1
|
Abrol S, Tandon M, Raghu AM, Pandey C. Role of Preoperative Neutrophil-Lymphocyte Ratio in Predicting Prognosis After Liver Transplantation for Chronic Liver Failure. Cureus 2025; 17:e80749. [PMID: 40114845 PMCID: PMC11922682 DOI: 10.7759/cureus.80749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 03/22/2025] Open
Abstract
INTRODUCTION The neutrophil-lymphocyte ratio (NLR) is an easily calculable biomarker known to have a predictive value in cardiac disease, malignancy, and renal failure. However, it has not been studied before in chronic liver disease patients undergoing liver transplantation. We aimed to evaluate the role of the pre-transplantation NLR in predicting the prognosis of patients with chronic liver failure undergoing liver transplantation. METHOD Data was retrospectively collected from 46 patients with chronic liver disease who underwent liver transplantation. The patients were divided into two groups. Group A had 23 patients who survived after liver transplantation. Group B had 23 patients who did not survive. NLR was calculated by dividing the percentage of neutrophils by the percentage of lymphocytes in peripheral blood. The NLR cut-off value was based on a receiver operating characteristic curve analysis. Postoperative complications were also noted. RESULTS Preoperative NLR of 3.46 can predict post-transplantation mortality, with the area under the curve (AUC) of 0.86, having a sensitivity of 86.96% and a specificity of 73.91%. NLR emerged as an independent predictor of mortality (hazard ratio (HR) = 4.1, p = 0.028) after adjusting for the Model for End-Stage Liver Disease-Sodium (MELD-Na), creatinine, and neutrophil count. A rising NLR trend was significantly associated with the development of postoperative complications like neurological disease (p < 0.001), coagulopathy (p = 0.004), and acute kidney injury (p = 0.043). CONCLUSION A high preoperative NLR is a predictor of poor outcomes in liver transplantation patients with chronic liver disease.
Collapse
Affiliation(s)
- Surbhi Abrol
- Department of Anesthesia and Critical Care, Northampton General Hospital, Northampton, GBR
| | - Manish Tandon
- Department of Anesthesiology, Dharamshila Narayana Superspeciality Hospital, Delhi, IND
| | - Arun M Raghu
- Department of Anesthesiology and Transplant Anesthesiology, Gleneagles Global Hospital, Bangalore, IND
| | | |
Collapse
|
2
|
Zhao Q, Chen DP, Chen HD, Wang YZ, Shi W, Lu YT, Ren YZ, Wu YK, Pang YH, Deng H, He X, Kuang DM, Guo ZY. NK-cell-elicited gasdermin-D-dependent hepatocyte pyroptosis induces neutrophil extracellular traps that facilitate HBV-related acute-on-chronic liver failure. Hepatology 2025; 81:917-931. [PMID: 38537134 DOI: 10.1097/hep.0000000000000868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/27/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND AND AIMS HBV infection is a major etiology of acute-on-chronic liver failure (ACLF). At present, the pattern and regulation of hepatocyte death during HBV-ACLF progression are still undefined. Evaluating the mode of cell death and its inducers will provide new insights for developing therapeutic strategies targeting cell death. In this study, we aimed to elucidate whether and how immune landscapes trigger hepatocyte death and lead to the progression of HBV-related ACLF. APPROACH AND RESULTS We identified that pyroptosis represented the main cell death pattern in the liver of patients with HBV-related ACLF. Deficiency of MHC-I in HBV-reactivated hepatocytes activated cytotoxic NK cells, which in turn operated in a perforin/granzyme-dependent manner to trigger GSDMD/caspase-8-dependent pyroptosis of hepatocytes. Neutrophils selectively accumulated in the pyroptotic liver, and HMGB1 derived from the pyroptotic liver constituted an important factor triggering the generation of pathogenic extracellular traps in neutrophils (NETs). Clinically, elevated plasma levels of myeloperoxidase-DNA complexes were a promising prognostic biomarker for HBV-related ACLF. More importantly, targeting GSDMD pyroptosis-HMGB1 release in the liver abrogates NETs that intercept the development of HBV-related ACLF. CONCLUSIONS Studying the mechanisms that selectively modulate GSDMD-dependent pyroptosis, as well as its immune landscapes, will provide a novel strategy for restoring the liver function of patients with HBV-related ACLF.
Collapse
Affiliation(s)
- Qiang Zhao
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong-Ping Chen
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Hua-Di Chen
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying-Zhe Wang
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wei Shi
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yi-Tong Lu
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yi-Zheng Ren
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yuan-Kai Wu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Department of Infectious Diseases, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi-Hua Pang
- Guangdong Provincial Key Laboratory of Liver Disease Research, Department of Infectious Diseases, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong Deng
- Guangdong Provincial Key Laboratory of Liver Disease Research, Department of Infectious Diseases, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoshun He
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
- NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Dong-Ming Kuang
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Yong Guo
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
- NHC key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| |
Collapse
|
3
|
He W, Yan L, Hu D, Hao J, Liou Y, Luo G. Neutrophil heterogeneity and plasticity: unveiling the multifaceted roles in health and disease. MedComm (Beijing) 2025; 6:e70063. [PMID: 39845896 PMCID: PMC11751288 DOI: 10.1002/mco2.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/04/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
Neutrophils, the most abundant circulating leukocytes, have long been recognized as key players in innate immunity and inflammation. However, recent discoveries unveil their remarkable heterogeneity and plasticity, challenging the traditional view of neutrophils as a homogeneous population with a limited functional repertoire. Advances in single-cell technologies and functional assays have revealed distinct neutrophil subsets with diverse phenotypes and functions and their ability to adapt to microenvironmental cues. This review provides a comprehensive overview of the multidimensional landscape of neutrophil heterogeneity, discussing the various axes along which diversity manifests, including maturation state, density, surface marker expression, and functional polarization. We highlight the molecular mechanisms underpinning neutrophil plasticity, focusing on the complex interplay of signaling pathways, transcriptional regulators, and epigenetic modifications that shape neutrophil responses. Furthermore, we explore the implications of neutrophil heterogeneity and plasticity in physiological processes and pathological conditions, including host defense, inflammation, tissue repair, and cancer. By integrating insights from cutting-edge research, this review aims to provide a framework for understanding the multifaceted roles of neutrophils and their potential as therapeutic targets in a wide range of diseases.
Collapse
Affiliation(s)
- Weifeng He
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Lingfeng Yan
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| | - Dongxue Hu
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and TreatmentZhuhai Institute of Translational MedicineZhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University)Jinan UniversityZhuhaiGuangdongChina
- The Biomedical Translational Research InstituteFaculty of Medical ScienceJinan UniversityGuangzhouGuangdongChina
| | - Yih‐Cherng Liou
- Department of Biological SciencesFaculty of ScienceNational University of SingaporeSingaporeSingapore
- National University of Singapore (NUS) Graduate School for Integrative Sciences and EngineeringNational University of SingaporeSingaporeSingapore
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of Trauma and Chemical Poisoningthe First Affiliated Hospital of Army Medical University (the Third Military Medical University)ChongqingChina
- Chongqing Key Laboratory for Wound Repair and Tissue RegenerationChongqingChina
| |
Collapse
|
4
|
Chen L, Elizalde M, Alvarez-Sola G. The Role of Sulfatides in Liver Health and Disease. FRONT BIOSCI-LANDMRK 2025; 30:25077. [PMID: 39862071 DOI: 10.31083/fbl25077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 01/27/2025]
Abstract
Sulfatides or 3-O-sulfogalactosylceramide are negatively charged sulfated glycosphingolipids abundant in the brain and kidneys and play crucial roles in nerve impulse conduction and urinary pH regulation. Sulfatides are present in the liver, specifically in the biliary tract. Sulfatides are self-lipid antigens presented by cholangiocytes to activate cluster of differentiation 1d (CD1d)-restricted type II natural killer T (NKT) cells. These cells are involved in alcohol-related liver disease (ArLD) and ischemic liver injury and exert anti-inflammatory effects by regulating the activity of pro-inflammatory type I NKT cells. Loss of sulfatides has been implicated in the chronic inflammatory disorder of the liver known as primary sclerosing cholangitis (PSC); bile ducts deficient in sulfatides increase their permeability, resulting in the spread of bile into the liver parenchyma. Previous studies have shown elevated levels of sulfatides in hepatocellular carcinoma (HCC), where sulfatides could act as adhesive molecules that contribute to cancer metastasis. We have recently demonstrated how loss of function of GAL3ST1, a limiting enzyme involved in sulfatide synthesis, reduces tumorigenic capacity in cholangiocarcinoma (CCA) cells. The biological function of sulfatides in the liver is still unclear; however, this review aims to summarize the existing findings on the topic.
Collapse
Affiliation(s)
- Lin Chen
- Department of Surgery, School of Nutrition and Translational Research in Metabolism, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Montserrat Elizalde
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Gloria Alvarez-Sola
- Department of Surgery, School of Nutrition and Translational Research in Metabolism, Maastricht University, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
5
|
Hortion J, Perthame E, Lafoux B, Soyer L, Reynard S, Journeaux A, Germain C, Lopez-Maestre H, Pietrosemoli N, Baillet N, Croze S, Rey C, Legras-Lachuer C, Baize S. Fatal Lassa fever in cynomolgus monkeys is associated with systemic viral dissemination and inflammation. PLoS Pathog 2024; 20:e1012768. [PMID: 39652618 DOI: 10.1371/journal.ppat.1012768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/19/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Abstract
The pathogenesis of Lassa fever has not yet been fully deciphered, particularly as concerns the mechanisms determining whether acute infection is controlled or leads to catastrophic illness and death. Using a cynomolgus monkey model of Lassa virus (LASV) infection reproducing the different outcomes of the disease, we performed histological and transcriptomic studies to investigate the dynamics of LASV infection and the immune mechanisms associated with survival or death. Lymphoid organs are an early major reservoir for replicating virus during Lassa fever, with LASV entering through the cortical sinus of draining lymph nodes regardless of disease outcome. However, subsequent viral tropism varies considerably with disease severity, with viral dissemination limited almost entirely to lymphoid organs and immune cells during nonfatal Lassa fever. By contrast, the systemic dissemination of LASV to all organs and diverse cell types, leading to infiltrations with macrophages and neutrophils and an excessive inflammatory response, is associated with a fatal outcome. These results provide new insight into early viral dynamics and the host response to LASV infection according to disease outcome.
Collapse
Affiliation(s)
- Jimmy Hortion
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, Lyon France
| | - Emeline Perthame
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Blaise Lafoux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, Lyon France
| | - Laura Soyer
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, Lyon France
| | - Stéphanie Reynard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, Lyon France
| | - Alexandra Journeaux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, Lyon France
| | - Clara Germain
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, Lyon France
| | - Hélène Lopez-Maestre
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Natalia Pietrosemoli
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Nicolas Baillet
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, Lyon France
| | | | | | | | - Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Université Paris Cité, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS UMR5308, Lyon France
| |
Collapse
|
6
|
Sakaguchi T, Nagahama Y, Hamada N, Singh SK, Mikami H, Maeda K, Akira S. Novel Choline-Deficient and 0.1%-Methionine-Added High-Fat Diet Induces Burned-Out Metabolic-Dysfunction-Associated Steatohepatitis with Inflammation by Rapid Immune Cell Infiltration on Male Mice. Nutrients 2024; 16:4151. [PMID: 39683544 DOI: 10.3390/nu16234151] [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: 11/01/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Metabolic-dysfunction-associated steatotic liver disease (MASLD) is a progressive liver disorder that possesses metabolic dysfunction and shows steatohepatitis. Although the number of patients is globally increasing and many clinical studies have developed medicine for MASLD, most of the studies have failed due to low efficacy. One reason for this failure is the lack of appropriate animal disease models that reflect human MASLD to evaluate the potency of candidate drugs. Methods: We developed a novel choline-deficient and 0.11%-methionine-added high-fat diet (CDAHFD)-based (MASH) diet that can induce murine metabolic-dysfunction-associated steatohepatitis (MASH) without severe body weight loss. We performed kinetic analyses post-feeding and proposed an appropriate timing of MASH pathogenesis by quantitatively analyzing steatosis, inflammation, and fibrosis. Results: This MASH diet induced liver fibrosis earlier than the conventional CDAHFD model. In brief, lipid accumulation, inflammation, and fibrosis started after 1 week from feeding. Lipid accumulation increased until 8 weeks and declined thereafter; on the other hand, liver fibrosis showed continuous progression. Additionally, immune cells, especially myeloid cells, specifically accumulated and induced inflammation in the initiation stage of MASH. Conclusions: The novel MASH diet promotes the dynamics of lipid deposition and fibrosis in the liver, similar to human MASH pathophysiology. Furthermore, immune-cell-derived inflammation possibly contributes to the initiation of MASH pathogenesis. We propose this model can be the new pre-clinical MASH model to discover the drugs against human MASH by evaluating the interaction between parenchymal and non-parenchymal cells.
