1
|
Wang J, Li J, Buist-Homan M, Harmsen MC, Moshage H. Extracellular vesicle-dependent crosstalk between hepatic stellate cells and Kupffer cells promotes their mutual activation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167914. [PMID: 40449190 DOI: 10.1016/j.bbadis.2025.167914] [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: 02/26/2025] [Revised: 04/30/2025] [Accepted: 05/16/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND & AIMS Hepatic fibrosis results from hepatic stellate cell (HSC) activation and excessive extracellular matrix (ECM) deposition, driven by chronic inflammation. Kupffer cells (KCs) play a central role in HSC activation. We previously showed that HSC-secreted factors, particularly extracellular vesicles (EVs), activate KCs. However, the reciprocal effects of activated KCs on HSCs remain poorly understood. This study investigates the bidirectional crosstalk between HSCs and KCs, focusing on the role of KC-derived EVs in regulating HSC activation and fibrosis progression. METHODS Primary HSCs and KCs were isolated from male Wistar rats. HSCs were co-cultured with KCs for 24 h to assess inflammatory and activation markers. LPS-stimulated KC-derived EVs and controls were administered to HSCs on day 1. LPS and the Toll-like receptor 4 (TLR4) inhibitor TAK-242 were used to investigate the intercellular communication in detail. RESULTS Co-cultured HSCs and KCs showed mutual activation, demonstrated by elevated inflammatory markers in both cell types and enhanced HSC pro-fibrotic activation. Pro-inflammatory (LPS)-activated KCs amplified HSC activation in a TLR4-dependent fashion. Part of this augmented HSC activation was attributed to EVs. CONCLUSIONS In co-culture, KCs and HSCs show mutual activation in a TLR4-dependent fashion. This bidirectional activation is augmented by pro-inflammatory mediators. KC-derived EVs (partially) activate HSCs, which might contribute to progression of liver fibrosis in vivo. Modulating KC activation, such as by blocking TLR4 signaling, may alter EV secretion or cargo composition, reducing HSC activation and fibrosis progression. Targeting this EV-mediated crosstalk could provide novel therapeutic strategies for liver fibrosis.
Collapse
Affiliation(s)
- Junyu Wang
- University Medical Center Groningen, University of Groningen, Department of Gastroenterology and Hepatology, Groningen, the Netherlands; University Medical Center Groningen, University of Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands
| | - Jia Li
- University Medical Center Groningen, University of Groningen, Department of Gastroenterology and Hepatology, Groningen, the Netherlands
| | - Manon Buist-Homan
- University Medical Center Groningen, University of Groningen, Department of Gastroenterology and Hepatology, Groningen, the Netherlands
| | - Martin C Harmsen
- University Medical Center Groningen, University of Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science, Groningen, the Netherlands; University of Groningen, University Medical Centre Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands
| | - Han Moshage
- University Medical Center Groningen, University of Groningen, Department of Gastroenterology and Hepatology, Groningen, the Netherlands.
