1
|
Gao C, Wang S, Xie X, Ramadori P, Li X, Liu X, Ding X, Liang J, Xu B, Feng Y, Tan X, Wang H, Zhang Y, Zhang H, Zhang T, Mi P, Li S, Zhang C, Yuan D, Heikenwalder M, Zhang P. Single-cell Profiling of Intrahepatic Immune Cells Reveals an Expansion of Tissue-resident Cytotoxic CD4 + T Lymphocyte Subset Associated With Pathogenesis of Alcoholic-associated Liver Diseases. Cell Mol Gastroenterol Hepatol 2024; 19:101411. [PMID: 39349248 PMCID: PMC11719870 DOI: 10.1016/j.jcmgh.2024.101411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND & AIMS The immunological mechanisms underpinning the pathogenesis of alcoholic-associated liver disease (ALD) remain incompletely elucidated. This study aims to explore the transcriptomic profiles of hepatic immune cells in ALD compared with healthy individuals and those with metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS We utilized single-cell RNA sequencing to analyze liver samples from healthy subjects and patients with MASLD and ALD, focusing on the immune cell landscapes within the liver. Key alterations in immune cell subsets were further validated using liver biopsy samples from additional patient cohorts. RESULTS We observed a significant accumulation of CD4+ T cells in livers of patients with ALD, surpassing the prevalence of CD8+ T cells, in contrast to patients with MASLD and healthy counterparts, whereas natural killer (NK) cells and γδT cells exhibited reduced intrahepatic infiltration. In-depth transcriptional and developmental trajectory analyses unveiled that a distinct CD4+ subset characterized by granzyme K (GZMK) expression, displaying a tissue-resident signature and terminal effector state, prominently enriched among CD4+ T cells infiltrating the livers of patients with ALD. Subsequent examination of an independent ALD patient cohort corroborated the substantial enrichment of GZMK+CD4+ T lymphocytes, primarily within liver fibrotic zones, suggesting their potential involvement in disease progression. Additionally, we noted shifts in myeloid populations, with expanded APOE+ macrophage and FCGR3B+ monocyte subsets in ALD samples relative to MASLD and healthy tissues. CONCLUSIONS In summary, this study unravels the intricate cellular diversity within hepatic immune cell populations, highlighting the pivotal immune pathogenic role of the GZMK+CD4+ T lymphocyte subset in ALD pathogenesis.
Collapse
Affiliation(s)
- Chao Gao
- Department of Hepatobiliary Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shiguan Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaoyu Xie
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Pierluigi Ramadori
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Tübingen, Germany
| | - Xinying Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiaoyu Liu
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
| | - Xue Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jinyuan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Bowen Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yawei Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xueying Tan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Haoran Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Haiyan Zhang
- Department of Biochemistry, Heze Medical College, Heze, Shandong, China
| | - Tingguo Zhang
- Institute of Pathology and Pathophysiology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ping Mi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shiyang Li
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
| | - Cuijuan Zhang
- Institute of Pathology and Pathophysiology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Pathology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Detian Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; University Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Tübingen, Germany.
| | - Peng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
2
|
Martinez-Castillo M, Altamirano-Mendoza I, Sánchez-Valle S, García-Islas L, Sánchez-Barragán M, Hernández-Santillán M, Hernández-Barragán A, Pérez-Hernández J, Higuera-de la Tijera F, Gutierrez-Reyes G. Desregulación inmunológica y fisiopatología del consumo de alcohol y la enfermedad hepática alcohólica. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO 2023; 88:136-154. [DOI: 10.1016/j.rgmx.2023.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
3
|
Martinez-Castillo M, Altamirano-Mendoza I, Sánchez-Valle S, García-Islas L, Sánchez-Barragán M, Hernández-Santillán M, Hernández-Barragán A, Pérez-Hernández J, Higuera-de la Tijera F, Gutierrez-Reyes G. Immune dysregulation and pathophysiology of alcohol consumption and alcoholic liver disease. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO (ENGLISH EDITION) 2023; 88:136-154. [PMID: 36973122 DOI: 10.1016/j.rgmxen.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 01/13/2023] [Indexed: 03/28/2023] Open
Abstract
Alcoholic liver disease (ALD) is a clinical-pathologic entity caused by the chronic excessive consumption of alcohol. The disease includes a broad spectrum of anomalies at the cellular and tissual level that can cause acute-on-chronic (alcoholic hepatitis) or chronic (fibrosis, cirrhosis, hepatocellular cancer) injury, having a great impact on morbidity and mortality worldwide. Alcohol is metabolized mainly in the liver. During alcohol metabolism, toxic metabolites, such as acetaldehyde and oxygen reactive species, are produced. At the intestinal level, alcohol consumption can cause dysbiosis and alter intestinal permeability, promoting the translocation of bacterial products and causing the production of inflammatory cytokines in the liver, perpetuating local inflammation during the progression of ALD. Different study groups have reported systemic inflammatory response disturbances, but reports containing a compendium of the cytokines and cells involved in the pathophysiology of the disease, from the early stages, are difficult to find. In the present review article, the role of the inflammatory mediators involved in ALD progression are described, from risky patterns of alcohol consumption to advanced stages of the disease, with the aim of understanding the involvement of immune dysregulation in the pathophysiology of ALD.
Collapse
|
4
|
Hasa E, Hartmann P, Schnabl B. Liver cirrhosis and immune dysfunction. Int Immunol 2022; 34:455-466. [PMID: 35792761 PMCID: PMC9447994 DOI: 10.1093/intimm/dxac030] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/27/2022] [Indexed: 01/05/2023] Open
Abstract
Cirrhosis is end-stage liver disease resulting from various etiologies and is a common cause of death worldwide. The progression from compensated to decompensated cirrhosis to acute-on-chronic liver failure (ACLF) is due to multiple factors, including continuation of alcohol use or continued exposure to other toxins, an imbalance of the gut microbiota (dysbiosis), increased gut permeability and a disrupted immune response. This disrupted immune response is also named cirrhosis-associated immune dysfunction, which is characterized by worsening systemic inflammation with concomitant immune paralysis, as liver disease deteriorates. This review highlights central immunologic events during the exacerbation of cirrhosis and characterizes the different immune cell populations involved therein.
