: 10.1186/s12950-022-00321-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 11/15/2022] [Indexed: 12/02/2022] Open
Abstract
Abstract
Background
The chemokine receptor CXCR2 and its ligands, especially CXCL8, are crucial mediators for the progression of liver inflammation and liver failure in sepsis. Neutrophils have the highest CXCR2 expression in mice and humans, and their activation via CXCL8 facilitates their migration to the inflamed liver for the clearance of the pathogens and, in turn, the inflammation.
Main body
In sepsis, the inflammatory insult causes extensive neutrophil migration to the liver that overwhelms the immune response. To compensate for the strong receptor activation, CXCR2 desensitizes, incapacitating the immune cells to efficiently clear pathogens, causing further life-threatening liver damage and uncontrolled pathogen spread.
Conclusion
CXCR2 function during infection strongly depends on the expressing cell type. It signals pro- and anti-inflammatory effects that may prompt novel cell-type-specific CXCR2-directed therapeutics.
Collapse
|
5
|
Single-Chain Fragment Variables Targeting Leukocidin ED Can Alleviate the Inflammation of Staphylococcus aureus-Induced Mastitis in Mice. Int J Mol Sci 2021; 23:ijms23010334. [PMID: 35008761 PMCID: PMC8745144 DOI: 10.3390/ijms23010334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 11/17/2022] Open
Abstract
Staphylococcus aureus is a vital bovine mastitis pathogen causing huge economic losses to the dairy industry worldwide. In our previous studies, leukotoxin ED (LukED) was detected in most S. aureus strains isolated from bovine mastitis. Here, four single-chain fragment variables (scFvs) (ZL8 and ZL42 targeting LukE, ZL22 and ZL23 targeting LukD) were obtained using purified LukE and LukD proteins as the antigens after five rounds of bio-panning. The complementarity-determining region 3 (CDR3) of the VH domain of these scFvs exhibited significant diversities. In vitro, the scFvs significantly decreased LukED-induced cell killing by inhibiting the binding of LukED to chemokine receptors (CCR5 and CXCR2) and reduced the death rates of bovine neutrophils and MAC-T cells caused by LukED and S. aureus (p < 0.05). In an S. aureus-induced mouse mastitis model, histopathology and MPO results revealed that scFvs ameliorated the histopathological damages and reduced the infiltration of inflammatory cells (p < 0.05). The ELISA and qPCR assays showed that scFvs reduced the transcription and expression levels of Tumor Necrosis Factor-alpha (TNF-α), interleukin-1β (IL-1β), IL-6, IL-8 and IL-18 (p < 0.05). The overall results demonstrated the protective anti-inflammatory effect of scFvs in vitro and in vivo, enlightening the potential role of scFvs in the prevention and treatment of S. aureus-induced mastitis.
Collapse
|
6
|
Anti- Staphylococcus aureus Single-Chain Fragment Variables Play a Protective Anti-Inflammatory Role In Vitro and In Vivo. Vaccines (Basel) 2021; 9:vaccines9111300. [PMID: 34835231 PMCID: PMC8618225 DOI: 10.3390/vaccines9111300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is a causative agent of bovine mastitis, capable of causing significant economic losses to the dairy industry worldwide. This study focuses on obtaining single-chain fragment variables (scFvs) against the virulence factors of S. aureus and evaluates the protective effect of scFvs on bovine mammary epithelial (MAC-T) cells and mice mammary gland tissues infected by S. aureus. After five rounds of bio-panning, four scFvs targeting four virulence factors of S. aureus were obtained. The complementarity-determining regions (CDRs) of these scFvs exhibited significant diversities, especially CDR3 of the VH domain. In vitro, each of scFvs was capable of inhibiting S. aureus growth and reducing the damage of MAC-T cells infected by S. aureus. Preincubation of MAC-T cells with scFvs could significantly attenuate the effect of apoptosis and necrosis compared with the negative control group. In vivo, the qPCR and ELISA results demonstrated that scFvs reduced the transcription and expression of Tumor Necrosis Factor alpha (TNF-α), interleukin-1β (IL-1β), IL-6, IL-8, and IL-18. Histopathology and myeloperoxidase (MPO) results showed that scFvs ameliorated the histopathological damages and reduced the inflammatory cells infiltration. The overall results demonstrated the positive anti-inflammatory effect of scFvs, revealing the potential role of scFvs in the prevention and treatment of S. aureus infections.