| |
Collapse
|
2
|
Vahrenbrink M, Coleman CD, Kuipers S, Lurje I, Hammerich L, Kunkel D, Keye J, Dittrich S, Schjeide BM, Hiß R, Müller J, Püschel GP, Henkel J. Dynamic changes in macrophage populations and resulting alterations in Prostaglandin E 2 sensitivity in mice with diet-induced MASH. Cell Commun Signal 2025; 23:227. [PMID: 40380177 DOI: 10.1186/s12964-025-02222-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND The transition from metabolic dysfunction-associated steatotic liver disease (MASLD) to steatohepatitis (MASH) is characterized by a chronic low-grade inflammation, involving activation of resident macrophages (Kupffer cells; KC) and recruitment of infiltrating macrophages. Macrophages produce cytokines and, after induction of Cyclooxygenase 2 (COX-2), the key enzyme of prostanoid synthesis, prostaglandin E2 (PGE2). PGE2 modulates cytokine production in an autocrine and paracrine manner, therefore playing a pivotal role in regulating inflammatory processes. Changes in the hepatic macrophage pool during MASLD progression to MASH could influence PGE2- and cytokine-mediated signaling processes. The aim of this study was to characterize these changes in mice with diet-induced MASH and further elucidate the role of COX-2-dependently formed PGE2 on the inflammatory response in different macrophage populations of mice with a macrophage-specific COX-2-deletion. METHODS Male, 6-7-week-old wildtype mice were fed either a Standard or high-fat, high-cholesterol MASH-inducing diet for 4, 12 and 20 weeks. Liver macrophages were isolated and analyzed by flow cytometry. For in vitro experiments primary KC, peritoneal macrophages (PM) and Bone-marrow-derived macrophages (BMDM) were isolated from macrophage-specific COX-2-deficient and wildtype mice and treated with lipopolysaccharide (LPS) and/or PGE2. RESULTS During MASH-development, the proportion of KC (Clec4F+Tim4+) decreased, while the proportion of monocyte-derived macrophages (Clec4F-Tim4-) and monocyte-derived cells exhibiting a phenotype similar to KC (Clec4F+Tim4-) significantly increased over time. In vitro experiments showed that exogenous PGE2 completely abrogated the LPS-induced mRNA expression and secretion of tumor necrosis factor-alpha (TNF-α) in primary KC, PM and BMDM from wildtype mice. PM and BMDM, as in vitro models for infiltrating macrophages, were more sensitive to PGE2 compared to KC. Deletion of COX-2 in all macrophage populations led to an impaired PGE2-dependent feedback inhibition of TNF-α production. LPSinduced TNF-α mRNA expression was higher compared to the respective wildtype macrophage population. CONCLUSION The current study, using a murine MASH model, indicates that PGE2 may have a protective, anti-inflammatory effect, especially by inhibiting the expression of pro-inflammatory cytokines such as TNFα in infiltrating monocyte-derived macrophages. An inhibition of endogenous PGE2 synthesis in macrophages by pharmacological inhibition of COX-2 could potentially increase inflammation and promote the progression of MASH.
Collapse
Affiliation(s)
- Madita Vahrenbrink
- Department of Nutritional Biochemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.
- Max Rubner Center (MRC) for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité-Universitätsmedizin Berlin, Hessische Straße 3-4, 10115, Berlin, Germany.
| | - C D Coleman
- Department of Nutritional Biochemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - S Kuipers
- Department of Nutritional Biochemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - I Lurje
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - L Hammerich
- Department of Hepatology and Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - D Kunkel
- Flow & Mass Cytometry Core Facility, Berlin Institute of Health at Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - J Keye
- Flow & Mass Cytometry Core Facility, Berlin Institute of Health at Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - S Dittrich
- Nutritional Biochemistry, Faculty of Life Sciences: Food, Nutrition and Health, University of Bayreuth, Kulmbach, Germany
| | - B M Schjeide
- Nutritional Biochemistry, Faculty of Life Sciences: Food, Nutrition and Health, University of Bayreuth, Kulmbach, Germany
| | - R Hiß
- Department of Nutritional Biochemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - J Müller
- Physics and Computer Sciences, Applied Computer Sciences VIII, Faculty of Mathematics, University of Bayreuth, Bayreuth, Germany
| | - G P Püschel