Collapse
|
5
|
Zuluaga P, Teniente-Serra A, Fuster D, Quirant-Sánchez B, Hernandez-Rubio A, Martínez-Cáceres E, Muga R. Increased Natural Killer Cells Are Associated with Alcohol Liver Fibrosis and with T Cell and Cytotoxic Subpopulations Change. J Clin Med 2022; 11:jcm11020305. [PMID: 35054000 PMCID: PMC8780875 DOI: 10.3390/jcm11020305] [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: 12/14/2021] [Accepted: 12/31/2021] [Indexed: 12/20/2022] Open
Abstract
Natural killer (NK) cells play a therapeutic role in liver fibrosis (LF). We aimed to analyze NK cells in heavy drinkers without cirrhosis or decompensated liver disease and establish correlations with other related subpopulations. Data on sociodemographic characteristics, alcohol consumption, laboratory parameters, and immunophenotyping of NK (CD16+/CD56+), T (CD3+), B (CD19+), NKT (CD16+/CD56+/CD3+), and cytotoxic (CD3-CD8+) cells were collected. Fibrosis-4 (FIB-4) scores were used to compare patients without (FIB-4 < 1.45) and with (FIB-4 > 3.25) advanced LF (ALF). We included 136 patients (76% male) with a mean age of 49 years who had a 15-year alcohol use disorder (AUD) and alcohol consumption of 164 g/day. Patients with ALF (n = 25) presented significantly lower absolute total lymphocyte, T cell, B cell, and NKT cell numbers than patients without LF (n = 50; p < 0.01). However, the NK cells count was similar (208 ± 109 cells/µL vs. 170 ± 105 cells/µL) in both groups. The T cells percentage was lower (80.3 ± 5.6% vs. 77 ± 7%; p = 0.03) and the NK cells percentage was higher (9.7 ± 5% vs. 13 ± 6%; p = 0.02) in patients with ALF than in those without LF. The percentages of NK cells and T cells were inversely correlated in patients without (r = –0.65, p < 0.01) and with ALF (r = −0.64; p < 0.01). Additionally, the NK cells and CD3-CD8+ cell percentages were positively correlated in patients without (r = 0.87, p < 0.01) and with (r = 0.92; p < 0.01) ALF. Conclusions: Heavy drinkers without decompensated liver disease showed an increase in NK cells related to T cells lymphopenia and an increase in cytotoxic populations. The interaction of NK cells with other subpopulations may modify alcohol-related liver disease progression.
Collapse
Affiliation(s)
- Paola Zuluaga
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol-IGTP, Universitat Autònoma de Barcelona, 08916 Barcelona, Spain; (D.F.); (A.H.-R.); (R.M.)
- Correspondence:
| | - Aina Teniente-Serra
- Department of Immunology, Hospital Universitari Germans Trias i Pujol-IGTP, Universitat Autònoma de Barcelona, 08916 Barcelona, Spain; (A.T.-S.); (B.Q.-S.); (E.M.-C.)
| | - Daniel Fuster
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol-IGTP, Universitat Autònoma de Barcelona, 08916 Barcelona, Spain; (D.F.); (A.H.-R.); (R.M.)
| | - Bibiana Quirant-Sánchez
- Department of Immunology, Hospital Universitari Germans Trias i Pujol-IGTP, Universitat Autònoma de Barcelona, 08916 Barcelona, Spain; (A.T.-S.); (B.Q.-S.); (E.M.-C.)
| | - Anna Hernandez-Rubio
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol-IGTP, Universitat Autònoma de Barcelona, 08916 Barcelona, Spain; (D.F.); (A.H.-R.); (R.M.)
| | - Eva Martínez-Cáceres
- Department of Immunology, Hospital Universitari Germans Trias i Pujol-IGTP, Universitat Autònoma de Barcelona, 08916 Barcelona, Spain; (A.T.-S.); (B.Q.-S.); (E.M.-C.)
| | - Roberto Muga
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol-IGTP, Universitat Autònoma de Barcelona, 08916 Barcelona, Spain; (D.F.); (A.H.-R.); (R.M.)
| |
Collapse
|
6
|
Petagine L, Zariwala MG, Patel VB. Alcoholic liver disease: Current insights into cellular mechanisms. World J Biol Chem 2021; 12:87-103. [PMID: 34630912 PMCID: PMC8473419 DOI: 10.4331/wjbc.v12.i5.87] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/20/2021] [Accepted: 08/12/2021] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) due to chronic alcohol consumption is a significant global disease burden and a leading cause of mortality. Alcohol abuse induces a myriad of aberrant changes in hepatocytes at both the cellular and molecular level. Although the disease spectrum of ALD is widely recognized, the precise triggers for disease progression are still to be fully elucidated. Oxidative stress, mitochondrial dysfunction, gut dysbiosis and altered immune system response plays an important role in disease pathogenesis, triggering the activation of inflammatory pathways and apoptosis. Despite many recent clinical studies treatment options for ALD are limited, especially at the alcoholic hepatitis stage. We have therefore reviewed some of the key pathways involved in the pathogenesis of ALD and highlighted current trials for treating patients.
Collapse
Affiliation(s)
- Lucy Petagine
- Center for Nutraceuticals, School of Life Sciences, University of Westminster, London W1W 6UW, United Kingdom
| | - Mohammed Gulrez Zariwala
- Center for Nutraceuticals, School of Life Sciences, University of Westminster, London W1W 6UW, United Kingdom
| | - Vinood B Patel
- Center for Nutraceuticals, School of Life Sciences, University of Westminster, London W1W 6UW, United Kingdom
| |
Collapse
|
7
|
Sakamoto Y, Yoshio S, Doi H, Mori T, Matsuda M, Kawai H, Shimagaki T, Yoshikawa S, Aoki Y, Osawa Y, Yoshida Y, Arai T, Itokawa N, Atsukawa M, Ito T, Honda T, Mise Y, Ono Y, Takahashi Y, Saiura A, Taketomi A, Kanto T. Increased Frequency of Dysfunctional Siglec-7 -CD57 +PD-1 + Natural Killer Cells in Patients With Non-alcoholic Fatty Liver Disease. Front Immunol 2021; 12:603133. [PMID: 33692781 PMCID: PMC7938755 DOI: 10.3389/fimmu.2021.603133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 01/25/2021] [Indexed: 12/16/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a progressive disorder that can develop into liver fibrosis and hepatocellular carcinoma. Natural killer (NK) cells have been shown to protect against liver fibrosis and tumorigenesis, suggesting that they may also play a role in the pathogenesis of NAFLD. Sialic acid-binding immunoglobulin-like lectins (Siglecs) are a family of inhibitory and activating receptors expressed by many cell types, including NK cells. Here, we investigated the phenotypic profiles of peripheral blood and intrahepatic NK cells, including expression of Siglecs and immune checkpoint molecules, and their association with NK cell function in patients with NAFLD. Immune cells in the peripheral blood of 42 patients with biopsy-proven NAFLD and 13 healthy volunteers (HVs) were identified by mass cytometry. The function of various NK cell subpopulations was assessed by flow cytometric detection of intracellular IFN-γ and CD107a/LAMP-1, a degranulation marker, after in vitro stimulation. We found that peripheral blood from NAFLD patients, regardless of fibrosis stage, contained significantly fewer total CD56+ NK cell and CD56dim NK cell populations compared with HVs, and the CD56dim cells from NAFLD patients were functionally impaired. Among the Siglecs examined, NK cells predominantly expressed Siglec-7 and Siglec-9, and both the expression levels of Siglec-7 and Siglec-9 on NK cells and the frequencies of Siglec-7+CD56dim NK cells were reduced in NAFLD patients. Notably, Siglec-7 levels on CD56dim NK cells were inversely correlated with PD-1, CD57, and ILT2 levels and positively correlated with NKp30 and NKp46 levels. Further subtyping of NK cells identified a highly dysfunctional Siglec-7-CD57+PD-1+CD56dim NK cell subset that was increased in patients with NAFLD, even those with mild liver fibrosis. Intrahepatic NK cells from NAFLD patients expressed elevated levels of NKG2D and CD69, suggesting a more activated phenotype than normal liver NK cells. These data identify a close association between NK cell function and expression of Siglec-7, CD57, and PD-1 that could potentially be therapeutically targeted in NAFLD.