Collapse
|
7
|
Lim Y, Ku NO. Revealing the Roles of Keratin 8/18-Associated Signaling Proteins Involved in the Development of Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:6401. [PMID: 34203895 PMCID: PMC8232640 DOI: 10.3390/ijms22126401] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 02/08/2023] Open
Abstract
Although hepatocellular carcinoma (HCC) is developed with various etiologies, protection of hepatocytes seems basically essential to prevent the incidence of HCC. Keratin 8 and keratin 18 (K8/K18) are cytoskeletal intermediate filament proteins that are expressed in hepatocytes. They maintain the cell shape and protect cells under stress conditions. Their protective roles in liver damage have been described in studies of mouse models, and K8/K18 mutation frequency in liver patients. Interestingly, K8/K18 bind to signaling proteins such as transcription factors and protein kinases involved in HCC development. Since K8/K18 are abundant cytoskeletal proteins, K8/K18 binding with the signaling factors can alter the availability of the factors. Herein, we discuss the potential roles of K8/K18 in HCC development.
Collapse
Affiliation(s)
- Younglan Lim
- Interdisciplinary Program of Integrated OMICS for Biomedical Sciences, Yonsei University, Seoul 03722, Korea;
| | - Nam-On Ku
- Interdisciplinary Program of Integrated OMICS for Biomedical Sciences, Yonsei University, Seoul 03722, Korea;
- Department of Bio-Convergence ISED, Underwood International College, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
8
|
Liu D, Han P, Gao C, Gao W, Yao X, Liu S. microRNA-155 Modulates Hepatic Stellate Cell Proliferation, Apoptosis, and Cell Cycle Progression in Rats With Alcoholic Hepatitis via the MAPK Signaling Pathway Through Targeting SOCS1. Front Pharmacol 2020; 11:270. [PMID: 32317960 PMCID: PMC7154100 DOI: 10.3389/fphar.2020.00270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 02/26/2020] [Indexed: 12/24/2022] Open
Abstract
The aim of this study was to investigate the regulatory function of the non-coding microRNA-155 (miR-155) and suppressor of cytokine signaling 1 (SOCS1) in alcoholic hepatitis (AH) and its potential mechanism associated with the mitogen-activated protein kinase (MAPK) signaling pathway. Levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), albumin (ALB), total bilirubin (TBIL), malondialdehyde (MDA), and superoxide dismutase (SOD) were measured in a rat model of AH. The biological prediction website microRNA.org and dual-luciferase reporter gene assay were used to identify whether SOCS1 was a direct target of miR-155, and the effects of miR-155 and SOCS1 on the viability, cycle progression, and apoptosis of hepatic stellate cells were assessed using RT-qPCR, Western blot assay, MTT assay, Annexin V/PI double staining, and PI single staining. The levels of ALT, AST, MDA, and TBIL and the liver cell morphology were all prominently changed in AH model rats. miR-155 suppressed SOCS1 by specifically binding to SOCS1-3'-UTR to activate the MAPK signaling pathway. SOCS1 had low expression while miR-155 was highly expressed in AH rats. miR-155 promoted hepatic stellate cell viability and cycle progression and reduced cell apoptosis by silencing SOCS1. Together, we find that silenced miR-155 could upregulate SOCS1 and inactivate the MAPK signaling pathway, thereby inhibiting the proliferation of alcoholic hepatic stellate cells and promoting cell apoptosis.