Collapse
Affiliation(s)
- Takatoshi Sakaguchi
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | - Yasuharu Nagahama
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
- Host Defense Laboratory, Immunology Unit, Osaka Research Center for Drug Discovery, Otsuka Pharmaceutical Co., Ltd., Minoh 562-0029, Japan
| | - Nanako Hamada
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | - Shailendra Kumar Singh
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
| | | | - Kazuhiko Maeda
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
- Department of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Suita 565-0871, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, Immunology Frontier Research Center, Osaka University, Suita 565-0871, Japan
- Department of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Suita 565-0871, Japan
- Center for Advanced Modalities and Drug Delivery System (CAMaD), Osaka University, Suita 565-0871, Japan
| |
Collapse
|
7
|
Feng R, Chen L, Yang M. Aluminum-induced oxidative stress promotes changes in the structure of the gut microbiota and liver deficiency. Heliyon 2024; 10:e36165. [PMID: 39224282 PMCID: PMC11367489 DOI: 10.1016/j.heliyon.2024.e36165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
As a low-toxicity metal, aluminum has garnered increasing attention in relation to its impact on the human body; however, the specific mechanism of action remains unclear. To bridge this knowledge gap and facilitate practical applications, this study took 8-week-old ICR mice as the research object to study the effects of dietary addition of aluminum potassium sulfate on intestinal flora structure and liver. As the concentration of aluminum increased, it inhibited mice weight growth rate and significantly altered the composition of white blood cells in their bloodstream. Histological examination revealed liver inflammation through HE staining sections. The oxidative stress markers MDA increased, GSH-PX and CAT decreased significantly. And liver function index MAO increased, TC and ALP decreased first and then increased. Moreover, there was a significant increase in pro-inflammatory factor TNF-α content. Further 16S rRNA sequencing analysis demonstrated substantial changes in both composition and structure of mouse intestinal microbiota induced by aluminum exposure; microbial phenotype prediction indicated that aluminum-induced oxidative stress promoted an increase in abundance of oxidation-resistant microbial types. Alterations in gut flora structure also influenced the liver via the gut-liver axis. These findings lay a foundation for further research on the regulation and interaction of aluminum on intestinal flora.
Collapse
Affiliation(s)
- Rong Feng
- Chongqing City Vocational College, Yongchuan, Chongqing Municipality, China
| | - Liang Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu province, China
| | - Ming Yang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu province, China
| |
Collapse
|
8
|
Abdelnabi MN, Hassan GS, Shoukry NH. Role of the type 3 cytokines IL-17 and IL-22 in modulating metabolic dysfunction-associated steatotic liver disease. Front Immunol 2024; 15:1437046. [PMID: 39156888 PMCID: PMC11327067 DOI: 10.3389/fimmu.2024.1437046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 08/20/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) comprises a spectrum of liver diseases that span simple steatosis, metabolic dysfunction-associated steatohepatitis (MASH) and fibrosis and may progress to cirrhosis and cancer. The pathogenesis of MASLD is multifactorial and is driven by environmental, genetic, metabolic and immune factors. This review will focus on the role of the type 3 cytokines IL-17 and IL-22 in MASLD pathogenesis and progression. IL-17 and IL-22 are produced by similar adaptive and innate immune cells such as Th17 and innate lymphoid cells, respectively. IL-17-related signaling is upregulated during MASLD resulting in increased chemokines and proinflammatory cytokines in the liver microenvironment, enhanced recruitment of myeloid cells and T cells leading to exacerbation of inflammation and liver disease progression. IL-17 may also act directly by activating hepatic stellate cells resulting in increased fibrosis. In contrast, IL-22 is a pleiotropic cytokine with a dominantly protective signature in MASLD and is currently being tested as a therapeutic strategy. IL-22 also exhibits beneficial metabolic effects and abrogates MASH-related inflammation and fibrosis development via inducing the production of anti-oxidants and anti-apoptotic factors. A sex-dependent effect has been attributed to both cytokines, most importantly to IL-22 in MASLD or related conditions. Altogether, IL-17 and IL-22 are key effectors in MASLD pathogenesis and progression. We will review the role of these two cytokines and cells that produce them in the development of MASLD, their interaction with host factors driving MASLD including sexual dimorphism, and their potential therapeutic benefits.
Collapse
Affiliation(s)
- Mohamed N. Abdelnabi
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
9
|
Sheikhrobat SB, Mahmoudvand S, Kazemipour-Khabbazi S, Ramezannia Z, Baghi HB, Shokri S. Understanding lactate in the development of Hepatitis B virus-related hepatocellular carcinoma. Infect Agent Cancer 2024; 19:31. [PMID: 39010155 PMCID: PMC11247867 DOI: 10.1186/s13027-024-00593-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024] Open
Abstract
Hepatitis B Virus (HBV) is a hepatotropic virus that can establish a persistent and chronic infection in humans. Chronic hepatitis B (CHB) infection is associated with an increased risk of hepatic decompensation, cirrhosis, and hepatocellular carcinoma (HCC). Lactate level, as the end product of glycolysis, plays a substantial role in metabolism beyond energy production. Emerging studies indicate that lactate is linked to patient mortality rates, and HBV increases overall glucose consumption and lactate production in hepatocytes. Excessive lactate plays a role in regulating the tumor microenvironment (TME), immune cell function, autophagy, and epigenetic reprogramming. The purpose of this review is to gather and summarize the existing knowledge of the lactate's functions in the dysregulation of the immune system, which can play a crucial role in the development of HBV-related HCC. Therefore, it is reasonable to hypothesize that lactate with intriguing functions can be considered an immunomodulatory metabolite in immunotherapy.
Collapse
Affiliation(s)
- Sheida Behzadi Sheikhrobat
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahab Mahmoudvand
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Virology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Salva Kazemipour-Khabbazi
- Department of English Language and Persian Literature, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zahra Ramezannia
- Department of Virology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hossein Bannazadeh Baghi
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Somayeh Shokri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Virology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
10
|
Maretti-Mira AC, Salomon MP, Chopra S, Yuan L, Golden-Mason L. Circulating Neutrophil Profiles Undergo a Dynamic Shift during Metabolic Dysfunction-Associated Steatohepatitis (MASH) Progression. Biomedicines 2024; 12:1105. [PMID: 38791066 PMCID: PMC11117983 DOI: 10.3390/biomedicines12051105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Neutrophils play a crucial role in host defense against infection. Aberrant neutrophil activation may induce tissue damage via sterile inflammation. Neutrophil accumulation has been identified as a feature of the inflammatory response observed in metabolic dysfunction-associated steatohepatitis (MASH) and has been associated with liver fibrosis and cirrhosis. Here, we performed the transcriptomic analysis of circulating neutrophils from mild and advanced MASH patients to identify the potential mechanism behind neutrophil contribution to MASH progression. Our findings demonstrated that circulating neutrophils from mild and advanced MASH display an increased activated transcriptional program, with the expression of pro-inflammatory factors and an amplified lifespan compared to cells from non-diseased controls. Our results also suggest that MASH progression is associated with a dynamic shift in the profile of circulating neutrophils. In the early stages of MASH, mature neutrophils predominate in the bloodstream. As hepatic inflammation and fibrosis progress, the premature release of immature neutrophils into the circulation occurs. These immature neutrophils exhibit a pro-inflammatory profile that may exacerbate inflammation and promote fibrosis in MASH.
Collapse
Affiliation(s)
- Ana C. Maretti-Mira
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (M.P.S.); (L.Y.); (L.G.-M.)
| | - Matthew P. Salomon
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (M.P.S.); (L.Y.); (L.G.-M.)
| | - Shefali Chopra
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Liyun Yuan
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (M.P.S.); (L.Y.); (L.G.-M.)
| | - Lucy Golden-Mason
- USC Research Center for Liver Diseases, Division of Gastrointestinal and Liver Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; (M.P.S.); (L.Y.); (L.G.-M.)
| |
Collapse
|
11
|
Tharmalingam J, Gangadaran P, Rajendran RL, Ahn BC. Impact of Alcohol on Inflammation, Immunity, Infections, and Extracellular Vesicles in Pathogenesis. Cureus 2024; 16:e56923. [PMID: 38665743 PMCID: PMC11043057 DOI: 10.7759/cureus.56923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Alcohol consumption is a widespread social activity with a complex and multifaceted impact on human health. Although moderate alcohol consumption has been associated with certain potential health benefits, excessive or chronic alcohol use can disrupt the body's immune balance, promote inflammation, and increase susceptibility to infections. The deleterious effects associated with alcohol toxicity include the loss of cell integrity. When cells lose their integrity, they also lose the capacity to communicate with other systems. One of the systems disturbed by alcohol toxicity is extracellular vesicle (EV)-mediated communication. EVs are critical mediators of cell-to-cell communication. They play a significant role in alcohol-induced pathogenesis, facilitating communication and molecular exchange between cells, thereby potentially contributing to alcohol-related health issues. Investigating their involvement in this context is fundamental to resolving the intricate mechanisms behind the health consequences of alcohol use and may pave the way for innovative approaches for mitigating the adverse effects of alcohol on immune health. Understanding the role of EVs in the context of alcohol-induced pathogenesis is essential for comprehending the mechanisms behind alcohol-related health issues.
Collapse
Affiliation(s)
| | - Prakash Gangadaran
- Department of Nuclear Medicine, Kyungpook National University, Daegu, KOR
- Department of Biomedical Science, BK (Brain Korea) 21 FOUR (Fostering Outstanding Universities for Research) Program, Kyungpook National University, Daegu, KOR
| | | | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University, Daegu, KOR
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, KOR
- Department of Biomedical Science, BK (Brain Korea) 21 FOUR (Fostering Outstanding Universities for Research) Program, Kyungpook National University, Daegu, KOR
| |
Collapse
|
12
|
Li G, Park HJ, Suh JH, Choi HS. 7-Ketocholesterol plays a key role in cholesterol-induced hepatitis via macrophage and neutrophil infiltration. J Nutr Biochem 2024; 125:109552. [PMID: 38134972 DOI: 10.1016/j.jnutbio.2023.109552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
This study sought to explore the role of 7-ketocholesterol (7-KC) in liver damage caused by high cholesterol intake and its potential pathological mechanism in mice. Our in vivo findings indicated that mice fed a high-cholesterol diet had elevated serum levels of 7-KC, accompanied by liver injury and inflammation, similar to human nonalcoholic steatohepatitis. Furthermore, the high-cholesterol diet induced neutrophil infiltration, which played a critical role in liver damage through myeloperoxidase (MPO) activity. Upon stimulation with 7-KC, macrophages exhibited increased expression of C-X-C motif chemokine ligand 1 (CXCL1) and CXCL2, as well as ATP-binding cassette transporter A1 (ABCA1) and ABCG1. Hepatocytes, on the other hand, exhibited increased expression of CXCL2 and ABCG1. The infiltration of neutrophils in the liver was primarily caused by CXCL1 and CXCL2, resulting in hepatocyte cell death due to elevated MPO activity. Our data also revealed that the activation of macrophages by 7-KC via ABCA1 or ABCG1 was not associated with lipid accumulation. Collectively, these findings suggest that high cholesterol-induced hepatitis in mice involves, at least partially, the recruitment of neutrophils to the liver by 7-KC-activated macrophages. This is mediated by increased expression of CXCL1 and CXCL2 through ABCA1 or ABCG1, which act as 7-KC efflux transporters. Additionally, hepatocytes contribute to this process by increased expression of CXCL2 through ABCG1. Therefore, our findings suggest that 7-KC may play a role in high cholesterol-induced hepatitis in mice by activating macrophages and hepatocytes, ultimately leading to neutrophil infiltration.