Collapse
Affiliation(s)
- Yuzuru Sakamoto
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Gastoenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Sachiyo Yoshio
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroyoshi Doi
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Taizo Mori
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Michitaka Matsuda
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hironari Kawai
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tomonari Shimagaki
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shiori Yoshikawa
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yoshihiko Aoki
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yosuke Osawa
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yuji Yoshida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Taeang Arai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Norio Itokawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Masanori Atsukawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Nippon Medical School, Tokyo, Japan
| | - Takanori Ito
- Division of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Honda
- Division of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Mise
- Department of Hepato-Pancreatic-Biliary Surgery, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoshihiro Ono
- Department of Hepato-Pancreatic-Biliary Surgery, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yu Takahashi
- Department of Hepato-Pancreatic-Biliary Surgery, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akio Saiura
- Department of Hepato-Pancreatic-Biliary Surgery, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akinobu Taketomi
- Department of Gastoenterological Surgery I, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tatsuya Kanto
- Department of Liver Diseases, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Keenan BP, Fong L, Kelley RK. Immunotherapy in hepatocellular carcinoma: the complex interface between inflammation, fibrosis, and the immune response. J Immunother Cancer 2019; 7:267. [PMID: 31627733 PMCID: PMC6798343 DOI: 10.1186/s40425-019-0749-z] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer deaths worldwide and confers a poor prognosis. Beyond standard systemic therapy with multikinase inhibitors, recent studies demonstrate the potential for robust and durable responses from immune checkpoint inhibition in subsets of HCC patients across disease etiologies. The majority of HCC arises in the context of chronic inflammation and from within a fibrotic liver, with many cases associated with hepatitis virus infections, toxins, and fatty liver disease. Many patients also have concomitant cirrhosis which is associated with both local and systemic immune deficiency. Furthermore, the liver is an immunologic organ in itself, which may enhance or suppress the immune response to cancer arising within it. Here, we explore the immunobiology of the liver from its native state to chronic inflammation, fibrosis, cirrhosis and then to cancer, and summarize how this unique microenvironment may affect the response to immunotherapy.
Collapse
Affiliation(s)
- Bridget P Keenan
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Room M1286, 505 Parnassus Ave., San Francisco, CA, 94143, USA. .,Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
| | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Room M1286, 505 Parnassus Ave., San Francisco, CA, 94143, USA.,Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Robin K Kelley
- Division of Hematology/Oncology, Department of Medicine, University of California San Francisco, Room M1286, 505 Parnassus Ave., San Francisco, CA, 94143, USA.,Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
9
|
Matsushita H, Takaki A. Alcohol and hepatocellular carcinoma. BMJ Open Gastroenterol 2019; 6:e000260. [PMID: 31139422 PMCID: PMC6505979 DOI: 10.1136/bmjgast-2018-000260] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/04/2019] [Accepted: 01/12/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Alcohol is classified as a Group 1 carcinogen by the International Agency for Research on Cancer because it induces hepatocellular carcinoma (among other cancers) in humans. An excessive alcohol intake may result in fatty liver, acute/chronic hepatitis, and cirrhosis and eventually lead to hepatocellular carcinoma. It has been reported that alcohol abuse increases the relative risk of hepatocellular carcinoma by 3- to 10-fold. AIM AND METHODS To clarify the known mechanisms of alcohol-related carcinogenesis, we searched Pubmed using the terms alcohol and immune mechanism, alcohol and cancer, and immune mechanism and cancer and summarized the articles as a qualitative review. RESULTS From a clinical perspective, it is well known that alcohol interacts with other factors, such as smoking, viral hepatitis, and diabetes, leading to an increased risk of hepatocellular carcinoma. There are several possible mechanisms through which alcohol may induce liver carcinogenicity, including the mutagenic effects of acetaldehyde and the production of ROS due to the excessive hepatic deposition of iron. Furthermore, it has been reported that alcohol accelerates hepatitis C virus-induced liver tumorigenesis through TLR4 signaling. Despite intense investigations to elucidate the mechanisms, they remain poorly understood. CONCLUSION This review summarizes the recent findings of clinical and pathological studies that have investigated the carcinogenic effects of alcohol in the liver.
Collapse
Affiliation(s)
- Hiroshi Matsushita
- Department of Gastroenterology and Hepatology, Okayama University, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Akinobu Takaki
- Department of Gastroenterology and Hepatology, Okayama University, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
10
|
Liver-resident NK cells suppress autoimmune cholangitis and limit the proliferation of CD4 + T cells. Cell Mol Immunol 2019; 17:178-189. [PMID: 30874628 DOI: 10.1038/s41423-019-0199-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 01/01/2019] [Indexed: 12/13/2022] Open
Abstract
Liver-resident NK cells are distinct from conventional NK cells and play an important role in the maintenance of liver homeostasis. How liver-resident NK cells participate in autoimmune cholangitis remains unclear. Here, we extensively investigated the impact of NK cells in the pathogenesis of autoimmune cholangitis utilizing the well-established dnTGFβRII cholangitis model, NK cell-deficient (Nfil3-/-) mice, adoptive transfer and in vivo antibody-mediated NK cell depletion. Our data demonstrated that disease progression was associated with a significantly reduced frequency of hepatic NK cells. Depletion of NK cells resulted in exacerbated autoimmune cholangitis in dnTGFβRII mice. We further confirmed that the DX5-CD11chi liver-resident NK cell subset colocalized with CD4+ T cells and inhibited CD4+ T cell proliferation. Gene expression microarray analysis demonstrated that liver-resident NK cells had a distinct gene expression pattern consisting of the increased expression of genes involved in negative regulatory functions in the context of the inflammatory microenvironment.