Collapse
Affiliation(s)
- Dengtao Liu
- Clinical Laboratory, Linyi People's Hospital, Linyi, China
| | - Ping Han
- Department of Pulmonary and Critical Care Medicine, Linyi People's Hospital, Linyi, China
| | - Chunhai Gao
- Clinical Laboratory, Linyi People's Hospital, Linyi, China
| | - Wei Gao
- Clinical Laboratory, Linyi People's Hospital, Linyi, China
| | - Xiaocui Yao
- Clinical Laboratory, Linyi People's Hospital, Linyi, China
| | - Shulan Liu
- Department of Imaging, Linyi People's Hospital, Linyi, China
| |
Collapse
|
9
|
Neuman MG, Seitz HK, French SW, Malnick S, Tsukamoto H, Cohen LB, Hoffman P, Tabakoff B, Fasullo M, Nagy LE, Tuma PL, Schnabl B, Mueller S, Groebner JL, Barbara FA, Yue J, Nikko A, Alejandro M, Brittany T, Edward V, Harrall K, Saba L, Mihai O. Alcoholic-Hepatitis, Links to Brain and Microbiome: Mechanisms, Clinical and Experimental Research. Biomedicines 2020; 8:E63. [PMID: 32197424 PMCID: PMC7148515 DOI: 10.3390/biomedicines8030063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/02/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023] Open
Abstract
The following review article presents clinical and experimental features of alcohol-induced liver disease (ALD). Basic aspects of alcohol metabolism leading to the development of liver hepatotoxicity are discussed. ALD includes fatty liver, acute alcoholic hepatitis with or without liver failure, alcoholic steatohepatitis (ASH) leading to fibrosis and cirrhosis, and hepatocellular cancer (HCC). ALD is fully attributable to alcohol consumption. However, only 10-20% of heavy drinkers (persons consuming more than 40 g of ethanol/day) develop clinical ALD. Moreover, there is a link between behaviour and environmental factors that determine the amount of alcohol misuse and their liver disease. The range of clinical presentation varies from reversible alcoholic hepatic steatosis to cirrhosis, hepatic failure, and hepatocellular carcinoma. We aimed to (1) describe the clinico-pathology of ALD, (2) examine the role of immune responses in the development of alcoholic hepatitis (ASH), (3) propose diagnostic markers of ASH, (4) analyze the experimental models of ALD, (5) study the role of alcohol in changing the microbiota, and (6) articulate how findings in the liver and/or intestine influence the brain (and/or vice versa) on ASH; (7) identify pathways in alcohol-induced organ damage and (8) to target new innovative experimental concepts modeling the experimental approaches. The present review includes evidence recognizing the key toxic role of alcohol in ALD severity. Cytochrome p450 CYP2E1 activation may change the severity of ASH. The microbiota is a key element in immune responses, being an inducer of proinflammatory T helper 17 cells and regulatory T cells in the intestine. Alcohol consumption changes the intestinal microbiota and influences liver steatosis and liver inflammation. Knowing how to exploit the microbiome to modulate the immune system might lead to a new form of personalized medicine in ALF and ASH.
Collapse
Affiliation(s)
- Manuela G. Neuman
- In Vitro Drug Safety and Biotechnology, Toronto, ON M5G 1L5, Canada;
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L5, Canada
| | - Helmut Karl Seitz
- Department of Medicine, Centre of Alcohol Research, University of Heidelberg, Salem Medical Centre, 337374 Heidelberg, Germany; (H.K.S.); (S.M.)
| | - Samuel W. French
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Stephen Malnick
- Department Internal Medicine C, Kaplan Medical Centre and Hebrew University of Jerusalem, Rehovot 76100, Israel;
| | - Heidekazu Tsukamoto
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089-5311, USA;
- Department of Veterans; Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Lawrence B. Cohen
- Division of Gastroenterology, Sunnybrook Health Sciences Centre, Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON M4N 3M5, Canada;
| | - Paula Hoffman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045-0511, USA; (P.H.); (B.T.); (K.H.); (L.S.)