Collapse
Affiliation(s)
- Guoen Li
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Hyun-Jung Park
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea
| | - Jae-Hee Suh
- Department of Pathology, Ulsan University Hospital, Ulsan, Korea
| | - Hye-Seon Choi
- Department of Biological Sciences, University of Ulsan, Ulsan, Korea.
| |
Collapse
|
13
|
Zhang JB, Zhang QR, Jin Q, Yang J, Lin SZ, Fan JG. Sestrin2 maintains hepatic immune homeostasis and redox balance partially via inhibiting RIPK3-mediated necroptosis in metabolic dysfunction-associated steatohepatitis. Mol Metab 2024; 80:101865. [PMID: 38163459 PMCID: PMC10825057 DOI: 10.1016/j.molmet.2023.101865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND & AIMS Necroptosis, a novel type of programmed cell death, is intricately associated with inflammatory response. Currently, most studies focus on the activation of necroptosis, while the mechanisms underlying the negative regulation of necroptosis remain poorly understood. METHODS The effects of sestrin2 (SESN2) overexpression or knockdown on the regulation of necroptosis were assessed in the TNFα/Smac-mimetic/Z-VAD-FMK (T/S/Z)-induced necroptosis model and palmitic acid (PA)-induced lipotoxicity model. Western-blot, co-Immunoprecipitation, Glutathione S-transferase pull-down, and confocal assays were employed to explore the regulatory mechanisms including protein-protein interactions and post-translational modification. Furthermore, we used GSK'872, a specific inhibitor of receptor-interacting serine/threonine-protein kinase (RIPK) 3, to evaluate the relationship between SESN2-related alterations and RIPK3-mediated necroptosis in T/S/Z-induced necroptosis model, PA-induced lipotoxicity model, and high-fat high-cholesterol diet (HFHCD)-induced non-alcoholic steatohepatitis model. RESULTS Our findings revealed that SESN2 was upregulated under conditions that induce necroptosis and functioned as a negative regulator of necroptosis. High levels of SESN2 could equipped hepatocytes with the ability to defend against necroptotic inflammation and oxidative stress. Mechanistically, SESN2 interacted with RIPK3 and tuned down necroptosis by inhibiting the phosphorylation of RIPK3, promoting the ubiquitination of RIPK3, and preventing the formation of the RIPK1/RIPK3 necrosome. The depletion of SESN2 resulted in excessive necroptosis, accompanied by increased fat accumulation, inflammation, and oxidative stress in the experimental steatohepatitis model. Blocking necroptosis by GSK'872 reduced the liberation of pro-inflammatory cytokines and reactive oxygen species generation, but not hepatocyte fat deposition, in both PA-treated SESN2 knockout cells and HFHCD-fed SESN2 knockout mice, suggesting that the activation of RIPK3-mediated necroptosis may partially account for the hyperinflammation and excessive oxidative stress induced by SESN2 deficiency. CONCLUSION Our results suggested that SESN2 inhibited RIPK3-mediated necroptosis; this regulation is an important for the immune homeostasis and the redox balance in the liver.
Collapse
Affiliation(s)
- Jian-Bin Zhang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qian-Ren Zhang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Qian Jin
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jing Yang
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Shuang-Zhe Lin
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Department of Gastroenterology, Xin Hua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
14
|
Zhang Y, Zhang X, Zhang M, Zhang F, Chen T, Zha J, Shen Q, Wang D, Hou C. Hepatocytes-derived Prdx1 regulates macrophage phenotypes via TLR4 activation in acute liver injury. Int Immunopharmacol 2024; 127:111439. [PMID: 38159556 DOI: 10.1016/j.intimp.2023.111439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Acute liver injury (ALI) is a significant causative factor for multiple hepatic diseases. The excessive inflammatory response triggers proinflammatory immune cells recruitment, infiltration and differentiation, further contributing to inflammatory injuries in liver. As a proinflammatory factor, circulating Peroxiredoxin 1 (Prdx1) is elevated in ALI patients and mice. In this study, through carbon tetrachloride (CCl4) and cecal puncture and ligation (CLP)-induced liver injury mice model, we found hepatocytes-derived Prdx1 expression was increased in ALI. After AAV8-Prdx1-mediated Prdx1 knockdown, CCl4 and CLP-induced ALI was alleviated, along with the reduced proinflammatory cytokines, suppressed myeloid cells recruitment, decreased proportions of hepatic macrophages and neutrophils, restrained proinflammatory macrophage differentiation and infiltration. Mechanistically, hepatocyte-derived Prdx1 regulated macrophages through paracrine activation of the TLR4 signal. Our data support the immune and inflammatory regulatory role of Prdx1 in ALI pathological process to suggest its potential therapeutic application and clinical value.
Collapse
Affiliation(s)
- Yujing Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xinru Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Mingxun Zhang
- The First Affiliated Hospital of University of Science and Technology of China, China
| | - Fanrong Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Tong Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jingjing Zha
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Qiying Shen
- The First Affiliated Hospital of Anhui Medical University, China.
| | - Dong Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Chao Hou
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
15
|
Puengel T, Tacke F. Role of Kupffer cells and other immune cells. SINUSOIDAL CELLS IN LIVER DISEASES 2024:483-511. [DOI: 10.1016/b978-0-323-95262-0.00024-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
16
|
Bao X, Leng X, Yu T, Zhu J, Zhao Y, Tenzindrogar, Yang Z, Wu S, Sun Q. Integrated Multi-omics Analyses Identify CDCA5 as a Novel Biomarker Associated with Alternative Splicing, Tumor Microenvironment, and Cell Proliferation in Colon Cancer Via Pan-cancer Analysis. J Cancer 2024; 15:825-840. [PMID: 38213717 PMCID: PMC10777042 DOI: 10.7150/jca.91082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/28/2023] [Indexed: 01/13/2024] Open
Abstract
Background: CDCA5 has been reported as a gene involved in the cell cycle, however current research provides little details. Our goal was to figure out its functions and probable mechanisms in pan-cancer. Methods: Pan-cancer bulk sequencing data and web-based analysis tools were applied to analyze CDCA5's correlations with the gene expression, clinical prognosis, genetic alterations, promoter methylation, alternative splicing, immune checkpoints, tumor microenvironment and enrichment. Real‑time PCR, cell clone formation assay, CCK-8 assay, cell proliferation assay, migration assay, invasion assay and apoptosis assay were used to evaluate the effect of CDCA5 silencing on colon cancer cell lines. Results: CDCA5 is highly expressed in most tumors, which has been linked to a poor prognosis. Immune checkpoints analysis revealed that CDCA5 was associated with the immune gene CD276 in various tumors. Single-cell analysis showed that CDCA5 correlated with proliferating T cell infiltration in COAD. Enrichment analysis demonstrated that CDCA5 may modify cell cycle genes to influence p53 signaling. The examination of DLD1 cells revealed that CDCA5 increased the proliferation and blocked cell apoptosis. Conclusion: This study contributes to the knowledge of the role of CDCA5 in carcinogenesis, highlighting the prognostic potential and carcinogenic involvement of CDCA5 in pan-cancer.
Collapse
Affiliation(s)
- Xinyue Bao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xin Leng
- Department of Urology, Affiliated Kunshan Hospital of Jiangsu University, Suzhou215300, China
| | - Tianyu Yu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Junzheya Zhu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Yunhan Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tenzindrogar
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Zhiluo Yang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Shaobo Wu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Qi Sun
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
17
|
Szafranska K, Sørensen KK, Lalor PF, McCourt P. Sinusoidal cells and liver immunology. SINUSOIDAL CELLS IN LIVER DISEASES 2024:53-75. [DOI: 10.1016/b978-0-323-95262-0.00003-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
18
|
Schefczyk S, Luo X, Liang Y, Hasenberg M, Walkenfort B, Trippler M, Schuhenn J, Sutter K, Lu M, Wedemeyer H, Schmidt HH, Broering R. Tg1.4HBV-s-rec mice, a crossbred hepatitis B virus-transgenic model, develop mild hepatitis. Sci Rep 2023; 13:22829. [PMID: 38129531 PMCID: PMC10739827 DOI: 10.1038/s41598-023-50090-8] [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: 09/28/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Hepatitis B virus (HBV)-transgenic mice exhibit competent innate immunity and are therefore an ideal model for considering intrinsic or cell-based mechanisms in HBV pathophysiology. A highly replicative model that has been little used, let alone characterized, is the Tg1.4HBV-s-rec strain derived from cross breeding of HBV-transgenic mouse models that either accumulate (Alb/HBs, Tg[Alb1-HBV]Bri44) or lack (Tg1.4HBV-s-mut) the hepatitis B surface antigen (HBsAg). Tg1.4HBV-s-rec hepatocytes secreted HBsAg, Hepatitis B extracellular antigen (HBeAg) and produced HBV virions. Transmission electron microscopy visualised viral particles (Tg1.4HBV-s-rec), nuclear capsid formations (Tg1.4HBV-s-mut and Tg1.4HBV-s-rec) and endoplasmic reticulum malformations (Alb/HBs). Viral replication in Tg1.4HBV-s-rec and Tg1.4HBV-s-mut differed in HBsAg expression and interestingly in the distribution of HBV core antigen (HBcAg) and HBV × protein. While in Tg1.4HBV-s-mut hepatocytes, the HBcAg was located in the cytoplasm, in Tg1.4HBV-s-rec hepatocytes, the HBcAg appeared in the nuclei, suggesting a more productive replication. Finally, Tg1.4HBV-s-rec mice showed symptoms of mild hepatitis, with reduced liver function and elevated serum transaminases, which appeared to be related to natural killer T cell activation. In conclusion, the study of Alb/HBs, Tg1.4HBV-s-mut and their F1 progeny provides a powerful tool to elucidate HBV pathophysiology, especially in the early HBeAg-positive phases of chronic infection and chronic hepatitis.
Collapse
Affiliation(s)
- Stefan Schefczyk
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Xufeng Luo
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
- Institute for Lymphoma Research, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Yaojie Liang
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Mike Hasenberg
- Electron Microscopy Unit, Imaging Center Essen, Medical Faculty, Germany Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Walkenfort
- Electron Microscopy Unit, Imaging Center Essen, Medical Faculty, Germany Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Martin Trippler
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Jonas Schuhenn
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hartmut H Schmidt
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Ruth Broering
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
19
|
Yang S, Zhang L, Jin Q, Wang J, Ma D, Gao J, Huang R. Meld-sarcopenia score and skeletal muscle density predicts short-term readmission of patients with hepatic encephalopathy. Eur J Radiol 2023; 169:111178. [PMID: 37922620 DOI: 10.1016/j.ejrad.2023.111178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/10/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
PURPOSE To investigate whether the quality of skeletal muscle mass could predict short-term readmission in patients with hepatic encephalopathy (HE). METHOD Patients with HE were enrolled from 2018 to 2022. Sarcopenia and myosteatosis were defined using the L3 skeletal muscle index (SMI) and skeletal muscle density (SMD) obtained from CT imaging. MELD-Sarcopenia score was calculated. Multivariable analysis and multiple linear regression were applied to identify predictors of 30-day readmission and length of hospitalization. RESULTS 123 patients with HE were included. 55 (44.7%) and 87 (70.7%) patients were identified with sarcopenia and myosteatosis, respectively. Patients with sarcopenia exhibited a higher prevalence of myosteatosis, lower SMI and SMD (p < 0.05). Patients with myosteatosis were older, had a lower body mass index, a higher neutrophil-to-lymphocyte ratio and MELD-sarcopenia scores (p < 0.05). 10 (8.1%) patients were readmitted within 30 days. The readmitted group had a higher MELD-sarcopenia score (25.0 ± 6.6 vs. 19.5 ± 7.8, p = 0.034) and lower L3 SMD (28.3 ± 5.9 vs. 33.8 ± 6.9, p = 0.015). In the multivariable analysis, MELD-sarcopenia score (95% CI 1.388 [1.074-1.793], p = 0.012) and SMD (95% CI 0.778 [0.610-0.991], p = 0.042) were found to be significantly associated with the 30-day readmission of patients with HE. Age (p = 0.028), alcohol liver disease (p = 0.025), and hypertension (p = 0.003) were associated with the length of hospitalization for patients with HE. CONCLUSIONS The MELD-sarcopenia score and SMD were identified as predictive factors for short-term readmission in patients diagnosed as HE.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Radiology, Peking University People' s Hospital, No.11 Xizhimen South Street, Beijing 100044, China
| | - Lin Zhang
- Peking University Hepatology Institute, Peking University People' s Hospital, No.11 Xizhimen South Street, Beijing 100044, China
| | - Qian Jin
- Peking University Hepatology Institute, Peking University People' s Hospital, No.11 Xizhimen South Street, Beijing 100044, China
| | - Jian Wang
- Peking University Hepatology Institute, Peking University People' s Hospital, No.11 Xizhimen South Street, Beijing 100044, China
| | - Danli Ma
- Peking University Hepatology Institute, Peking University People' s Hospital, No.11 Xizhimen South Street, Beijing 100044, China
| | - Jie Gao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, China.
| | - Rui Huang
- Peking University Hepatology Institute, Peking University People' s Hospital, No.11 Xizhimen South Street, Beijing 100044, China.
| |
Collapse
|
20
|
Balazs I, Stadlbauer V. Circulating neutrophil anti-pathogen dysfunction in cirrhosis. JHEP Rep 2023; 5:100871. [PMID: 37822786 PMCID: PMC10562928 DOI: 10.1016/j.jhepr.2023.100871] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 07/16/2023] [Accepted: 07/22/2023] [Indexed: 10/13/2023] Open
Abstract
Neutrophils are the largest population of leucocytes and are among the first cells of the innate immune system to fight against intruding pathogens. In patients with cirrhosis, neutrophils exhibit altered functionality, including changes in phagocytic ability, bacterial killing, chemotaxis, degranulation, reactive oxygen species production and NET (neutrophil extracellular trap) formation. This results in their inability to mount an adequate antibacterial response and protect the individual from infection. Prognosis and survival in patients with cirrhosis are greatly influenced by the development of infectious complications. Multidrug-resistant bacterial infections in patients with cirrhosis are currently a growing problem worldwide; therefore, alternative methods for the prevention and treatment of bacterial infections in cirrhosis are urgently needed. The prevention and treatment of neutrophil dysfunction could be a potential way to protect patients from bacterial infections. However, the reasons for changes in neutrophil function in cirrhosis are still not completely understood, which limits the development of efficient therapeutic strategies. Both cellular and serum factors have been proposed to contribute to the functional impairment of neutrophils. Herein, we review the current knowledge on features and proposed causes of neutrophil dysfunction in cirrhosis, with a focus on current knowledge gaps and limitations, as well as opportunities for future investigations in this field.