Collapse
|
11
|
Huang H, Lu Y, Zhou T, Gu G, Xia Q. Innate Immune Cells in Immune Tolerance After Liver Transplantation. Front Immunol 2018; 9:2401. [PMID: 30473690 PMCID: PMC6237933 DOI: 10.3389/fimmu.2018.02401] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/27/2018] [Indexed: 12/13/2022] Open
Abstract
Currently, liver transplantation is the most effective treatment for end-stage liver disease. Immunosuppressive agents are required to be taken after the operations, which have significantly reduced rejection rates and improved the short-term (<1 year) survival rates. However, post-transplant complications related to the immunosuppressive therapy have led to the development of new protocols aimed at protecting renal function and preventing de novo cancer and dysmetabolic syndrome. Donor specific immune tolerance, which means the mature immune systems of recipients will not attack the grafts under the conditions without any immunosuppression therapies, is considered the optimal state after liver transplantation. There have been studies that have shown that some patients can reach this immune tolerance state after liver transplantation. The intrahepatic immune system is quite different from that in other solid organs, especially the innate immune system. It contains a variety of liver specific cells, such as liver-derived dendritic cells, Kupffer cells, liver sinusoidal endothelial cells, liver-derived natural killer (NK) cells, natural killer T (NKT) cells, and so on. Depending on their specific structures and functions, these intrahepatic innate immune cells play important roles in the development of intrahepatic immune tolerance. In this article, in order to have a deeper understanding of the tolerogenic functions of liver, we summarized the molecular mechanisms of immune tolerance induced by intrahepatic innate immune cells after liver transplantation.
Collapse
Affiliation(s)
- Hongting Huang
- Department of Hepatic Surgery and Liver Transplantation Center, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yefeng Lu
- Department of Hepatic Surgery and Liver Transplantation Center, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Tao Zhou
- Department of Hepatic Surgery and Liver Transplantation Center, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Guangxiang Gu
- Department of Hepatic Surgery and Liver Transplantation Center, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Qiang Xia
- Department of Hepatic Surgery and Liver Transplantation Center, School of Medicine, Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
12
|
|
13
|
Wahid B, Ali A, Rafique S, Saleem K, Waqar M, Wasim M, Idrees M. Role of altered immune cells in liver diseases: a review. GASTROENTEROLOGIA Y HEPATOLOGIA 2018; 41:377-388. [PMID: 29605453 DOI: 10.1016/j.gastrohep.2018.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 01/12/2018] [Accepted: 01/22/2018] [Indexed: 12/17/2022]
Abstract
Immune cells play an important role in controlling liver tumorigenesis, viral hepatitis, liver fibrosis and contribute to pathogenesis of liver inflammation and injury. Accumulating evidence suggests the effectiveness of natural killer (NK) cells and Kupffer cells (KCs) against viral hepatitis, hepatocellular damage, liver fibrosis, and carcinogenesis. Activation of natural killer cells provides a novel therapeutic strategy to cure liver related diseases. This review discusses the emerging roles of immune cells in liver disorders and it will provide baseline data to scientists to design better therapies for treatment.
Collapse
Affiliation(s)
- Braira Wahid
- Centre for Applied Molecular Biology (CAMB), 87-West Canal Bank Road Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan; Genome Centre for Molecular Based Diagnostics and Research, Al-Sudais Plaza Abdalian Cooperative Society, Lahore, Pakistan
| | - Amjad Ali
- Genome Centre for Molecular Based Diagnostics and Research, Al-Sudais Plaza Abdalian Cooperative Society, Lahore, Pakistan
| | - Shazia Rafique
- Genome Centre for Molecular Based Diagnostics and Research, Al-Sudais Plaza Abdalian Cooperative Society, Lahore, Pakistan
| | - Komal Saleem
- Centre for Applied Molecular Biology (CAMB), 87-West Canal Bank Road Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan; Genome Centre for Molecular Based Diagnostics and Research, Al-Sudais Plaza Abdalian Cooperative Society, Lahore, Pakistan
| | - Muhammad Waqar
- Centre for Applied Molecular Biology (CAMB), 87-West Canal Bank Road Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan; Genome Centre for Molecular Based Diagnostics and Research, Al-Sudais Plaza Abdalian Cooperative Society, Lahore, Pakistan
| | - Muhammad Wasim
- Department of Medicine, Khyber Teaching Hospital Peshawar KPK, Pakistan
| | - Muhammad Idrees
- Centre for Applied Molecular Biology (CAMB), 87-West Canal Bank Road Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan; Genome Centre for Molecular Based Diagnostics and Research, Al-Sudais Plaza Abdalian Cooperative Society, Lahore, Pakistan; Department of Medicine, Khyber Teaching Hospital Peshawar KPK, Pakistan; Division of Molecular Virology and Diagnostics Center of Excellence in Molecular Biology (CEMB), 87-West Canal Bank Road Thokar Niaz Baig, University of the Punjab, Lahore, Pakistan; Vice Chancellor Hazara University Mansehra, Pakistan.
| |
Collapse
|
14
|
Hepatic Immune Microenvironment in Alcoholic and Nonalcoholic Liver Disease. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6862439. [PMID: 28852648 PMCID: PMC5567444 DOI: 10.1155/2017/6862439] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/06/2017] [Indexed: 01/18/2023]
Abstract
Many types of innate (natural killer cells, natural killer T cells, and Kupffer cells/macrophages) and adaptive (T cells and B cells) immune cells are enriched within the liver and function in liver physiology and pathology. Liver pathology is generally induced by two types of immunologic insults: failure to eliminate antigens derived from the gastrointestinal tract which are important for host defense and an impaired tissue protective tolerance mechanism that helps reduce the negative outcomes of immunopathology. Accumulating evidence from the last several decades suggests that hepatic immune cells play an important role in the pathogenesis of alcoholic and nonalcoholic liver injury and inflammation in humans and mice. Here, we focus on the roles of innate and adaptive immune cells in the development and maintenance of alcoholic liver disease and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Additionally, the pathogenesis of liver disease and new therapeutic targets for preventing and treating alcoholic liver disease and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis are discussed.
Collapse
|
15
|
Jamil KM, Hydes TJ, Cheent KS, Cassidy SA, Traherne JA, Jayaraman J, Trowsdale J, Alexander GJ, Little AM, McFarlane H, Heneghan MA, Purbhoo MA, Khakoo SI. STAT4-associated natural killer cell tolerance following liver transplantation. Gut 2017; 66:352-361. [PMID: 26887815 PMCID: PMC5284485 DOI: 10.1136/gutjnl-2015-309395] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 09/29/2015] [Accepted: 10/20/2015] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Natural killer (NK) cells are important mediators of liver inflammation in chronic liver disease. The aim of this study was to investigate why liver transplants (LTs) are not rejected by NK cells in the absence of human leukocyte antigen (HLA) matching, and to identify a tolerogenic NK cell phenotype. DESIGN Phenotypic and functional analyses on NK cells from 54 LT recipients were performed, and comparisons made with healthy controls. Further investigation was performed using gene expression analysis and donor:recipient HLA typing. RESULTS NK cells from non-HCV LT recipients were hypofunctional, with reduced expression of NKp46 (p<0.05) and NKp30 (p<0.001), reduced cytotoxicity (p<0.001) and interferon (IFN)-γ secretion (p<0.025). There was no segregation of this effect with HLA-C, and these functional changes were not observed in individuals with HCV. Microarray and RT-qPCR analysis demonstrated downregulation of STAT4 in NK cells from LT recipients (p<0.0001). Changes in the expression levels of the transcription factors Helios (p=0.06) and Hobit (p=0.07), which control NKp46 and IFNγ expression, respectively, were also detected. Hypofunctionality of NK cells was associated with impaired STAT4 phosphorylation and downregulation of the STAT4 target microRNA-155. Conversely in HCV-LT NK cell tolerance was reversed, consistent with the more aggressive outcome of LT for HCV. CONCLUSIONS LT is associated with transcriptional and functional changes in NK cells, resulting in reduced activation. NK cell tolerance occurs upstream of major histocompatibility complex (MHC) class I mediated education, and is associated with deficient STAT4 phosphorylation. STAT4 therefore represents a potential therapeutic target to induce NK cell tolerance in liver disease.