| | - Boris Tabakoff
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045-0511, USA; (P.H.); (B.T.); (K.H.); (L.S.)
| | - Michael Fasullo
- College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12205, USA;
| | - Laura E. Nagy
- Departments of Pathobiology and Gastroenterology, Center for Liver Disease Research, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Pamela L. Tuma
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA; (P.L.T.); (J.L.G.)
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA;
| | - Sebastian Mueller
- Department of Medicine, Centre of Alcohol Research, University of Heidelberg, Salem Medical Centre, 337374 Heidelberg, Germany; (H.K.S.); (S.M.)
| | - Jennifer L. Groebner
- Department of Biology, The Catholic University of America, Washington, DC 20064, USA; (P.L.T.); (J.L.G.)
| | - French A. Barbara
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Jia Yue
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Afifiyan Nikko
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Mendoza Alejandro
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Tillman Brittany
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Vitocruz Edward
- Department of Pathology, Harbor-UCLA Medical Center and Los Angeles BioMedical Institute, Torrance, CA Harbor-UCLA Medical Center, Torrance, CA 90509, USA; (S.W.F.); (F.A.B.); (J.Y.); (A.N.); (M.A.); (T.B.); (V.E.)
| | - Kylie Harrall
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045-0511, USA; (P.H.); (B.T.); (K.H.); (L.S.)
| | - Laura Saba
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045-0511, USA; (P.H.); (B.T.); (K.H.); (L.S.)
| | - Opris Mihai
- In Vitro Drug Safety and Biotechnology, Toronto, ON M5G 1L5, Canada;
- Department Family Medicine Clinic CAR, 010164 Bucharest, Romania
| |
Collapse
|
10
|
Cheng Y, Ma XL, Wei YQ, Wei XW. Potential roles and targeted therapy of the CXCLs/CXCR2 axis in cancer and inflammatory diseases. Biochim Biophys Acta Rev Cancer 2019; 1871:289-312. [DOI: 10.1016/j.bbcan.2019.01.005] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 11/19/2018] [Accepted: 01/09/2019] [Indexed: 12/16/2022]
|
11
|
Massey VL, Qin L, Cabezas J, Caballeria J, Sancho-Bru P, Bataller R, Crews FT. TLR7-let-7 Signaling Contributes to Ethanol-Induced Hepatic Inflammatory Response in Mice and in Alcoholic Hepatitis. Alcohol Clin Exp Res 2018; 42:2107-2122. [PMID: 30103265 PMCID: PMC6282707 DOI: 10.1111/acer.13871] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/08/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Toll-like receptor 7 (TLR7) is an endosomal TLR that is activated by single-stranded RNA, including endogenous microRNAs (e.g., let-7b). Increased hepatic expression of TLRs, microRNAs, and inflammatory mediators is linked to ethanol (EtOH) exposure and to alcoholic liver disease (ALD). ALD invovles chronic hepatic inflammation that can progress to alcoholic hepatitis (AH), a particularly severe form of ALD. This study aimed to investigate TLR7 expression in patients with different liver disease phenotypes and in mouse liver following alcohol exposure. METHODS Hepatic mRNA expression was determined by RNA sequencing of liver tissue from patients with liver disease or normal liver tissue. Mice were exposed to subchronic EtOH followed by administration of the TLR7 agonist imiquimod. Primary human hepatocytes were exposed to EtOH or imiquimod in vitro. RESULTS RNAseq analysis revealed that hepatic expression of TLR7 and let-7b microRNA, an endogenous TLR7 ligand, was significantly increased in AH patients. Hepatic expression of TLR7 and let-7b positively correlated with hepatic IL-8 mRNA expression. In mice, EtOH increased hepatic TLR7 mRNA expression and enhanced imiquimod-induced expression of the pro-inflammatory mediators TNFα, MCP-1, and iNOS. In vitro, EtOH significantly increased hepatocyte TLR7 mRNA and the TLR7 agonist, imiquimod, induced hepatocyte expression of TNFα and IL-8 mRNA. EtOH also increased the release of let-7b in microvesicles from hepatocytes, suggesting that EtOH can increase the expression of both the receptor and its endogenous ligand. CONCLUSIONS These studies suggest that increased TLR7 signaling caused by increased expression of TLR7 and its endogenous ligand let-7b may contribute to the enhanced inflammatory response associated with AH.