Collapse
Affiliation(s)
- Irina Balazs
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Vanessa Stadlbauer
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| |
Collapse
|
21
|
Ji Y, Liang Y, Chu PH, Ge M, Yeung SC, Ip MSM, Mak JCW. The effects of intermittent hypoxia on hepatic expression of fatty acid translocase CD36 in lean and diet-induced obese mice. Biomed J 2023; 46:100566. [PMID: 36244649 PMCID: PMC10498409 DOI: 10.1016/j.bj.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 09/01/2022] [Accepted: 10/11/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Both obstructive sleep apnea (OSA) and non-alcoholic fatty liver disease (NAFLD) are prevalent within obese individuals. We aimed to investigate the effects of intermittent hypoxia (IH), a clinical feature of OSA, on hepatic expression of fatty acid translocase (CD36) in relation to liver injury in lean and diet-induced obese mice. METHODS Four-week-old male C57BL/6J mice were randomized to standard diet (SD) or high fat (HF) diet groups. At 13-week-old, all mice were exposed to either air or IH (IH30; thirty hypoxic episodes per hour) for four weeks. We assessed liver injury through lipid profile, oxidative and inflammatory stress, histological scoring and hepatic CD36 expression. RESULTS In lean mice, IH elevated serum and hepatic triglyceride and free fatty acid (FFA) levels, in line with upregulation of hepatic CD36 expression and myeloperoxidase (MPO)-positive cells in support of inflammatory infiltrates along with increase in serum malondialdehyde (MDA), C-X-C motif chemokine ligand 1(CXCL-1) and monocyte chemoattractant protein-1 (MCP-1). In diet-induced obese mice, an increase in hepatic alanine transaminase (ALT) activity, serum and hepatic levels of lipid parameters and inflammatory markers, serum MDA level, hepatic expressions of CD36 and α-smooth muscle actin (α-SMA), and MPO-positive cells was observed. IH potentiated hepatic ALT activity, serum CXCL-1 and hepatic interleukin-6 (IL-6), in line with inflammatory infiltrates, but paradoxically, reduced hepatic FFA level and hepatic CD36 expression, compared to obese mice without IH exposure. However, IH further augmented diet-induced liver steatosis and fibrosis as shown by histological scores. CONCLUSION This study contributes to support that IH featuring OSA may lead to liver injury via differential regulation of hepatic CD36 expression in lean and diet-induced obese mice.
Collapse
Affiliation(s)
- Yang Ji
- Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yingmin Liang
- Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Pak Hin Chu
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Mengqin Ge
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sze Chun Yeung
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Mary Sau Man Ip
- Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Judith Choi Wo Mak
- Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
22
|
Abstract
Chronic liver diseases such as nonalcoholic fatty liver disease (NAFLD) or viral hepatitis are characterized by persistent inflammation and subsequent liver fibrosis. Liver fibrosis critically determines long-term morbidity (for example, cirrhosis or liver cancer) and mortality in NAFLD and nonalcoholic steatohepatitis (NASH). Inflammation represents the concerted response of various hepatic cell types to hepatocellular death and inflammatory signals, which are related to intrahepatic injury pathways or extrahepatic mediators from the gut-liver axis and the circulation. Single-cell technologies have revealed the heterogeneity of immune cell activation concerning disease states and the spatial organization within the liver, including resident and recruited macrophages, neutrophils as mediators of tissue repair, auto-aggressive features of T cells as well as various innate lymphoid cell and unconventional T cell populations. Inflammatory responses drive the activation of hepatic stellate cells (HSCs), and HSC subsets, in turn, modulate immune mechanisms via chemokines and cytokines or transdifferentiate into matrix-producing myofibroblasts. Current advances in understanding the pathogenesis of inflammation and fibrosis in the liver, mainly focused on NAFLD or NASH owing to the high unmet medical need, have led to the identification of several therapeutic targets. In this Review, we summarize the inflammatory mediators and cells in the diseased liver, fibrogenic pathways and their therapeutic implications.
Collapse
Affiliation(s)
- Linda Hammerich
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
23
|
Ding R, Liu D, Feng Y, Liu H, Ji H, He L, Liu S. Unexcited Light Source Imaging for Biomedical Applications. Chemistry 2023; 29:e202301689. [PMID: 37401914 DOI: 10.1002/chem.202301689] [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: 05/27/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/05/2023]
Abstract
Optical imaging has a wide range of applications in the biomedical field, allowing the visualization of physiological processes and helping in the diagnosis and treatment of diseases. Unexcited light source imaging technologies, such as chemiluminescence imaging, bioluminescence imaging and afterglow imaging have attracted great attention in recent years because of the absence of excitation light interference in their application and the advantages of high sensitivity and high signal-to-noise ratio. In this review, the latest advances in unexcited light source imaging technology for biomedical applications are highlighted. The design strategies of unexcited light source luminescent probes in improving luminescence brightness, penetration depth, quantum yield and targeting, and their applications in inflammation imaging, tumor imaging, liver and kidney injury imaging and bacterial infection imaging are introduced in detail. The research progress and future prospects of unexcited light source imaging for medical applications are further discussed.
Collapse
Affiliation(s)
- Ruihao Ding
- School of Material Science and Chemical Engineering, Harbin University of Science and Technology, 150040, Harbin, China
| | - Danqing Liu
- School of Material Science and Chemical Engineering, Harbin University of Science and Technology, 150040, Harbin, China
| | - Yu Feng
- School of Material Science and Chemical Engineering, Harbin University of Science and Technology, 150040, Harbin, China
| | - Haoxin Liu
- Augustana Faculty, University of Alberta, T4V2R3, Camrose, Canada
| | - Hongrui Ji
- School of Material Science and Chemical Engineering, Harbin University of Science and Technology, 150040, Harbin, China
| | - Liangcan He
- Key Laboratory of Micro-systems and, Micro-structures Manufacturing of Ministry of Education, Harbin Institute of Technology, 150001, Harbin, China
| | - Shaoqin Liu
- Key Laboratory of Micro-systems and, Micro-structures Manufacturing of Ministry of Education, Harbin Institute of Technology, 150001, Harbin, China
| |
Collapse
|
24
|
Zhang L, Zhang W, Wang J, Jin Q, Ma D, Huang R. Neutrophil-to-lymphocyte ratio predicts 30-, 90-, and 180-day readmissions of patients with hepatic encephalopathy. Front Med (Lausanne) 2023; 10:1185182. [PMID: 37457569 PMCID: PMC10348710 DOI: 10.3389/fmed.2023.1185182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Hepatic encephalopathy (HE) is a significant complication of cirrhosis, known to be associated with hospital readmission. However, few new serological indicators associated with readmission in HE patients have been identified and reported. The objective of our study was to identify simple and effective predictors reated to readmission in HE patients. Materials and methods We conducted a retrospective study at a single center on adult patients admitted with HE from January 2018 to December 2022. The primary endpoint was the first liver-related readmission within 30, 90, and 180 days, and we collected electronic medical records from our hospital for sociodemographic, clinical, and hospitalization characteristics. We utilized logistic regression analysis and multiple linear regression analysis to determine the predictors that were associated with the readmission rate and the length of the first hospitalization. Results A total of 424 patients were included in the study, among whom 24 (5.7%), 63 (14.8%), and 92 (21.7%) were readmitted within 30, 90, and 180 days, respectively. Logistic regression analysis showed that insurance status, alcoholic liver disease (ALD), ascites, the model for end-stage liver disease (MELD) score, and neutrophil-to-lymphocyte ratio (NLR) were significantly associated with 30-, 90-, and 180-day readmissions. Age and hepatocellular carcinoma (HCC) were predictors of 90- and 180-day readmissions. ALD was identified as a unique predictor of readmission in men, while hypertension was a predictor of 180-day readmission in women. Variceal bleeding, chronic kidney disease, and MELD score were associated with the length of the first hospitalization. Conclusions NLR at discharge was identified as a significant predictor of 30-, 90- and 180-day readmissions in patients with HE. Our findings suggest that incorporating NLR into routine clinical assessments could improve the evaluation of the prognosis of liver cirrhosis.
Collapse
|
25
|
Otumala AE, Hellen DJ, Luna CA, Delgado P, Dissanayaka A, Ugwumadu C, Oshinowo O, Islam MM, Shen L, Karpen SJ, Myers DR. Opportunities and considerations for studying liver disease with microphysiological systems on a chip. LAB ON A CHIP 2023; 23:2877-2898. [PMID: 37282629 DOI: 10.1039/d2lc00940d] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Advances in microsystem engineering have enabled the development of highly controlled models of the liver that better recapitulate the unique in vivo biological conditions. In just a few short years, substantial progress has been made in creating complex mono- and multi-cellular models that mimic key metabolic, structural, and oxygen gradients crucial for liver function. Here we review: 1) the state-of-the-art in liver-centric microphysiological systems and 2) the array of liver diseases and pressing biological and therapeutic challenges which could be investigated with these systems. The engineering community has unique opportunities to innovate with new liver-on-a-chip devices and partner with biomedical researchers to usher in a new era of understanding of the molecular and cellular contributors to liver diseases and identify and test rational therapeutic modalities.
Collapse
Affiliation(s)
- Adiya E Otumala
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dominick J Hellen
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - C Alessandra Luna
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Priscilla Delgado
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anjana Dissanayaka
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Chidozie Ugwumadu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Oluwamayokun Oshinowo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Md Mydul Islam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Luyao Shen
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Saul J Karpen
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - David R Myers
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
26
|
Adugna A. Antigen Recognition and Immune Response to Acute and Chronic Hepatitis B Virus Infection. J Inflamm Res 2023; 16:2159-2166. [PMID: 37223107 PMCID: PMC10202203 DOI: 10.2147/jir.s411492] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/02/2023] [Indexed: 05/25/2023] Open
Abstract
The antigen recognition and immune response to acute and chronic hepatitis B virus (HBV) infections are the result of both the innate and adaptive immune response. The innate immune response comprises Dendritic Cells (DCs), which served as professional antigen-presenting cells and a bridge between innate and adaptive immunity, Kupffer cells and inflammatory monocytes for the continuous inflammation of hepatocyte, neutrophils for hepatic tissue damage due to acute inflammation, type I interferons (IFN), which induce an antiviral state on infected cells, directs natural killer (NK) cells to kill virally infected cells, reduces the population of infected cells, and promotes the effective maturation and site recruitment of adaptive immunity through the production of pro-inflammatory cytokines and chemokines. Through stimulating B cells, T-helper, and cytotoxic T cells, the adaptive immune system also protects against hepatitis B infection. During HBV infection, a network of cell types that can either play protective or harmful functions creates the anti-viral adaptive immune response. These many elements, such as Cluster of differentiation four (CD4) T cells (traditionally known as helper T cells), are potent cytokine producers and necessary for the effective maturation of effector cytotoxic cluster of differentiation eight (CD8) T cells and B cell antibody production. By cytolytic and non-cytolytic processes, CD8 T cells are able to eliminate HBV-infected hepatocytes and directly detect virus-infected cells, and circulating CD4+ CD25+ regulatory T cells for the modulation of immune system. In order to avoid reinfection, B cells can produce antibodies that destroy free viral particles. Moreover, by presenting HBV antigens to helper T cells, B cells may also influence how well these cells operate.