Collapse
Affiliation(s)
- K M Jamil
- Department of Hepatology, Imperial College, London, UK
| | - T J Hydes
- Department of Hepatology, Southampton University, Southampton, UK
| | - K S Cheent
- Department of Hepatology, Imperial College, London, UK
| | - S A Cassidy
- Department of Hepatology, Imperial College, London, UK
| | - J A Traherne
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - J Jayaraman
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - J Trowsdale
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - G J Alexander
- Department of Hepatology, Addenbrookes Hospital, Cambridge, UK
| | - A-M Little
- Histocompatibility and Immunogenetics Service, Gartnavel General Hospital, Glasgow, UK
| | - H McFarlane
- Histocompatibility and Immunogenetics Service, Gartnavel General Hospital, Glasgow, UK
| | - M A Heneghan
- Institute of Liver Studies, Kings College Hospital London, London, UK
| | - M A Purbhoo
- Department of Hepatology, Imperial College, London, UK
| | - S I Khakoo
- Department of Hepatology, Imperial College, London, UK.,Department of Hepatology, Southampton University, Southampton, UK
| |
Collapse
|
16
|
Zuluaga P, Sanvisens A, Teniente A, Fuster D, Tor J, Martínez-Cáceres E, Muga R. Wide array of T-cell subpopulation alterations in patients with alcohol use disorders. Drug Alcohol Depend 2016; 162:124-9. [PMID: 27038675 DOI: 10.1016/j.drugalcdep.2016.02.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/12/2016] [Accepted: 02/27/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Alcohol abuse impacts innate and adaptive immunity and predisposes to infections. However, prevalence and correlations of cellular immune alterations in large case series is underreported. We aimed to analyze quantitative alterations of T-lymphocyte subpopulations in patients with alcohol use disorder (AUD). METHODS cross-sectional study in patients admitted for detoxification between January 1, 2002 and December 31, 2012. Socio-demographic and alcohol use characteristics and blood samples for biochemistry, hematology and immune phenotype was obtained at admission. RESULTS 238 patients (79.8%M) were eligible; age at admission was 43 years (interquartile range [IQR]: 38-51 years), the amount of alcohol consumption was 180 g/day (IQR: 120-200 g/day) and median duration of AUD was 18 years (IQR: 9-25 years). Compared to healthy individuals, 50% of patients had significantly fewer double-negative (DN) T-lymphocytes (<34 × 10(9)/L) and 23% had more double-positive (DP) T-cells (>52 × 10(9)/L). In addition, 24% of patients had high number of CD8(+) cells (>735 × 10(9)/L) and 13% had low CD4(+) cell counts (<600 × 10(9)/L). In multivariable analysis, age, sex, serum albumin, and current cocaine use were predictors of T-cell subpopulation alterations. Women were three-times (OR=3.5, 95%CI:1.3-9.5) more likely to present with higher DP T-lymphocytes than men. CONCLUSIONS Quantitative alterations of T-cell subpopulations are frequent in patients seeking treatment of AUD. Assessment of cellular immunity in this population may help to identify those at increased risk of immune alterations.
Collapse
Affiliation(s)
- Paola Zuluaga
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Arantza Sanvisens
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Aina Teniente
- Department of Immunology, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Daniel Fuster
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Jordi Tor
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Eva Martínez-Cáceres
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Roberto Muga
- Department of Internal Medicine, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain.
| |
Collapse
|
17
|
Seo W, Jeong WI. Hepatic non-parenchymal cells: Master regulators of alcoholic liver disease? World J Gastroenterol 2016; 22:1348-1356. [PMID: 26819504 PMCID: PMC4721970 DOI: 10.3748/wjg.v22.i4.1348] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 09/28/2015] [Accepted: 12/14/2015] [Indexed: 02/06/2023] Open
Abstract
Chronic alcohol consumption is one of the most common causes of the progression of alcoholic liver disease (ALD). In the past, alcohol-mediated hepatocyte injury was assumed to be a significantly major cause of ALD. However, a huge number of recent and brilliant studies have demonstrated that hepatic non-parenchymal cells including Kupffer cells, hepatic stellate cells, liver sinusoidal endothelial cells and diverse types of lymphocytes play crucial roles in the pathogenesis of ALD by producing inflammatory mediators such as cytokines, oxidative stress, microRNA, and lipid-originated metabolites (retinoic acid and endocannabinoids) or by directly interacting with parenchymal cells (hepatocytes). Therefore, understanding the comprehensive roles of hepatic non-parenchymal cells during the development of ALD will provide new integrative directions for the treatment of ALD. This review will address the roles of non-parenchymal cells in alcoholic steatosis, inflammation, and liver fibrosis and might help us to discover possible therapeutic targets and treatments involving modulating the non-parenchymal cells in ALD.
Collapse
|
18
|
Zaldivar Fujigaki JL, Arroyo Valerio AG, López Alvarenga JC, Gutiérrez Reyes EG, Kershenobich D, Hernández Ruiz J. Alterations in Activation, Cytotoxic Capacity and Trafficking Profile of Peripheral CD8 T Cells in Young Adult Binge Drinkers. PLoS One 2015; 10:e0132521. [PMID: 26151816 PMCID: PMC4494878 DOI: 10.1371/journal.pone.0132521] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/15/2015] [Indexed: 12/14/2022] Open
Abstract
Background Excess of alcohol consumption is a public health problem and has documented effects on the immune system of humans and animals. Animal and in vitro studies suggest that alcohol abuse changes CD8 T cell (CD8) characteristics, however it remains unknown if the CD8 profile of binge drinkers is different in terms of activation, trafficking and cytotoxic capacity. Aim To analyze the peripheral CD8 cytotoxic capacity, activation and trafficking phenotypic profile of Mexican young adults with regard to alcohol consumption pattern. Methods 55 Mexican young adults were stratified as Light (20), Intermediate (18) or Binge drinkers (17) according to their reported alcohol consumption pattern. Blood samples were obtained and hematic biometry and liver enzyme analysis were performed. Peripheral CD8 profile was established by expression of Granzyme B (GB), CD137, CD127, CD69, TLR4, PD1, CCR2, CCR4, CCR5 and CXCR4 by FACS. Data was analyzed by ANOVA, posthoc DMS and Tamhane, and principal component analysis (PCA) with varimax rotation, p<0.05. Results The Binge drinking group showed increased γGT together with increased expression of CD69 and reduced expression of TLR4, PD1, CCR2 and CXCR4 in peripheral CD8 cells. Other parameters were also specific to Binge drinkers. PCA established 3 factors associated with alcohol consumption: “Early Activation” represented by CD69 and TLR4 expression in the CD8 population; “Effector Activation” by CD69 expression in CD8 CD127+CD137+ and CD8 CD25+ CD137+; and Trafficking by CXCR4 expression on total CD8 and CD8 GB+CXCR4+, and CCR2 expression on total CD8. Binge drinking pattern showed low expression of Early Activation and Trafficking factors while Light drinking pattern exhibited high expression of Effector Activation factor. Conclusions Alcohol consumption affects the immune phenotype of CD8 cells since binge drinking pattern was found to be associated with high CD69 and low TLR4, CXCR4 and CCR2 expression, which suggest recent activation, decreased sensitivity to LPS and lower migration capacity in response to chemokines SDF-1 and MCP-1. These results indicate that a binge-drinking pattern of alcohol consumption may induce an altered immune profile that could be related with liver damage and the increased susceptibility to infection reported to this behavior.