- Department of Nutritional Biochemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - J Henkel
- Department of Nutritional Biochemistry, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
- Nutritional Biochemistry, Faculty of Life Sciences: Food, Nutrition and Health, University of Bayreuth, Kulmbach, Germany
| |
Collapse
|
3
|
Han M, Qing Y, Fu J, He W, Huang J, Zhu X, Yang L, Yao L, Peng T, Wang Z, Li Z, Wu L, Yang Q, Hu B, Lv Y, Zhang H, Wan L, Meng X, Wang F, Qin S, Zhang Y, Wang Z. Mechanism of Jizhi syrup's prevention and treatment of acute bronchitis based on LPS-iNOS inflammatory mediators' signalling. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118708. [PMID: 39197804 DOI: 10.1016/j.jep.2024.118708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/03/2024] [Accepted: 08/17/2024] [Indexed: 09/01/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jizhi syrup (JZTJ) is composed of eight medicinal herbs, including Houttuynia cordata, Fagopyrum dibotrys, Ilex chinensis, Ephedra sinica, Aster tataricus, Peucedanum praeruptorum, Citrus aurantium and Glycyrrhiza uralensis. It is mainly used for coughing caused by exogenous wind heat. Symptoms include fever, aversion to cold, chest and diaphragm tightness, cough and sore throat; and acute bronchitis and acute exacerbation of chronic bronchitis with the above symptoms. PURPOSE This study aimed to preliminary analyse the chemical components in the liposoluble part of JZTJ, evaluate the anti-inflammatory effect of JZTJ by using six animal and cell models and predict the target and mechanism of acute bronchitis prevention and treatment with JZTJ. METHODS The chemical components in the liposoluble fraction of JZTJ (extracted by cyclohexane) were quantitatively analysed using gas chromatography-mass spectrometry (GC-MS). Classic non-specific inflammation models and acute bronchitis models were established to systematically evaluate the anti-inflammatory effect of JZTJ. The anti-inflammatory intensity and characteristics of three doses of JZTJ were comprehensively compared on the basis of principal component analysis method at the cellular and overall animal levels. By using lipopolysaccharides (LPSs) as modelling factors, a RAW264.7 macrophage inflammatory response model and a rat acute bronchitis model were created to study the effect of JZTJ on the in-vitro and - vivo LPS-iNOS-inflammatory mediators' inflammatory signalling pathway to reveal the mechanism of acute bronchitis prevention and treatment by JZTJ at the levels of genes, proteins, and inflammatory mediators. RESULTS Seventeen alkane and ester compounds were preliminarily qualitatively identified from the lipid soluble fraction of JZTJ: dibutyl phthalate, tetradecane, ridecane, n-hexadecanoic acid, pentadecane, n-decanoic acid, 2,6,10,14,18,22-tetracosahexaene, 2,6,10,15,19,23-hexamethyl-(all-E)-; phenol, 2,2'-methylenebis[6-(1,1-dimethylethyl)-4-methyl-; hexadecane. JZTJ has a significant inhibitory effect on acute non-specific inflammation, specifically inhibiting 'xylene-induced ear swelling in mice', 'acetic acid-induced increased permeability of abdominal capillaries in mice' and 'egg white-induced foot swelling in rats'. The above effects are most evident in high doses, followed by medium doses, whereas low doses have poorer or no effects. JZTJ can prevent and treat acute bronchitis induced by LPS in mice and rats, significantly improve the pathological changes in patchy interstitial and alveolar bleeding with excessive neutrophil infiltration and inhibit the release of inflammatory mediators by LPS-induced RAW264.7 macrophages. Its mechanism of action may be by downregulating the phosphorylation level of p-ERK1/2 protein, thereby inhibiting inducible nitric oxide synthase (iNOS) mRNA, tumour necrosis factor (TNF)-α mRNA and IL-1β. The expression levels of genes, such as mRNA and IL-6 mRNA, thereby reducing iNOS, TNF-α and IL-1β. The expression of proteins in the cytoplasm of lung and bronchial tissue cells reduced the release of downstream inflammatory mediators NO and IL-6. CONCLUSION Preliminary analysis of the chemical components in the lipid soluble fraction of JZTJ can lay the foundation for subsequent research on its effective components. Evaluating the anti-inflammatory effect of JZTJ is helpful for further research on its mechanism of action. The anti-inflammatory effects are exerted by regulating the inflammatory signalling pathway of LPS-iNOS inflammatory mediators, providing a scientific basis for their clinical application.