Collapse
Affiliation(s)
- Adane Adugna
- Medical Microbiology, Medical Laboratory Sciences, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
27
|
Kountouras J, Kazakos E, Kyrailidi F, Polyzos SA, Zavos C, Arapoglou S, Boziki M, Mouratidou MC, Tzitiridou-Chatzopoulou M, Chatzopoulos D, Doulberis M, Papaefthymiou A, Vardaka E. Innate immunity and nonalcoholic fatty liver disease. Ann Gastroenterol 2023; 36:244-256. [PMID: 37144011 PMCID: PMC10152810 DOI: 10.20524/aog.2023.0793] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/14/2023] [Indexed: 05/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently renamed as metabolic (dysfunction)-associated fatty liver disease (MAFLD), is a complex, multifactorial disease that progresses via nonalcoholic steatohepatitis (NASH) towards severe liver complications. MAFLD/NAFLD affects up to a third of the global population. It is connected with metabolic syndrome parameters and has been increasing in parallel with the rates of metabolic syndrome parameters worldwide. This disease entity exhibits a strong immune-inflammatory dimension. In MAFLD/NAFLD/NASH, a vast network of innate immune cells is mobilized that can provoke liver damage, leading to advanced fibrosis, cirrhosis and its complications, including hepatocellular carcinoma. However, our understanding of the inflammatory signals that drive the onset and progression of MAFLD/NAFLD/NASH is fragmented. Thus, further investigation is required to better understand the role of specific innate immune cell subsets in the disease, and to aid the design of innovative therapeutic agents to target MAFLD/NAFLD/NASH. In this review, we discuss current concepts regarding the role of innate immune system involvement in MAFLD/NAFLD/NASH onset and progression, along with presenting potential stress signals affecting immune tolerance that may trigger aberrant immune responses. A comprehensive understanding of the innate immune mechanisms involved in MAFLD/NAFLD/NASH pathophysiology will help the discovery of early interventions to prevent the disease, and lead to potential innovative therapeutic strategies that may limit its worldwide burden.
Collapse
Affiliation(s)
- Jannis Kountouras
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
| | - Evangelos Kazakos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
- School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, Macedonia, Greece (Evangelos Kazakos)
| | - Foteini Kyrailidi
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
| | - Stergios A. Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Macedonia, Greece (Stergios A. Polyzos, Michael Doulberis, Apostolis Papaefthymiou)
| | - Christos Zavos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
| | - Stergios Arapoglou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
- Fifth Surgical Department, Medical School, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Stergios Arapoglou)
| | - Marina Boziki
- 2 Neurology Department, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA Hospital, Macedonia, Greece (Marina Boziki)
| | - Maria C. Mouratidou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
| | - Maria Tzitiridou-Chatzopoulou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
- School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, Macedonia, Greece (Maria Tzitiridou-Chatzopoulou)
| | - Dimitrios Chatzopoulos
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Macedonia, Greece (Stergios A. Polyzos, Michael Doulberis, Apostolis Papaefthymiou)
- Department of Gastroenterology and Hepatology, University of Zurich, Zurich, Switzerland (Michael Doulberis)
| | - Apostolis Papaefthymiou
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Macedonia, Greece (Stergios A. Polyzos, Michael Doulberis, Apostolis Papaefthymiou)
- Pancreaticobiliary Medicine Unit, University College London Hospitals (UCLH), London, UK (Apostolis Papaefthymiou)
| | - Elisabeth Vardaka
- Second Medical Clinic, School of Medicine, Ippokration Hospital, Aristotle University of Thessaloniki, Macedonia, Greece (Jannis Kountouras, Evangelos Kazakos, Foteini Kyrailidi, Christos Zavos, Stergios Arapoglou, Maria C. Mouratidou, Maria Tzitiridou-Chatzopoulou, Dimitrios Chatzopoulos, Michael Doulberis, Apostolis Papaefthymiou, Elisabeth Vardaka)
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, Alexander Campus, Macedonia, Greece (Elisabeth Vardaka)
| |
Collapse
|
28
|
Ma DW, Ha J, Yoon KS, Kang I, Choi TG, Kim SS. Innate Immune System in the Pathogenesis of Non-Alcoholic Fatty Liver Disease. Nutrients 2023; 15:2068. [PMID: 37432213 DOI: 10.3390/nu15092068] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 07/12/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent condition characterized by lipid accumulation in hepatocytes with low alcohol consumption. The development of sterile inflammation, which occurs in response to a range of cellular stressors or injuries, has been identified as a major contributor to the pathogenesis of NAFLD. Recent studies of the pathogenesis of NAFLD reported the newly developed roles of damage-associated molecular patterns (DAMPs). These molecules activate pattern recognition receptors (PRRs), which are placed in the infiltrated neutrophils, dendritic cells, monocytes, or Kupffer cells. DAMPs cause the activation of PRRs, which triggers a number of immunological responses, including the generation of cytokines that promote inflammation and the localization of immune cells to the site of the damage. This review provides a comprehensive overview of the impact of DAMPs and PRRs on the development of NAFLD.
Collapse
Affiliation(s)
- Dae Won Ma
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
29
|
Li N, Liu H, Xue Y, Xu Z, Miao X, Guo Y, Li Z, Fan Z, Xu Y. Targetable Brg1-CXCL14 axis contributes to alcoholic liver injury by driving neutrophil trafficking. EMBO Mol Med 2023; 15:e16592. [PMID: 36722664 PMCID: PMC9994483 DOI: 10.15252/emmm.202216592] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 02/02/2023] Open
Abstract
Alcoholic liver disease (ALD) accounts for a large fraction of patients with cirrhosis and hepatocellular carcinoma. In the present study we investigated the involvement of Brahma-related gene 1 (Brg1) in ALD pathogenesis and implication in ALD intervention. We report that Brg1 expression was elevated in mouse models of ALD, in hepatocyte exposed to alcohol, and in human ALD specimens. Manipulation of Brg1 expression in hepatocytes influenced the development of ALD in mice. Flow cytometry showed that Brg1 deficiency specifically attenuated hepatic infiltration of Ly6G+ neutrophils in the ALD mice. RNA-seq identified C-X-C motif chemokine ligand 14 (CXCL14) as a potential target for Brg1. CXCL14 knockdown alleviated whereas CXCL14 over-expression enhanced ALD pathogenesis in mice. Importantly, pharmaceutical inhibition of Brg1 with a small-molecule compound PFI-3 or administration of an antagonist to the CXCL14 receptor ameliorated ALD pathogenesis in mice. Finally, a positive correlation between Brg1 expression, CXCL14 expression, and neutrophil infiltration was detected in ALD patients. In conclusion, our data provide proof-of-concept for targeting the Brg1-CXCL14 axis in ALD intervention.
Collapse
Affiliation(s)
- Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
| | - Hong Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
| | - Yujia Xue
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
| | - Zheng Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
| | - Xiulian Miao
- Collage of Life Sciences and Institute of Biomedical Research, Liaocheng UniversityLiaochengChina
| | - Yan Guo
- Collage of Life Sciences and Institute of Biomedical Research, Liaocheng UniversityLiaochengChina
| | - Zilong Li
- State Key Laboratory of Natural Medicines, Department of PharmacologyChina Pharmaceutical UniversityNanjingChina
| | - Zhiwen Fan
- Department of PathologyNanjing Drum Tower Hospital Affiliated to Nanjing University Medical SchoolNanjingChina
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of PathophysiologyNanjing Medical UniversityNanjingChina
- Collage of Life Sciences and Institute of Biomedical Research, Liaocheng UniversityLiaochengChina
- State Key Laboratory of Natural Medicines, Department of PharmacologyChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
30
|
Timmer KD, Floyd DJ, Scherer AK, Crossen AJ, Atallah J, Viens AL, Sykes DB, Mansour MK. Multiparametric Profiling of Neutrophil Function via a High-Throughput Flow Cytometry-Based Assay. Cells 2023; 12:743. [PMID: 36899878 PMCID: PMC10000770 DOI: 10.3390/cells12050743] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023] Open
Abstract
Neutrophils are a vital component of the innate immune system and play an essential function in the recognition and clearance of bacterial and fungal pathogens. There is great interest in understanding mechanisms of neutrophil dysfunction in the setting of disease and deciphering potential side effects of immunomodulatory drugs on neutrophil function. We developed a high throughput flow cytometry-based assay for detecting changes to four canonical neutrophil functions following biological or chemical triggers. Our assay detects neutrophil phagocytosis, reactive oxygen species (ROS) generation, ectodomain shedding, and secondary granule release in a single reaction mixture. By selecting fluorescent markers with minimal spectral overlap, we merge four detection assays into one microtiter plate-based assay. We demonstrate the response to the fungal pathogen, Candida albicans and validate the assay's dynamic range using the inflammatory cytokines G-CSF, GM-CSF, TNFα, and IFNγ. All four cytokines increased ectodomain shedding and phagocytosis to a similar degree while GM-CSF and TNFα were more active in degranulation when compared to IFNγ and G-CSF. We further demonstrated the impact of small molecule inhibitors such as kinase inhibition downstream of Dectin-1, a critical lectin receptor responsible for fungal cell wall recognition. Bruton's tyrosine kinase (Btk), Spleen tyrosine kinase (Syk), and Src kinase inhibition suppressed all four measured neutrophil functions but all functions were restored with lipopolysaccharide co-stimulation. This new assay allows for multiple comparisons of effector functions and permits identification of distinct subpopulations of neutrophils with a spectrum of activity. Our assay also offers the potential for studying the intended and off-target effects of immunomodulatory drugs on neutrophil responses.
Collapse
Affiliation(s)
- Kyle D. Timmer
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Daniel J. Floyd
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Allison K. Scherer
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 20114, USA
| | - Arianne J. Crossen
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Johnny Atallah
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 20114, USA
| | - Adam L. Viens
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David B. Sykes
- Harvard Medical School, Boston, MA 20114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA 02114, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02114, USA
| | - Michael K. Mansour
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 20114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| |
Collapse
|
31
|
Dukić M, Radonjić T, Jovanović I, Zdravković M, Todorović Z, Kraišnik N, Aranđelović B, Mandić O, Popadić V, Nikolić N, Klašnja S, Manojlović A, Divac A, Gačić J, Brajković M, Oprić S, Popović M, Branković M. Alcohol, Inflammation, and Microbiota in Alcoholic Liver Disease. Int J Mol Sci 2023; 24:ijms24043735. [PMID: 36835145 PMCID: PMC9966185 DOI: 10.3390/ijms24043735] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Alcoholic liver disease (ALD) is a consequence of excessive alcohol use. According to many studies, alcohol represents a significant socioeconomic and health risk factor in today's population. According to data from the World Health Organization, there are about 75 million people who have alcohol disorders, and it is well known that its use leads to serious health problems. ALD is a multimodality spectrum that includes alcoholic fatty liver disease (AFL) and alcoholic steatohepatitis (ASH), consequently leading to liver fibrosis and cirrhosis. In addition, the rapid progression of alcoholic liver disease can lead to alcoholic hepatitis (AH). Alcohol metabolism produces toxic metabolites that lead to tissue and organ damage through an inflammatory cascade that includes numerous cytokines, chemokines, and reactive oxygen species (ROS). In the process of inflammation, mediators are cells of the immune system, but also resident cells of the liver, such as hepatocytes, hepatic stellate cells, and Kupffer cells. These cells are activated by exogenous and endogenous antigens, which are called pathogen and damage-associated molecular patterns (PAMPs, DAMPs). Both are recognized by Toll-like receptors (TLRs), which activation triggers the inflammatory pathways. It has been proven that intestinal dysbiosis and disturbed integrity of the intestinal barrier perform a role in the promotion of inflammatory liver damage. These phenomena are also found in chronic excessive use of alcohol. The intestinal microbiota has an important role in maintaining the homeostasis of the organism, and its role in the treatment of ALD has been widely investigated. Prebiotics, probiotics, postbiotics, and symbiotics represent therapeutic interventions that can have a significant effect on the prevention and treatment of ALD.
Collapse
Affiliation(s)
- Marija Dukić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Tijana Radonjić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Igor Jovanović
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Marija Zdravković
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Zoran Todorović
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Nemanja Kraišnik
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Bojana Aranđelović
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Olga Mandić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Višeslav Popadić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Novica Nikolić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Slobodan Klašnja
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Andrea Manojlović
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Anica Divac
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Jasna Gačić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Brajković
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Svetlana Oprić
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Maja Popović
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
| | - Marija Branković
- University Hospital Medical Center Bežanijska Kosa, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
- Correspondence: or
| |
Collapse
|
32
|
IL-1β neutralization prevents diastolic dysfunction development, but lacks hepatoprotective effect in an aged mouse model of NASH. Sci Rep 2023; 13:356. [PMID: 36611037 PMCID: PMC9825403 DOI: 10.1038/s41598-022-26896-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 12/21/2022] [Indexed: 01/09/2023] Open
Abstract
Interleukin-1β (IL-1β) is a key mediator of non-alcoholic steatohepatitis (NASH), a chronic liver disease, and of systemic inflammation-driven aging. IL-1β contributes to cardio-metabolic decline, and may promote hepatic oncogenic transformation. Therefore, IL-1β is a potential therapeutic target in these pathologies. We aimed to investigate the hepatic and cardiac effects of an IL-1β targeting monoclonal antibody in an aged mouse model of NASH. 24 months old male C57Bl/6J mice were fed with control or choline deficient (CDAA) diet and were treated with isotype control or anti-IL-1β Mab for 8 weeks. Cardiac functions were assessed by conventional-and 2D speckle tracking echocardiography. Liver samples were analyzed by immunohistochemistry and qRT-PCR. Echocardiography revealed improved cardiac diastolic function in anti-IL-1β treated mice with NASH. Marked hepatic fibrosis developed in CDAA-fed group, but IL-1β inhibition affected fibrosis only at transcriptomic level. Hepatic inflammation was not affected by the IL-1β inhibitor. PCNA staining revealed intensive hepatocyte proliferation in CDAA-fed animals, which was not influenced by neutralization of IL-1β. IL-1β inhibition increased hepatic expression of Pd-1 and Ctla4, while Pd-l1 expression increased in NASH. In conclusion, IL-1β inhibition improved cardiac diastolic function, but did not ameliorate features of NASH; moreover, even promoted hepatic immune checkpoint expression, with concomitant NASH-related hepatocellular proliferation.