Collapse
Affiliation(s)
- José Luis Zaldivar Fujigaki
- Laboratory of Liver, Pancreas and Motility, Department of Experimental Medicine, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | - Esperanza Gabriela Gutiérrez Reyes
- Laboratory of Liver, Pancreas and Motility, Department of Experimental Medicine, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - David Kershenobich
- Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Mexico City, Mexico
| | - Joselin Hernández Ruiz
- Laboratory of Liver, Pancreas and Motility, Department of Experimental Medicine, Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Dirección de Investigación, Hospital General de México "Dr. Eduardo Liceaga", Mexico City, Mexico
- * E-mail:
| |
Collapse
|
19
|
Almeida J, Polvorosa MA, Gonzalez-Quintela A, Madruga I, Marcos M, Pérez-Nieto MA, Hernandez-Cerceño ML, Orfao A, Laso FJ. Altered Distribution of Peripheral Blood Maturation-Associated B-Cell Subsets in Chronic Alcoholism. Alcohol Clin Exp Res 2015; 39:1476-84. [PMID: 26146763 DOI: 10.1111/acer.12783] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/14/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although decreased counts of peripheral blood (PB) B cells-associated with an apparently contradictory polyclonal hypergammaglobulinemia-have been reported in chronic alcoholism, no information exists about the specific subsets of circulating B cells altered and their relationship with antibody production. Here, we analyzed for the first time the distribution of multiple maturation-associated subpopulations of PB B cells in alcoholism and its potential relationship with the onset of liver disease. METHODS PB samples from 35 male patients-20 had alcoholic hepatitis (AH) and 15 chronic alcoholism without liver disease (AWLD)-were studied, in parallel to 19 male healthy donors (controls). The distribution of PB B-cell subsets (immature/regulatory, naïve, CD27(-) and CD27(+) memory B lymphocytes, and circulating plasmablasts of distinct immunoglobulin-Ig-isotypes) was analyzed by flow cytometry. RESULTS Patients with AH showed significantly decreased numbers of total PB B lymphocytes (vs. controls and AWLD), at the expense of immature, memory, and, to a lesser extent, also naïve B cells. AWLD showed reduced numbers of immature and naïve B cells (vs. controls), but higher PB counts of plasmablasts (vs. the other 2 groups). Although PB memory B cells were reduced among the patients, the percentage of surface (s)IgA(+) cells (particularly CD27(-) /sIgA(+) cells) was increased in AH, whereas both sIgG(+) and sIgA(+) memory B cells were significantly overrepresented in AWLD versus healthy donors. Regarding circulating plasmablasts, patients with AH only showed significantly reduced counts of sIgG(+) cells versus controls. In contrast, the proportion of both sIgA(+) and sIgG(+) plasmablasts-from all plasmablasts-was reduced in AH and increased in AWLD (vs. the other 2 groups). CONCLUSIONS AH and AWLD patients display a significantly reduced PB B-cell count, at the expense of decreased numbers of recently produced immature/regulatory B cells and naïve B cells, together with an increase in Ig-switched memory B lymphocytes and plasmablasts, particularly of IgA(+) cells.
Collapse
Affiliation(s)
- Julia Almeida
- Instituto de Biologia Molecular y Celular del Cancer, Centro de Investigacion del Cancer/IBMCC (CSIC-USAL), Institute of Biomedical Research of Salamanca (IBSAL), Cytometry Service, Salamanca, Spain.,Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Maria Angeles Polvorosa
- Alcoholism Unit, Department of Internal Medicine, University Hospital of Salamanca, Salamanca, Spain.,Department of Medicine, IBSAL, University of Salamanca, Salamanca, Spain
| | - Arturo Gonzalez-Quintela
- Department of Internal Medicine, University Hospital of Santiago de Compostela, Santiago de Compostela, Spain
| | - Ignacio Madruga
- Alcoholism Unit, Department of Internal Medicine, University Hospital of Salamanca, Salamanca, Spain.,Department of Medicine, IBSAL, University of Salamanca, Salamanca, Spain
| | - Miguel Marcos
- Alcoholism Unit, Department of Internal Medicine, University Hospital of Salamanca, Salamanca, Spain.,Department of Medicine, IBSAL, University of Salamanca, Salamanca, Spain
| | - Maria Angeles Pérez-Nieto
- Alcoholism Unit, Department of Internal Medicine, University Hospital of Salamanca, Salamanca, Spain.,Department of Medicine, IBSAL, University of Salamanca, Salamanca, Spain
| | | | - Alberto Orfao
- Instituto de Biologia Molecular y Celular del Cancer, Centro de Investigacion del Cancer/IBMCC (CSIC-USAL), Institute of Biomedical Research of Salamanca (IBSAL), Cytometry Service, Salamanca, Spain.,Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Francisco Javier Laso
- Alcoholism Unit, Department of Internal Medicine, University Hospital of Salamanca, Salamanca, Spain.,Department of Medicine, IBSAL, University of Salamanca, Salamanca, Spain
| |
Collapse
|
20
|
Zhang H, Zhang F, Zhu Z, Luong D, Meadows GG. Chronic alcohol consumption enhances iNKT cell maturation and activation. Toxicol Appl Pharmacol 2014; 282:139-50. [PMID: 25499027 DOI: 10.1016/j.taap.2014.11.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/13/2014] [Accepted: 11/25/2014] [Indexed: 01/16/2023]
Abstract
Alcohol consumption exhibits diverse effects on different types of immune cells. NKT cells are a unique T cell population and play important immunoregulatory roles in different types of immune responses. The effects of chronic alcohol consumption on NKT cells remain to be elucidated. Using a mouse model of chronic alcohol consumption, we found that alcohol increases the percentage of NKT cells, especially iNKT cells in the thymus and liver, but not in the spleen or blood. Alcohol consumption decreases the percentage of NK1.1(-) iNKT cells in the total iNKT cell population in all of the tissues and organs examined. In the thymus, alcohol consumption increases the number of NK1.1(+)CD44(hi) mature iNKT cells but does not alter the number of NK1.1(-) immature iNKT cells. A BrdU incorporation assay shows that alcohol consumption increases the proliferation of thymic NK1.1(-) iNKT cells, especially the NK1.1(-)CD44(lo) Stage I iNKT cells. The percentage of NKG2A(+) iNKT cells increases in all of the tissues and organs examined; whereas CXCR3(+) iNKT cells only increases in the thymus of alcohol-consuming mice. Chronic alcohol consumption increases the percentage of IFN-γ-producing iNKT cells and increases the blood concentration of IFN-γ and IL-12 after in vivo α-galactosylceramide (αGalCer) stimulation. Consistent with the increased cytokine production, the in vivo activation of iNKT cells also enhances the activation of dendritic cells (DC) and NK, B, and T cells in the alcohol-consuming mice. Taken together the data indicate that chronic alcohol consumption enhances iNKT cell maturation and activation, which favors the Th1 immune response.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA 99210-1495, USA.