Collapse
Affiliation(s)
- Mengtian Han
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuling Qing
- Taiji Group Co., Ltd., Chongqing, 408099, China
| | - Jiaqing Fu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wencan He
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jing Huang
- Taiji Group Co., Ltd., Chongqing, 408099, China
| | - Xiaoqi Zhu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lijuan Yang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China
| | - Lincai Yao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Peng
- Taiji Group Co., Ltd., Chongqing, 408099, China
| | - Zhihua Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zhangyu Li
- Taiji Group Co., Ltd., Chongqing, 408099, China
| | - Lian Wu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | | | - Boyang Hu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yongjun Lv
- Taiji Group Co., Ltd., Chongqing, 408099, China
| | - Hai Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li Wan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xianli Meng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fei Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, 610213, China.
| | - Shaorong Qin
- Taiji Group Co., Ltd., Chongqing, 408099, China.
| | - Yi Zhang
- College of Ethnomedicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Zhang Wang
- College of Ethnomedicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
4
|
Tang S, Wu S, Zhang W, Ma L, Zuo L, Wang H. Immunology and treatments of fatty liver disease. Arch Toxicol 2025; 99:127-152. [PMID: 39692857 DOI: 10.1007/s00204-024-03920-1] [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: 09/05/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) are two major chronic liver diseases worldwide. The triggers for fatty liver can be derived from external sources such as adipose tissue, the gut, personal diet, and genetics, or internal sources, including immune cell responses, lipotoxicity, hepatocyte death, mitochondrial dysfunction, and extracellular vesicles. However, their pathogenesis varies to some extent. This review summarizes various immune mechanisms and therapeutic targets associated with these two types of fatty liver disease. It describes the gut-liver axis and adipose tissue-liver crosstalk, as well as the roles of different immune cells (both innate and adaptive immune cells) in fatty liver disease. Additionally, mitochondrial dysfunction, extracellular vesicles, microRNAs (miRNAs), and gastrointestinal hormones are also related to the pathogenesis of fatty liver. Understanding the pathogenesis of fatty liver and corresponding therapeutic strategies provides a new perspective for developing novel treatments for fatty liver disease.
Collapse
Affiliation(s)
- Sainan Tang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Shanshan Wu
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Wenzhe Zhang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Lili Ma
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Li Zuo
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China.
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
5
|
Mercado-Gómez M, Goikoetxea-Usandizaga N, Kerbert AJC, Gracianteparaluceta LU, Serrano-Maciá M, Lachiondo-Ortega S, Rodriguez-Agudo R, Gil-Pitarch C, Simón J, González-Recio I, Fondevila MF, Santamarina-Ojeda P, Fraga MF, Nogueiras R, Heras JDL, Jalan R, Martínez-Chantar ML, Delgado TC. The lipopolysaccharide-TLR4 axis regulates hepatic glutaminase 1 expression promoting liver ammonia build-up as steatotic liver disease progresses to steatohepatitis. Metabolism 2024; 158:155952. [PMID: 38906371 DOI: 10.1016/j.metabol.2024.155952] [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: 02/22/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/23/2024]
Abstract
INTRODUCTION Ammonia is a pathogenic factor implicated in the progression of metabolic-associated steatotic liver disease (MASLD). The contribution of the glutaminase 1 (GLS) isoform, an enzyme converting glutamine to glutamate and ammonia, to hepatic ammonia build-up and the mechanisms underlying its upregulation in metabolic-associated steatohepatitis (MASH) remain elusive. METHODS Multiplex transcriptomics and targeted metabolomics analysis of liver biopsies in dietary mouse models representing the whole spectra of MASLD were carried out to characterize the relevance of hepatic GLS during disease pathological progression. In addition, the acute effect of liver-specific GLS inhibition in hepatic ammonia content was evaluated in cultured hepatocytes and in in vivo mouse models of diet-induced MASLD. Finally, the regulatory mechanisms of hepatic GLS overexpression related to the lipopolysaccharide (LPS)/Toll-like receptor 4 (TLR4) axis were explored in the context of MASH. RESULTS In mouse models of diet-induced MASLD, we found that augmented liver GLS expression is closely associated with the build-up of hepatic ammonia as the disease progresses from steatosis to steatohepatitis. Importantly, the acute silencing/pharmacological inhibition of GLS diminishes the ammonia burden in cultured primary mouse hepatocytes undergoing dedifferentiation, in steatotic hepatocytes, and in a mouse model of diet-induced steatohepatitis, irrespective of changes in ureagenesis and gut permeability. Under these conditions, GLS upregulation in the liver correlates positively with the hepatic expression of TLR4 that recognizes LPS. In agreement, the pharmacological inhibition of TLR4 reduces GLS and hepatic ammonia content in LPS-stimulated mouse hepatocytes and hyperammonemia animal models of endotoxemia. CONCLUSIONS Overall, our results suggest that the LPS/TLR4 axis regulates hepatic GLS expression promoting liver ammonia build-up as steatotic liver disease progresses to steatohepatitis.