Collapse
|
33
|
Liu CX, Xiao SY, Gong XL, Zhu X, Wang YW, Peng Y. A Near-Infrared Fluorescent Probe for Recognition of Hypochlorite Anions Based on Dicyanoisophorone Skeleton. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28010402. [PMID: 36615593 PMCID: PMC9823594 DOI: 10.3390/molecules28010402] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 01/09/2023]
Abstract
A novel near-infrared (NIR) fluorescent probe (SWJT-9) was designed and synthesized for the detection of hypochlorite anion (ClO-) using a diaminomaleonitrile group as the recognition site. SWJT-9 had large Stokes shift (237 nm) and showed an excellent NIR fluorescence response to ClO- with the color change under the visible light. It showed a low detection limit (24.7 nM), high selectivity, and rapid detection (within 2 min) for ClO-. The new detection mechanism of SWJT-9 on ClO- was confirmed by 1H NMR, MS spectrum, and the density functional theory (DFT) calculations. In addition, the probe was successfully used to detect ClO- in HeLa cells.
Collapse
Affiliation(s)
- Chang-Xiang Liu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Shu-Yuan Xiao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiu-Lin Gong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xi Zhu
- Department of Neurology, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China
- Correspondence: (X.Z.); (Y.-W.W.)
| | - Ya-Wen Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
- Correspondence: (X.Z.); (Y.-W.W.)
| | - Yu Peng
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
34
|
Kasztelan-Szczerbinska B, Zygo B, Rycyk-Bojarzynska A, Surdacka A, Rolinski J, Cichoz-Lach H. Blood concentrations of mediators released from activated neutrophils are related to the severity of alcohol-induced liver damage. PLoS One 2023; 18:e0280068. [PMID: 36607987 PMCID: PMC9821433 DOI: 10.1371/journal.pone.0280068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/20/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Immune dysregulation and neutrophil infiltration are hallmarks of alcohol-related liver disease (ALD). Our objective was to evaluate the blood profile of neutrophil-derived mediators [neutrophil elastase (NE), myeloperoxidase (MPO), alpha1-antitrypsin (A1AT)], and their potential relevance in ALD. METHODS 62 patients with ALD /47 males, and 15 females, aged 49,2 ± 9,9/ were prospectively recruited and distributed according to their 1/ gender, 2/ severity of liver dysfunction (by Child-Turcotte-Pugh, MELD scores, and mDF) 3/ presence of complications of ALD complications, and followed for 90 days. 24 age- and sex-matched healthy volunteers served as the control group. Neutrophil-derived biomarkers were quantified using enzyme-linked immunosorbent assays (ELISAs). RESULTS Blood concentrations of MPO and NE were significantly higher in ALD patients in comparison with controls. A1AT levels were not different. There were no gender-related differences in the studied biomarker levels. Both NE and MPO correlated with routine markers of inflammation, while NE with MELD and mDF scores. Patients with a severe ALD course i.e. MELD>20 or mDF>32, presented with significantly higher NE blood concentrations. CONCLUSIONS Our results point out the critical role of neutrophils in the pathogenesis of ALD. NE and MPO correlated with the intensity of inflammation, and NE was related to the severity of liver dysfunction.
Collapse
Affiliation(s)
| | - Bartosz Zygo
- Department of Gastroenterology with Endoscopy Unit, Independent Public Academic Hospital No. 4 in Lublin, Lublin, Poland
| | - Anna Rycyk-Bojarzynska
- Department of Gastroenterology with Endoscopy Unit, Medical University of Lublin, Lublin, Poland
| | - Agata Surdacka
- Department of Clinical Immunology, Medical University of Lublin, Lublin, Poland
| | - Jacek Rolinski
- Department of Clinical Immunology, Medical University of Lublin, Lublin, Poland
| | - Halina Cichoz-Lach
- Department of Gastroenterology with Endoscopy Unit, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
35
|
Yoon JS, Lee CW. Protein phosphatases regulate the liver microenvironment in the development of hepatocellular carcinoma. Exp Mol Med 2022; 54:1799-1813. [PMID: 36380016 PMCID: PMC9722691 DOI: 10.1038/s12276-022-00883-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
The liver is a complicated heterogeneous organ composed of different cells. Parenchymal cells called hepatocytes and various nonparenchymal cells, including immune cells and stromal cells, are distributed in liver lobules with hepatic architecture. They interact with each other to compose the liver microenvironment and determine its characteristics. Although the liver microenvironment maintains liver homeostasis and function under healthy conditions, it also shows proinflammatory and profibrogenic characteristics that can induce the progression of hepatitis and hepatic fibrosis, eventually changing to a protumoral microenvironment that contributes to the development of hepatocellular carcinoma (HCC). According to recent studies, phosphatases are involved in liver diseases and HCC development by regulating protein phosphorylation in intracellular signaling pathways and changing the activities and characteristics of liver cells. Therefore, this review aims to highlight the importance of protein phosphatases in HCC development and in the regulation of the cellular components in the liver microenvironment and to show their significance as therapeutic targets.
Collapse
Affiliation(s)
- Joon-Sup Yoon
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419 Republic of Korea
| | - Chang-Woo Lee
- grid.264381.a0000 0001 2181 989XDepartment of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, 16419 Republic of Korea ,grid.264381.a0000 0001 2181 989XDepartment of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351 Republic of Korea
| |
Collapse
|
36
|
Si-Wu-Tang ameliorates bile duct ligation-induced liver fibrosis via modulating immune environment. Biomed Pharmacother 2022; 155:113834. [DOI: 10.1016/j.biopha.2022.113834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 11/18/2022] Open
|
37
|
Wu L, Jin J, Zhou T, Wu Y, Li X, Li X, Zeng J, Wang J, Ren J, Chong Y, Zheng R. A Prognostic Nomogram with High Accuracy Based on 2D-SWE in Patients with Acute-on-chronic Liver Failure. J Clin Transl Hepatol 2022; 10:803-813. [PMID: 36304504 PMCID: PMC9547255 DOI: 10.14218/jcth.2021.00278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/22/2021] [Accepted: 11/29/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Acute-on-chronic liver failure (ACLF) is associated with very high mortality. Accurate prediction of prognosis is critical in navigating optimal treatment decisions to improve patient survival. This study was aimed to develop a new nomogram integrating two-dimensional shear wave elastography (2D-SWE) values with other independent prognostic factors to improve the precision of predicting ACLF patient outcomes. METHODS A total of 449 consecutive patients with ACLF were recruited and randomly allocated to a training cohort (n=315) or a test cohort (n=134). 2D-SWE values, conventional ultrasound features, laboratory tests, and other clinical characteristics were included in univariate and multivariate analysis. Factors with prognostic value were then used to construct a novel prognostic nomogram. Receiver operating curves (ROCs) were generated to evaluate and compare the performance of the novel and published models including the Model for End-Stage Liver Disease (MELD), MELD combined with sodium (MELD-Na), and Jin's model. The model was validated in a prospective cohort (n=102). RESULTS A ACLF prognostic nomogram was developed with independent prognostic factors, including 2D-SWE, age, total bilirubin (TB), neutrophils (Neu), and the international normalized ratio (INR). The area under the ROC curve (AUC) was 0.849 for the new model in the training cohort and 0.861 in the prospective validation cohort, which were significantly greater than those for MELD (0.758), MELD-Na (0.750), and Jin's model (0.777, all p <0.05). Calibration curve analysis revealed good agreement between the predicted and observed probabilities. The new nomogram had superior overall net benefit and clinical utility. CONCLUSIONS We established and validated a 2D-SWE-based noninvasive nomogram to predict the prognosis of ACLF patients that was more accurate than other prognostic models.
Collapse
Affiliation(s)
- Lili Wu
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-Sen University, Guangdong Key Laboratory of Liver Disease Research, Guangzhou, Guangdong, China
| | - Jieyang Jin
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-Sen University, Guangdong Key Laboratory of Liver Disease Research, Guangzhou, Guangdong, China
| | - Taicheng Zhou
- Department of Gastroenterological Surgery and Hernia Center, Sixth Affiliated Hospital of Sun Yat-Sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, Guangzhou, Guangdong, China
| | - Yuankai Wu
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xinhua Li
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xiangyong Li
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jie Zeng
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-Sen University, Guangdong Key Laboratory of Liver Disease Research, Guangzhou, Guangdong, China
| | - Jinfen Wang
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-Sen University, Guangdong Key Laboratory of Liver Disease Research, Guangzhou, Guangdong, China
| | - Jie Ren
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-Sen University, Guangdong Key Laboratory of Liver Disease Research, Guangzhou, Guangdong, China
- Correspondence to: Jie Ren, Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, Guangdong 510630, China. ORCID: https://orcid.org/0000-0003-2599-9001. Tel: +86-20-85252010, Fax: +86-20-87583501, E-mail:
| | - Yutian Chong
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Rongqin Zheng
- Department of Medical Ultrasonics, Third Affiliated Hospital of Sun Yat-Sen University, Guangdong Key Laboratory of Liver Disease Research, Guangzhou, Guangdong, China
| |
Collapse
|
38
|
Antitoxic Effects of Curcumin against Obesity-Induced Multi-Organs' Biochemical and Histopathological Abnormalities in an Animal Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9707278. [PMID: 36248416 PMCID: PMC9560822 DOI: 10.1155/2022/9707278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/07/2022]
Abstract
Background Obesity is a significant public health problem that is characterized by an increase in oxidative stress and enhanced inflammatory responses associated with immune cell invasion of adipose tissues. This study assessed several biochemical abnormalities, apoptosis, oxidative stress status, and associated histological changes in the liver, duodenum, and heart brought on by high-fat diet-induced obesity in rats. It also assessed the mechanistic benefits of curcumin in reversing these inflammatory, metabolic, and histological impairments. Methods Rats were assigned into three groups each including ten rats: the control group (CD), the high-fat diet group (HFD), and the high-fat diet + curcumin (HFDC) group. Serum glucose, insulin, and triglycerides (TAGs) were observed. In addition, apoptosis (indicated by hepatic DNA fragmentation) and oxidative stress status (indicated by hepatic MPO, GSH, and SOD) were assessed. Histopathological examinations included the GIT (liver and duodenum) and heart in addition to quantitative real-time polymerase chain reaction (qRT-PCR) assays of the adipose tissue genetic expressions for inflammatory signaling pathways (TLR4, IL-6, and TNF-α). Results The overall findings showed that the HFD group exhibited significantly higher levels of glucose, TAGs, and insulin than the control group (P < 0.01). The histological abnormalities of the studied organs in the HFD group were paralleled by these biochemical abnormalities, which were strongly associated with increased apoptosis, increased oxidative stress, and increased expression of the inflammatory signaling markers. There were significant improvements in the HFDC group in terms of biochemical, inflammatory, and histological investigations. Conclusions This study's findings concluded that obesity is significantly associated with biochemical and microscopic alterations in many organs. Curcumin exerted potent antitoxic, antioxidant, tissue-protective, and antiobesity effects. Curcumin is recommended to be added to various dietary regimens to prevent or delay the organs' dysfunction among obese people.