| | - Faya Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA 99210-1495, USA
| | - Zhaohui Zhu
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA 99210-1495, USA
| | - Dung Luong
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA 99210-1495, USA
| | - Gary G Meadows
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA 99210-1495, USA
| |
Collapse
|
21
|
González-Reimers E, Santolaria-Fernández F, Martín-González MC, Fernández-Rodríguez CM, Quintero-Platt G. Alcoholism: A systemic proinflammatory condition. World J Gastroenterol 2014; 20:14660-14671. [PMID: 25356029 PMCID: PMC4209532 DOI: 10.3748/wjg.v20.i40.14660] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Excessive ethanol consumption affects virtually any organ, both by indirect and direct mechanisms. Considerable research in the last two decades has widened the knowledge about the paramount importance of proinflammatory cytokines and oxidative damage in the pathogenesis of many of the systemic manifestations of alcoholism. These cytokines derive primarily from activated Kupffer cells exposed to Gram-negative intestinal bacteria, which reach the liver in supra-physiological amounts due to ethanol-mediated increased gut permeability. Reactive oxygen species (ROS) that enhance the inflammatory response are generated both by activation of Kupffer cells and by the direct metabolic effects of ethanol. The effects of this increased cytokine secretion and ROS generation lie far beyond liver damage. In addition to the classic consequences of endotoxemia associated with liver cirrhosis that were described several decades ago, important research in the last ten years has shown that cytokines may also induce damage in remote organs such as brain, bone, muscle, heart, lung, gonads, peripheral nerve, and pancreas. These effects are even seen in alcoholics without significant liver disease. Therefore, alcoholism can be viewed as an inflammatory condition, a concept which opens the possibility of using new therapeutic weapons to treat some of the complications of this devastating and frequent disease. In this review we examine some of the most outstanding consequences of the altered cytokine regulation that occurs in alcoholics in organs other than the liver.
Collapse
|
22
|
Giannitrapani L, Soresi M, Balasus D, Licata A, Montalto G. Genetic association of interleukin-6 polymorphism (-174 G/C) with chronic liver diseases and hepatocellular carcinoma. World J Gastroenterol 2013; 19:2449-2455. [PMID: 23674845 PMCID: PMC3646134 DOI: 10.3748/wjg.v19.i16.2449] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2012] [Revised: 03/06/2013] [Accepted: 03/22/2013] [Indexed: 02/06/2023] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine which is expressed in many inflammatory cells in response to different types of stimuli, regulating a number of biological processes. The IL-6 gene is polymorphic in both the 5' and 3' flanking regions and more than 150 single nucleotide polymorphisms have been identified so far. Genetic polymorphisms of IL-6 may affect the outcomes of several diseases, where the presence of high levels of circulating IL-6 have been correlated to the stage and/or the progression of the disease itself. The -174 G/C polymorphism is a frequent polymorphism, that is located in the upstream regulatory region of the IL-6 gene and affects IL-6 production. However, the data in the literature on the genetic association between the -174 G/C polymorphism and some specific liver diseases characterized by different etiologies are still controversial. In particular, most of the studies are quite unanimous in describing a correlation between the presence of the high-producer genotype and a worse evolution of the chronic liver disease. This is valid for patients with hepatitis C virus (HCV)-related chronic hepatitis and liver cirrhosis and hepatocellular carcinoma (HCC) whatever the etiology. Studies in hepatitis B virus-related chronic liver diseases are not conclusive, while specific populations like non alcoholic fatty liver disease/non-alcoholic steatohepatitis, autoimmune and human immunodeficiency virus/HCV co-infected patients show a higher prevalence of the low-producer genotype, probably due to the complexity of these clinical pictures. In this direction, a systematic revision of these data should shed more light on the role of this polymorphism in chronic liver diseases and HCC.
Collapse
MESH Headings
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Fatty Liver/genetics
- Fatty Liver/immunology
- Genetic Predisposition to Disease
- Hepatitis B, Chronic/genetics
- Hepatitis B, Chronic/immunology
- Hepatitis C, Chronic/genetics
- Hepatitis C, Chronic/immunology
- Hepatitis, Autoimmune/genetics
- Hepatitis, Autoimmune/immunology
- Humans
- Interleukin-6/genetics
- Liver Cirrhosis/genetics
- Liver Cirrhosis/immunology
- Liver Diseases, Alcoholic/genetics
- Liver Diseases, Alcoholic/immunology
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Non-alcoholic Fatty Liver Disease
- Phenotype
- Polymorphism, Single Nucleotide
- Risk Factors
Collapse
|
23
|
Tian Z, Chen Y, Gao B. Natural killer cells in liver disease. Hepatology 2013; 57:1654-62. [PMID: 23111952 PMCID: PMC3573257 DOI: 10.1002/hep.26115] [Citation(s) in RCA: 227] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 10/01/2012] [Accepted: 10/12/2012] [Indexed: 12/12/2022]
Abstract
Natural killer (NK) cells are enriched in lymphocytes within the liver and have unique phenotypic features and functional properties, including tumor necrosis factor-related apoptosis-inducing ligand-dependent cytotoxicity and specific cytokine profiles. As a key component of innate immunity in the liver, NK cells perform critical roles in host defense against pathogens and tumors through their natural cytotoxicity and cytokine production, and they also act as regulatory cells by engaging in reciprocal interactions with other types of liver cells through cell-to-cell contact and the production of cytokines. Accumulating evidence from the last decade suggests that NK cells play an important role in controlling viral hepatitis, liver fibrosis, and liver tumorigenesis, but also contribute to the pathogenesis of liver injury and inflammation. The characterization of intrahepatic NK cell functions has not only helped us to better understand the pathogenesis of liver disease, but has also revealed new therapeutic targets for managing this disease.