Collapse
Affiliation(s)
- Veronica L Massey
- Bowles Center for Alcohol Studies, University of North Carolina Medical School, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Liya Qin
- Bowles Center for Alcohol Studies, University of North Carolina Medical School, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joaquin Cabezas
- Gastroenterology and Hepatology, Hospital Marques de Valdecilla, Research Institute Valdecilla, Santander, Spain
| | - Juan Caballeria
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Catalonia, Spain.,Liver Unit, Hospital Clinic, Barcelona, Catalonia, Spain
| | - Pau Sancho-Bru
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), CIBER de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Catalonia, Spain
| | - Ramon Bataller
- Bowles Center for Alcohol Studies, University of North Carolina Medical School, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Division of Gastroenterology, Hepatology and Nutrition, Center for Liver Diseases, Pittsburgh Liver Research Center, University of Pittsburgh Medical Center (UPMC), Pittsburgh, Pennsylvania
| | - Fulton T Crews
- Bowles Center for Alcohol Studies, University of North Carolina Medical School, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
12
|
The role of the IL-8 signaling pathway in the infiltration of granulocytes into the livers of patients with alcoholic hepatitis. Exp Mol Pathol 2017; 103:137-140. [DOI: 10.1016/j.yexmp.2017.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 08/11/2017] [Indexed: 11/18/2022]
|
13
|
Liu Q, Ni X, Wang Q, Peng Z, Niu L, Wang H, Zhou Y, Sun H, Pan K, Jing B, Zeng D. Lactobacillus plantarum BSGP201683 Isolated from Giant Panda Feces Attenuated Inflammation and Improved Gut Microflora in Mice Challenged with Enterotoxigenic Escherichia coli. Front Microbiol 2017; 8:1885. [PMID: 29018435 PMCID: PMC5623042 DOI: 10.3389/fmicb.2017.01885] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/14/2017] [Indexed: 12/28/2022] Open
Abstract
In this work, we searched for an effective probiotic that can help control intestinal infection, particularly enterotoxigenic Escherichia coli K88 (ETEC) invasion, in giant panda (Ailuropoda melanoleuca). As a potential probiotic strain, Lactobacillus plantarum BSGP201683 (L. plantarum G83) was isolated from the feces of giant panda and proven beneficial in vitro. This study was aimed to evaluate the protective effect of L. plantarum G83 in mice challenged with ETEC. The mice were orally administered with 0.2 mL of PBS containing L. plantarum G83 at 0 colony-forming units (cfu) mL−1 (control; negative control, ETEC group), 5.0 × 108 cfu mL−1 (LDLP), 5.0 × 109 cfu mL−1 (MDLP), and 5.0 × 1010 cfu mL−1 (HDLP) for 14 consecutive days. At day 15, the mice (LDLP, MDLP, HDLP, and ETEC groups) were challenged with ETEC and assessed at 0, 24, and 144 h. Animal health status; chemical and biological intestinal barriers; and body weight were measured. Results showed that L. plantarum G83 supplementation protected the mouse gut mainly by attenuating inflammation and improving the gut microflora. Most indices significantly changed at 24 h after challenge compared to those at 0 and 144 h. All treatment groups showed inhibited plasma diamine oxidase activity and D-lactate concentration. Tight-junction protein expression was down-regulated, and interleukin (IL)-1β, IL-6, IL-8, TLR4, and MyD88 levels were up-regulated in the jejunum in the LDLP and MDLP groups. The number of the Enterobacteriaceae family and the heat-labile enterotoxin (LT) gene decreased (P < 0.05) in the colons in the LDLP and MDLP groups. All data indicated that L. plantarum G83 could attenuate acute intestinal inflammation caused by ETEC infection, and the low and intermediate doses were superior to the high dose. These findings suggested that L. plantarum G83 may serve as a protective probiotic for intestinal disease and merits further investigation.