Collapse
|
39
|
Flores Molina M, Abdelnabi MN, Mazouz S, Villafranca-Baughman D, Trinh VQH, Muhammad S, Bédard N, Osorio Laverde D, Hassan GS, Di Polo A, Shoukry NH. Distinct spatial distribution and roles of Kupffer cells and monocyte-derived macrophages in mouse acute liver injury. Front Immunol 2022; 13:994480. [PMID: 36248843 PMCID: PMC9562324 DOI: 10.3389/fimmu.2022.994480] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022] Open
Abstract
Macrophages are key regulators of inflammation and repair, but their heterogeneity and multiple roles in the liver are not fully understood. We aimed herein to map the intrahepatic macrophage populations and their function(s) during acute liver injury. We used flow cytometry, gene expression analysis, multiplex-immunofluorescence, 3D-reconstruction, and spatial image analysis to characterize the intrahepatic immune landscape in mice post-CCl4-induced acute liver injury during three distinct phases: necroinflammation, and early and late repair. We observed hepatocellular necrosis and a reduction in liver resident lymphocytes during necroinflammation accompanied by the infiltration of circulating myeloid cells and upregulation of inflammatory cytokines. These parameters returned to baseline levels during the repair phase while pro-repair chemokines were upregulated. We identified resident CLEC4F+ Kupffer cells (KCs) and infiltrating IBA1+CLEC4F- monocyte-derived macrophages (MoMFs) as the main hepatic macrophage populations during this response to injury. While occupying most of the necrotic area, KCs and MoMFs exhibited distinctive kinetics, distribution and morphology at the site of injury. The necroinflammation phase was characterized by low levels of KCs and a remarkable invasion of MoMFs suggesting their potential role in phagoctosing necrotic hepatocytes, while opposite kinetics/distribution were observed during repair. During the early repair phase, yolksac - derived KCs were restored, whereas MoMFs diminished gradually then dissipated during late repair. MoMFs interacted with hepatic stellate cells during the necroinflammatory and early repair phases, potentially modulating their activation state and influencing their fibrogenic and pro-repair functions that are critical for wound healing. Altogether, our study reveals novel and distinct spatial and temporal distribution of KCs and MoMFs and provides insights into their complementary roles during acute liver injury.
Collapse
Affiliation(s)
- Manuel Flores Molina
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Mohamed N. Abdelnabi
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Sabrina Mazouz
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Deborah Villafranca-Baughman
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de neurosciences, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Vincent Quoc-Huy Trinh
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Shafi Muhammad
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Nathalie Bédard
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - David Osorio Laverde
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Ghada S. Hassan
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Adriana Di Polo
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de neurosciences, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
- Département de médecine, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
- *Correspondence: Naglaa H. Shoukry,
| |
Collapse
|
40
|
Ma J, Guillot A, Yang Z, Mackowiak B, Hwang S, Park O, Peiffer BJ, Ahmadi AR, Melo L, Kusumanchi P, Huda N, Saxena R, He Y, Guan Y, Feng D, Sancho-Bru P, Zang M, Cameron AM, Bataller R, Tacke F, Sun Z, Liangpunsakul S, Gao B. Distinct histopathological phenotypes of severe alcoholic hepatitis suggest different mechanisms driving liver injury and failure. J Clin Invest 2022; 132:e157780. [PMID: 35838051 PMCID: PMC9282929 DOI: 10.1172/jci157780] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/27/2022] [Indexed: 12/13/2022] Open
Abstract
Intrahepatic neutrophil infiltration has been implicated in severe alcoholic hepatitis (SAH) pathogenesis; however, the mechanism underlying neutrophil-induced injury in SAH remains obscure. This translational study aims to describe the patterns of intrahepatic neutrophil infiltration and its involvement in SAH pathogenesis. Immunohistochemistry analyses of explanted livers identified two SAH phenotypes despite a similar clinical presentation, one with high intrahepatic neutrophils (Neuhi), but low levels of CD8+ T cells, and vice versa. RNA-Seq analyses demonstrated that neutrophil cytosolic factor 1 (NCF1), a key factor in controlling neutrophilic ROS production, was upregulated and correlated with hepatic inflammation and disease progression. To study specifically the mechanisms related to Neuhi in AH patients and liver injury, we used the mouse model of chronic-plus-binge ethanol feeding and found that myeloid-specific deletion of the Ncf1 gene abolished ethanol-induced hepatic inflammation and steatosis. RNA-Seq analysis and the data from experimental models revealed that neutrophilic NCF1-dependent ROS promoted alcoholic hepatitis (AH) by inhibiting AMP-activated protein kinase (a key regulator of lipid metabolism) and microRNA-223 (a key antiinflammatory and antifibrotic microRNA). In conclusion, two distinct histopathological phenotypes based on liver immune phenotyping are observed in SAH patients, suggesting a separate mechanism driving liver injury and/or failure in these patients.
Collapse
Affiliation(s)
- Jing Ma
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH, Bethesda, Maryland, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Adrien Guillot
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH, Bethesda, Maryland, USA
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Zhihong Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bryan Mackowiak
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH, Bethesda, Maryland, USA
| | - Seonghwan Hwang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH, Bethesda, Maryland, USA
| | - Ogyi Park
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH, Bethesda, Maryland, USA
| | - Brandon J. Peiffer
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ali Reza Ahmadi
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Luma Melo
- Center for Liver Diseases, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Praveen Kusumanchi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nazmul Huda
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Romil Saxena
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yong He
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH, Bethesda, Maryland, USA
| | - Yukun Guan
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH, Bethesda, Maryland, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH, Bethesda, Maryland, USA
| | - Pau Sancho-Bru
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Mengwei Zang
- Department of Molecular Medicine, Barshop Institute for Longevity and Aging Studies, Center for Healthy Aging, University of Texas Health San Antonio, San Antonio, Texas, USA
- Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| | | | - Ramon Bataller
- Center for Liver Diseases, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism (NIAAA), NIH, Bethesda, Maryland, USA
| |
Collapse
|
41
|
MFG-E8 Knockout Aggravated Nonalcoholic Steatohepatitis by Promoting the Activation of TLR4/NF- κB Signaling in Mice. Mediators Inflamm 2022; 2022:5791915. [PMID: 35769208 PMCID: PMC9236848 DOI: 10.1155/2022/5791915] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/23/2022] [Accepted: 06/04/2022] [Indexed: 12/30/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the common liver disease characterized by hepatic steatosis, inflammation, and fibrosis; there are no approved drugs to treat this disease because of incomplete understanding of pathophysiological mechanisms of NASH. Milk fat globule-epidermal growth factor-factor 8 (MFG-E8), a multifunctional glycoprotein, has shown anti-inflammation and antifibrosis. Here, MFG-E8 was shown to play a key role in NASH progression. Using methionine and choline deficient (MCD) diet-fed mice, we found MFG-E8 knockout exacerbated hepatic damage and steatosis as indicated by increased plasma transaminases activities and hepatic histopathologic change, higher hepatic triglycerides (TGs), and lipid accumulation. Moreover, liver fibrosis and inflammation elicited by MCD were aggravated in MFG-E8 knockout mice. Mechanistically, MFG-E8 knockout facilitated activation of hepatic toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway in MCD-fed mice. In vitro experiment, the TLR4 specific antagonist TAK-242 rescued palmitic acid- (PA-) primed lipid formation and inflammation in MFG-E8 knockout primary murine hepatocytes. These findings indicated that MFG-E8 is involved in the progression of NASH and the possible mechanism by which MFG-E8 knockout exacerbated NASH in mice is associated with activation of the TLR4/NF-κB signaling pathway.
Collapse
|
42
|
Vaz K, Little R, Majeed A, Kemp W, Roberts SK. Determinants of Short- and Long-Term Outcomes of an Australian Cohort of Patients Admitted with Alcoholic Hepatitis. Dig Dis Sci 2022; 67:3356-3365. [PMID: 34231100 DOI: 10.1007/s10620-021-07140-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/26/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS Alcoholic hepatitis is a common condition with high mortality. This study aimed to firstly describe the presentation, treatment, and short- and long-term outcomes of an Australian cohort of patients admitted to hospital with alcoholic hepatitis and secondly to validate existing prognostic models. METHODS This is a retrospective study of consecutive patients admitted with alcoholic hepatitis to a major academic liver center in Melbourne, Australia, between January 1, 2010, and December 31, 2019. Cases were identified through appropriate International Classification of Diseases version 10 coding as well as review of non-coded patients with compatible biochemistry. Baseline demographic data, alcohol consumption, laboratory values, treatment, and outcomes at 30 days, 90 days, and 12 months post-diagnosis were collected from electronic medical records. Mortality data were extracted from an independent state government death registry. RESULTS In total, 126 patients (72 males [57%], median age 51 years) were included in the final analysis. Ninety-five (75%) were cirrhotic at diagnosis, 81 (64%) met criteria for severe alcoholic hepatitis, and 41 (33%) had an infection during their index admission. 54% of eligible patients were treated with corticosteroids. 30-day and 12-month mortality rates were 8.7% and 27.1%, respectively, with hepatic encephalopathy (hazard ratio 5.45) and neutrophil-to-lymphocyte ratio (hazard ratio 1.09) independent markers for 12-month mortality on Cox regression analysis. Glasgow alcoholic hepatitis score outperformed other major prognostic models for short-term mortality. CONCLUSIONS The 12-month mortality rate of 27% following alcoholic hepatitis is lower than previously reported studies, with hepatic encephalopathy and neutrophil-to-lymphocyte ratio predictive of long-term outcome.
Collapse
Affiliation(s)
- Karl Vaz
- Department of Gastroenterology and Hepatology, Alfred Health, Melbourne, Australia.
- Department of Gastroenterology and Hepatology, Austin Health, 145 Studley Rd, Heidelberg, VIC, 3084, Australia.
| | - Robert Little
- Department of Gastroenterology and Hepatology, Alfred Health, Melbourne, Australia
| | - Ammar Majeed
- Department of Gastroenterology and Hepatology, Alfred Health, Melbourne, Australia
| | - William Kemp
- Department of Gastroenterology and Hepatology, Alfred Health, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
| | - Stuart K Roberts
- Department of Gastroenterology and Hepatology, Alfred Health, Melbourne, Australia
- Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
43
|
Shariare MH, Pinky NJK, Abedin J, Kazi M, Aldughaim MS, Uddin MN. Liposomal Drug Delivery of Blumea lacera Leaf Extract: In-Vivo Hepatoprotective Effects. NANOMATERIALS 2022; 12:nano12132262. [PMID: 35808096 PMCID: PMC9268469 DOI: 10.3390/nano12132262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022]
Abstract
Background: Blumea lacera (B. lacera) is a herbaceous plant commonly found in south-east Asia. It shows significant therapeutic activities against various diseases. The objectives of this study were to evaluate hepatoprotective effects of Blumea lacera leaf extract and also to investigate the comparative effectiveness between a liposomal preparation and a suspension of B. lacera leaf extract against carbon tetrachloride (CCl4)-induced liver damage. Methods: B. lacera leaf extract was characterized using a GC-MS method. A liposomal preparation of B. lacera leaf extract was developed using an ethanol injection method and characterized using dynamic light scattering (DLS) and electronic microscopic systems. The hepatoprotective effects of B. lacera leaf extracts and its liposomal preparation were investigated using CCl4-induced liver damage in Long Evan rats. Results: GC-MS data showed the presence of different components (e.g., phytol) in the B. lacera leaf extract. DLS and microscopic data showed that a liposomal preparation of B. lacera leaf extracts was in the nano size range. In vivo study results showed that liposomal preparation and a suspension of B. lacera leaf extract normalized liver biochemical parameters, enzymes and oxidative stress markers which were elevated due to CCl4 administration. However, a liposomal formulation of B. lacera leaf extract showed significantly better hepatoprotective effects compared to a suspension of leaf extract. In addition, histopathological evaluation showed that B. lacera leaf extract and its liposomal preparation treatments decreased the extent of CCl4-induced liver inflammations. Conclusion: Results demonstrated that B. lacera leaf extract was effective against CCl4-induced liver injury possibly due to the presence of components such as phytol. A liposomal preparation exhibited significantly better activity compared to a B. lacera suspension, probably due to improved bioavailability and stability of the leaf extract.
Collapse
Affiliation(s)
- Mohammad Hossain Shariare
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh; (M.H.S.); (N.J.K.P.); (J.A.)
| | - Nusrat Jahan Khan Pinky
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh; (M.H.S.); (N.J.K.P.); (J.A.)
| | - Joynal Abedin
- Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh; (M.H.S.); (N.J.K.P.); (J.A.)