Collapse
Affiliation(s)
- Zhigang Tian
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yongyan Chen
- Department of Microbiology and Immunology, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
24
|
Almeida J, Polvorosa MA, Gonzalez-Quintela A, Marcos M, Pastor I, Hernandez Cerceño ML, Orfao A, Laso FJ. Decreased peripheral blood CD4+/CD25+ regulatory T cells in patients with alcoholic hepatitis. Alcohol Clin Exp Res 2013; 37:1361-9. [PMID: 23550693 DOI: 10.1111/acer.12095] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 01/09/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Development of alcoholic hepatitis (AH) may be favored by the activation of the innate immune response. Recently, decreased numbers of circulating regulatory T cells (Tregs) have been reported in diseases associated with an immune activation status, but no studies have focused so far, in investigating the distribution of Tregs in chronic alcoholism and its potential association with liver disease. Here, we analyzed for the first time the frequency of peripheral blood (PB) Tregs and Treg subsets in AH and its relationship with the production of inflammatory cytokines by PB monocytes and dendritic cells (DCs). METHODS PB samples from 25 male patients with AH were studied; in parallel, 15 male chronic alcoholic patients without liver disease (AWLD) and 17 male healthy donors were also studied, as controls. The distribution of CD4⁺CD25hiCD127-/lo Tregs and their maturation subsets (naïve, central memory, and peripheral memory Tregs) was analyzed by flow cytometry. Spontaneous and in vitro-stimulated production of inflammatory cytokines by PB monocytes and DCs was analyzed by flow cytometry at the cytoplasmic level. RESULTS Patients with AH showed decreased (p < 0.05) numbers of PB CD4⁺CD25hiCD127-/lo Tregs at the expense of all maturation-associated subsets, while AWLD and healthy subjects showed a similar (p > 0.05) distribution of PB CD4⁺CD25hiCD127-/lo Tregs. Interestingly, significantly increased amounts of spontaneously produced inflammatory cytokines were found among circulating monocyte-derived DCs and monocytes from AH (and AWLD) patients in comparison with healthy donors. Conversely, the ability of these cell subsets to produce cytokines after in vitro stimulation was lower (p < 0.05) in AH versus the 2 control groups. CONCLUSIONS PB CD4⁺CD25hiCD127-/lo Tregs are significantly decreased in patients with AH when compared to both healthy and AWLD; this may contribute to explain the more pronounced activation of the innate immune response observed in AH, as reflected by an increased secretion of inflammatory cytokines by PB DCs and monocytes, and could facilitate the development of liver disease.
Collapse
Affiliation(s)
- Julia Almeida
- Cancer Research Center, (IBMCC; CSIC-USAL), IBSAL and Cytometry Service, University of Salamanca, Salamanca, Spain
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Zhang HM, Nie JS, Li X, Niu Q. Characteristic analysis of peripheral blood mononuclear cell apoptosis in coke oven workers. J Occup Health 2011; 54:44-50. [PMID: 22186298 DOI: 10.1539/joh.11-0155-oa] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES The aim of the present study was to determine the peripheral blood mononuclear cell (PBMC) apoptosis in coke oven workers so that we can take effective measures to protect coke oven workers. METHODS The subjects, 129 coke oven workers and 37 warehouse workers (controls), were investigated using a questionnaire to collect information about their age, working years, smoking and drinking habits, vocational history and other general information. The coke oven workers were divided into the oven-bottom group (34), oven-side group (48) and oven-top group (47) according to their working sites and environmental monitoring data. The concentration of benzo[a]pyrene (B[a]P) and the subjects' urinary 1-hydroxypyrene (1-OH-Py) levels were determined by HPLC. Additionally, the PBMCs were separated from blood samples, and the early and late apoptosis rates were determined by flow cytometry. RESULTS The airborne B[a]P concentrations were 19.5 ± 13.2, 185.9 ± 38.6 and 1,623.5 ± 435.8 ng/m(3) at the bottom, side and top of the oven, respectively, and were higher than in the controls' workplaces 10.2 ± 7.6 ng/m(3). Urinary 1-OH-Py, indicating the B[a]P's internal exposure level, was significantly higher in the exposed groups than in the controls (p<0.05). Compared with the controls, the coke oven workers' PBMC apoptosis rates were significantly increased and increased in association with the B[a]P level. PBMC apoptosis increased in association with the 1-OH-Py level and coking operation years and decreased in association with years of alcohol consumption. CONCLUSIONS PBMC apoptosis in the coke oven workers was associated with the 1-OH-Py level, coke operation years and years of alcohol consumption and may be induced by B[a]P.
Collapse
Affiliation(s)
- Hong Mei Zhang
- Department of Occupational and Environmental Health, Shanxi Medical University
| | | | | | | |
Collapse
|
26
|
Chandrasekaran K, Swaminathan K, Kumar SM, Chatterjee S, Clemens DL, Dey A. Elevated glutathione level does not protect against chronic alcohol mediated apoptosis in recombinant human hepatoma cell line VL-17A over-expressing alcohol metabolizing enzymes--alcohol dehydrogenase and Cytochrome P450 2E1. Toxicol In Vitro 2011; 25:969-78. [PMID: 21414402 DOI: 10.1016/j.tiv.2011.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 02/04/2011] [Accepted: 03/09/2011] [Indexed: 12/11/2022]
Abstract
Chronic consumption of alcohol leads to liver injury. Ethanol-inducible Cytochrome P450 2E1 (CYP2E1) plays a critical role in alcohol mediated oxidative stress due to its ability to metabolize ethanol. In the present study, using the recombinant human hepatoma cell line VL-17A that over-expresses the alcohol metabolizing enzymes-alcohol dehydrogenase (ADH) and CYP2E1; and control HepG2 cells, the mechanism and mode of cell death due to chronic ethanol exposure were studied. Untreated VL-17A cells exhibited apoptosis and oxidative stress when compared with untreated HepG2 cells. Chronic alcohol exposure, i.e., 100 mM ethanol treatment for 72 h caused a significant decrease in viability (47%) in VL-17A cells but not in HepG2 cells. Chronic ethanol mediated cell death in VL-17A cells was predominantly apoptotic, with increased oxidative stress as the underlying mechanism. Chronic ethanol exposure of VL-17A cells resulted in 1.1- to 2.5-fold increased levels of ADH and CYP2E1. Interestingly, the level of the antioxidant GSH was found to be 3-fold upregulated in VL-17A cells treated with ethanol, which may be a metabolic adaptation to the persistent and overwhelming oxidative stress. In conclusion, the increased GSH level may not be sufficient enough to protect VL-17A cells from chronic alcohol mediated oxidative stress and resultant apoptosis.
Collapse
Affiliation(s)
- Karthikeyan Chandrasekaran
- Life Science Division, AU-KBC Research Centre, MIT Campus of Anna University, Chromepet, Chennai 600 044, India
| | | | | | | | | | | |
Collapse
|