Collapse
Affiliation(s)
- Qian Liu
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Qiang Wang
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, China
| | - Zhirong Peng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lili Niu
- Chengdu Wildlife Institute, Chengdu Zoo, Chengdu, China
| | - Hengsong Wang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yi Zhou
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hao Sun
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Kangcheng Pan
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Bo Jing
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
14
|
Liu H, French BA, Li J, Tillman B, French SW. Altered regulation of miR-34a and miR-483-3p in alcoholic hepatitis and DDC fed mice. Exp Mol Pathol 2015; 99:552-7. [PMID: 26403328 DOI: 10.1016/j.yexmp.2015.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 09/18/2015] [Indexed: 01/05/2023]
Abstract
MicroRNAs are small noncoding RNAs that negatively regulate gene expression by binding to the untranslated regions of their target mRNAs. Deregulation of miRNAs is shown to play pivotal roles in tumorigenesis and progression. Mallory-Denk Bodies (MDBs) are prevalent in various liver diseases including alcoholic hepatitis (AH) and are formed in mice livers by feeding DDC. By comparing AH livers where MDBs had formed with normal livers, there were significant changes of miR-34a and miR-483-3p by RNA sequencing (RNA-Seq) analyses. Real-time PCR further shows a 3- and 6-fold upregulation (respectively) of miR-34a in the AH livers and in the livers of DDC re-fed mice, while miR-483-3p was significantly downregulated in AH and DDC re-fed mice livers. This indicates that miR-34a and miR-483-3p may be crucial for liver MDB formation. P53 mRNA was found to be significantly downregulated both in the AH livers and in the livers of DDC re-fed mice, indicating that the upregulation of miR-34a is permitted by the decrease of p53 in AH since miR-34a is a main target of p53. Overexpression of miR-34a leads to an increase of p53 targets such as p27, which inhibits the cell cycle leading to cell cycle arrest. Importantly, BRCA1 is a target gene of miR-483-3p by RNA-Seq analyses and the downregulation of miR-483-3p may be the mechanism for liver MDB formation since the BRCA1 signal was markedly upregulated in AH livers. These results constitute a demonstration of the altered regulation of miR-34a and miR-483-3p in the livers of AH and mice fed DDC where MDBs formed, providing further insight into the mechanism of MDB formation mediated by miR-34a and miR-483-3p in AH.
Collapse
Affiliation(s)
- Hui Liu
- Department of Pathology, LABioMed at Harbor UCLA Medical Center, 1000 West Carson Street, Torrance, CA 90509, USA
| | - Barbara A French
- Department of Pathology, LABioMed at Harbor UCLA Medical Center, 1000 West Carson Street, Torrance, CA 90509, USA
| | - Jun Li
- Department of Pathology, LABioMed at Harbor UCLA Medical Center, 1000 West Carson Street, Torrance, CA 90509, USA
| | - Brittany Tillman
- Department of Pathology, LABioMed at Harbor UCLA Medical Center, 1000 West Carson Street, Torrance, CA 90509, USA
| | - Samuel W French
- Department of Pathology, LABioMed at Harbor UCLA Medical Center, 1000 West Carson Street, Torrance, CA 90509, USA.
| |
Collapse
|