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
- Correspondence: (M.K.); (M.N.U.)
| | | | - Mohammad N. Uddin
- College of Pharmacy, Mercer University, 3001 Mercer University Drive, Atlanta, GA 30341, USA
- Correspondence: (M.K.); (M.N.U.)
| |
Collapse
|
44
|
Zhang X, Li J, Liu T, Zhao M, Liang B, Chen H, Zhang Z. Identification of Key Biomarkers and Immune Infiltration in Liver Tissue after Bariatric Surgery. DISEASE MARKERS 2022; 2022:4369329. [PMID: 35789605 PMCID: PMC9250435 DOI: 10.1155/2022/4369329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/13/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022]
Abstract
Background Few drugs are clearly available for nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH); nevertheless, mounting studies have provided sufficient evidence that bariatric surgery is efficient for multiple metabolic diseases, including NAFLD and NASH, while the molecular mechanisms are still poorly understood. Methods The mRNA expression profiling of GSE48452 and GSE83452 were retrieved and obtained from the Gene Expression Omnibus (GEO) database. The limma package was employed for identifying differentially expressed genes (DEGs), followed by clusterProfiler for performing Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, and GSEA software for performing GSEA analyses. The PPI network analyses were constructed using Metascape online analyses. WGCNA was also utilized to identify and verify the hub genes. CIBERSORT tools contributed to the analysis of immune cell infiltration of liver diseases. Results We identify coexpressed differential genes including 10 upregulated and 55 downregulated genes in liver tissue after bariatric surgery. GO and KEGG enrichment analyses indicated that DEGs were remarkably involved in the immune response. GSEA demonstrated that DEGs were markedly enriched in the immune response before surgery, while most were enriched in metabolism after surgery. Seven genes were screened through the MCC algorithm and KME values, including SRGN, CD53, EVI2B, MPEG1, NCKAP1L, LCP1, and TYROBP. The mRNA levels of these genes were verified in the Attie Lab Diabetes Database, and only LCP1 was found to have significant differences and correlation with certain immune cells. Conclusion Our knowledge of the mechanisms by which bariatric surgery benefits the liver and the discovery of LCP1 is expected to serve as potential biomarkers or therapeutic targets for NAFLD and NASH.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Pediatrics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jingxin Li
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tiancai Liu
- School of Laboratory Medicine and Biotechnology, Institute of Antibody Engineering, Southern Medical University, Guangzhou, China
| | - Min Zhao
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Baozhu Liang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hong Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhen Zhang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
45
|
Chen X, Lin L, Chen G, Yan H, Li Z, Xiao M, He X, Zhang F, Zhang Y. High Levels of DEAH-Box Helicases Relate to Poor Prognosis and Reduction of DHX9 Improves Radiosensitivity of Hepatocellular Carcinoma. Front Oncol 2022; 12:900671. [PMID: 35814441 PMCID: PMC9256992 DOI: 10.3389/fonc.2022.900671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundLiver hepatocellular carcinoma (LIHC), one of the most common primary malignancies, exhibits high levels of molecular and clinical heterogeneity. Increasing evidence has confirmed the important roles of some RNA helicase families in tumor development, but the function of the DEAH-box RNA helicase family in LIHC therapeutic strategies has not yet been clarified.MethodsThe LIHC dataset was downloaded from The Cancer Genome Atlas (TCGA). Consensus clustering was applied to group the patients. Least absolute shrinkage and selection operator Cox regression and univariate and multivariate Cox regression were used to develop and validate a prognostic risk model. The Tumor Immune Estimation Resource and Tumor Immune Single Cell Hub databases were used to explore the role of DEAH-box RNA helicases in LIHC immunotherapy. In vitro experiments were performed to investigate the role of DHX9 in LIHC radiosensitivity.ResultsTwelve survival-related DEAH-box RNA helicases were identified. High helicase expression levels were associated with a poor prognosis and clinical features. A prognostic model comprising six DEAH-box RNA helicases (DHX8, DHX9, DHX34, DHX35, DHX38, and DHX57) was constructed. The risk score of this model was found to be an independent prognostic indicator, and LIHC patients with different prognosis were distinguished by the model in the training and test cohorts. DNA damage repair pathways were also enriched in patients with high-risk scores. The six DEAH-box RNA helicases in the risk model were substantially related to innate immune cell infiltration and immune inhibitors. In vitro experiments showed that DHX9 knockdown improved radiosensitivity by increasing DNA damage.ConclusionThe DEAH-box RNA helicase signature can be used as a reliable prognostic biomarker for LIHC. In addition, DHX9 may be a definitive indicator and therapeutic target in radiotherapy and immunotherapy for LIHC.
Collapse
Affiliation(s)
- Xi Chen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Letao Lin
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Guanyu Chen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Huzheng Yan
- Department of Interventional Radiology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhenyu Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Meigui Xiao
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xu He
- Interventional Medical Center, Zhuhai People’s Hospital, Zhuhai, China
| | - Fujun Zhang
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- *Correspondence: Fujun Zhang, ; Yanling Zhang,
| | - Yanling Zhang
- Department of Minimally Invasive Interventional Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center (SYSUCC), Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- *Correspondence: Fujun Zhang, ; Yanling Zhang,
| |
Collapse
|
46
|
Kanazawa J, Kakisaka K, Suzuki Y, Yonezawa T, Abe H, Wang T, Takikawa Y. Excess fructose enhances oleatic cytotoxicity via reactive oxygen species production and causes necroptosis in hepatocytes. J Nutr Biochem 2022; 107:109052. [DOI: 10.1016/j.jnutbio.2022.109052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 02/07/2023]
|
47
|
Lu Y, Chang N, Zhao X, Xue R, Liu J, Yang L, Li L. Activated Neutrophils Secrete Chitinase-Like 1 and Attenuate Liver Inflammation by Inhibiting Pro-Inflammatory Macrophage Responses. Front Immunol 2022; 13:824385. [PMID: 35529851 PMCID: PMC9069964 DOI: 10.3389/fimmu.2022.824385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/21/2022] [Indexed: 11/26/2022] Open
Abstract
Excessive activation and recruitment of neutrophils are generally considered to be associated with pathological aggravation of multiple diseases. However, as the role of neutrophils in tissue injury repair is receiving increasing attention, it is necessary to further explore the beneficial role of activated neutrophils in promoting the resolution of inflammation after injury. In this study, we found that activated neutrophils have a crucial function in suppressing liver inflammation. In methionine-choline-deficient and high-fat (MCDHF) diet induced liver inflammation in mice, tail vein injection of activated neutrophils (A-Neu, stimulated by sphingosine 1-phosphate) inhibited the expressions of pro-inflammatory cytokines in the liver, including C-C chemokine motif ligand 4, tumor necrosis factor and nitric oxide synthase 2, and attenuated liver injury. However, non-activated neutrophils (N-Neu) did not have these effects. In vitro, pro-inflammatory macrophages were co-cultured with N-Neu or A-Neu by transwell, respectively. A-Neu was found to suppress the pro-inflammatory phenotype of macrophages by using RT-qPCR, western blot and cytometric bead array. Microarray analysis showed that there were systematic variations in transcript expression levels between N-Neu and A-Neu. GeneVenn software was used to show the gene expression overlap between GO terms including Regulation of Cell Communication, Cytokine Secretion, Inflammatory Response and Extracellular Space clusters. We identified that Chitinase-like 1 (CHIL1) secreted by S1P activated neutrophils may be an important mediators affecting the pro-inflammatory macrophage responses. In the injured liver of mice induced by MCDHF diet, the expression of Chil1 mRNA increased and was positively correlated with the neutrophil marker Ly6g. Moreover, the secretion of CHIL1 in A-Neu increased significantly. Strikingly, the effect of A-Neu on macrophage response was reproduced by incubating pro-inflammatory macrophages with recombinant CHIL1. A-Neu conditioned medium were incubated with CHIL1 antibody-conjugated protein G beads, magnetically separated to immunodepletion CHIL1 from the A-Neu supernatant, which can partially weaken its inhibitory effect of A-Neu on the production of macrophage pro-inflammatory cytokines. Together, the conclusions indicated that A-Neu could inhibit the pro-inflammatory macrophage responses by secreting CHIL1, thereby effectively inhibiting liver inflammation.
Collapse
|
48
|
Carson JP, Robinson MW, Ramm GA, Gobert GN. Synthetic peptides derived from the Schistosoma mansoni secretory protein Sm16 induce contrasting responses in hepatic stellate cells. Exp Parasitol 2022; 236-237:108255. [PMID: 35385714 DOI: 10.1016/j.exppara.2022.108255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/04/2022]
|
49
|
Nava-Santana C, Rodríguez-Armida M, Jiménez JV, Vargas-Parra N, León DEA, Campos-Murguia A, Macías-Rodriguez R, Arteaga-Garrido A, Hernández-Villegas AC, Dominguez-Cherit G, Rivero-Sigarroa E, Gamboa-Dominguez A, Gullias-Herrero A, Sifuentes-Osornio J, Uribe-Uribe NO, Morales-Buenrostro LE. Clinicopathologic characteristics of severe COVID-19 patients in Mexico City: A post-mortem analysis using a minimally invasive autopsy approach. PLoS One 2022; 17:e0262783. [PMID: 35239660 PMCID: PMC8893646 DOI: 10.1371/journal.pone.0262783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/04/2022] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Describe the histological findings of minimally ultrasound-guided invasive autopsies in deceased patients with severe SARS-CoV-2 and compare the diagnostic yield with open autopsies. DESIGN Observational post-mortem cohort study. Minimally invasive ultrasound-guided autopsies were performed in fourteen deceased patients with a confirmed diagnosis of SARS-CoV-2 pneumonia. Histological and clinical findings of lung, kidney, and liver tissue are described and contrasted with those previously reported in the literature. SETTING Single-center COVID-19 reference center in Mexico City. RESULTS Fourteen minimally invasive autopsies revealed a gross correlation with open autopsies reports: 1) Lung histology was characterized mainly by early diffuse alveolar damage (12/13). Despite low lung compliances and prolonged mechanical ventilation, the fibrotic phase was rarely observed (2/13). 2) Kidney histopathology demonstrated acute tubular injury (12/13), interstitial nephritis (11/13), and glomerulitis (11/13) as the predominant features 3) Liver histology was characterized by neutrophilic inflammation in all of the cases, as well as hepatic necrosis (8/14) despite minimal alterations in liver function testing. Hepatic steatosis was observed in most cases (12/14). SARS-CoV-2 positivity was widely observed throughout the immunohistochemical analysis. However, endothelitis and micro thrombosis, two of the hallmark features of the disease, were not observed. CONCLUSION Our data represents the largest minimally invasive, ultrasound-guided autopsy report. We demonstrate a gross histological correlation with large open autopsy cohorts. However, this approach might overlook major histologic features of the disease, such as endothelitis and micro-thrombosis. Whether this represents sampling bias is unclear.
Collapse
Affiliation(s)
- Carlos Nava-Santana
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - María Rodríguez-Armida
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - José Víctor Jiménez
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Nancy Vargas-Parra
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Diana E. Aguilar León
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Alejandro Campos-Murguia
- Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Ricardo Macías-Rodriguez
- Department of Gastroenterology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Andrés Arteaga-Garrido
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | | | - Guillermo Dominguez-Cherit
- Department of Critical Care Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Eduardo Rivero-Sigarroa
- Department of Critical Care Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Armando Gamboa-Dominguez
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Alfonso Gullias-Herrero
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - José Sifuentes-Osornio
- Department of Medicine, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Norma Ofelia Uribe-Uribe
- Department of Pathology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Luis E. Morales-Buenrostro
- Department of Nephrology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| |
Collapse
|
50
|
Tsukanov VV, Gorchilova EG, Kolenchukova OA, Gvozdev II, Savchenko AA, Kasparov EV, Vasyutin AV, Tonkikh JL, Borisov AG, Rzhavicheva OS. Functional activity of blood neutrophilic granulocytes in patients with opisthorchiasis, depending on the severity of liver fibrosis. TERAPEVT ARKH 2021; 93:1271-1277. [PMID: 36286648 DOI: 10.26442/00403660.2021.11.201165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 11/22/2022]
Abstract
Aim. To study the activity of neutrophilic granulocytes in patients with opisthorchiasis, depending on the severity of liver fibrosis.
Materials and methods. A total of 74 patients with chronic opisthorchiasis (39 men and 35 women, average age 42.3 years) and 32 practically healthy patients (17 men and 15 women, average age 41.5 years) aged 24 to 60 years were examined. Diagnosis of opisthorchiasis was carried out by two methods: coprooscopy and identification of eggs or bodies of adult parasites in duodenal content. Liver fibrosis was determined by the method of elastometry according to the METAVIR scale in all 74 patients with opisthorchiasis. The study of the functional activity of neutrophils in the blood was performed to all 74 patients with opisthorchiasis and 32 healthy individuals from the control group by chemiluminescent analysis with measurement of the reactive oxygen species (ROS) production intensity in a spontaneous and zymosan-induced reaction in lucigenin and luminol-dependent processes.
Results. Liver fibrosis F2 by METAVIR was registered in 20.3% of the examined individuals, liver fibrosis F3F4 by METAVIR was detected in 17.6% of patients with opisthorchiasis. In patients with opisthorchiasis with liver fibrosis F3F4 by METAVIR, a significant decrease in the functional activity of neutrophilic granulocytes was registered in comparison with individuals with liver fibrosis F0F1 by METAVIR, as evidenced by a significant decrease in the maximum intensity of ROS production (Imax) and the area under the curve (S) chemiluminescence in lucigenin and luminol-dependent processes both in the spontaneous and zymosan-induced reaction.
Conclusion. These results provide new information to explain the mechanisms of liver fibrosis in patients opisthorchiasis and create opportunities for the development of diagnostics and preventive technologies.
Collapse
|