Collapse
Affiliation(s)
- Maria Mercado-Gómez
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Naroa Goikoetxea-Usandizaga
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Annarein J C Kerbert
- Liver Failure Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, United Kingdom
| | | | - Marina Serrano-Maciá
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Sofia Lachiondo-Ortega
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Rubén Rodriguez-Agudo
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Clàudia Gil-Pitarch
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Jorge Simón
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain
| | - Irene González-Recio
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Marcos F Fondevila
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain
| | - Pablo Santamarina-Ojeda
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; Spanish Biomedical Research Network in Rare Diseases (CIBERER), 28029 Madrid, Spain; Nanomaterials and Nanotechnology Research Center (CINN), Spanish National Research Council (CSIC), 33940 El Entrego, Asturias, Spain
| | - Mario F Fraga
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; Spanish Biomedical Research Network in Rare Diseases (CIBERER), 28029 Madrid, Spain; Nanomaterials and Nanotechnology Research Center (CINN), Spanish National Research Council (CSIC), 33940 El Entrego, Asturias, Spain; Institute of Oncology of Asturias (IUOPA), University of Oviedo, 33006 Oviedo, Asturias, Spain
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), 28029 Madrid, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Javier de Las Heras
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; Division of Paediatric Metabolism, CIBERER, MetabERN, Cruces University Hospital, 48903 Barakaldo, Spain.; Department of Paediatrics, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Rajiv Jalan
- Liver Failure Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus, London, United Kingdom
| | - María Luz Martínez-Chantar
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 28029 Madrid, Spain.
| | - Teresa C Delgado
- Liver Disease Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain; Biobizkaia Health Research Institute, 48903 Barakaldo, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
6
|
Huai Q, Zhu C, Zhang X, Dai H, Li X, Wang H. Mesenchymal stem/stromal cells armored by FGF21 ameliorate alcohol-induced liver injury through modulating polarization of macrophages. Hepatol Commun 2024; 8:e0410. [PMID: 38551384 PMCID: PMC10984668 DOI: 10.1097/hc9.0000000000000410] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/01/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Alcohol-associated liver disease (ALD) is a major health care challenge worldwide with limited therapeutic options. Although mesenchymal stem/stromal cells (MSCs) represent a newly emerging therapeutic approach to treat ALD, thus far, there have been extensive efforts to try and enhance their efficacy, including genetically engineering MSCs. FGF21, an endocrine stress-responsive hormone, has been shown to regulate energy balance, glucose, and lipid metabolism and to enhance the homing of MSCs toward injured sites. Therefore, the purpose of this study was to investigate whether MSCs that overexpress FGF21 (FGF21-MSCs) improve the therapeutic effect of MSCs in treating ALD. METHODS Human umbilical cord-derived MSCs served as the gene delivery vehicle for the FGF21 gene. Human umbilical cord-derived MSCs were transduced with the FGF21 gene using lentiviral vectors to mediate FGF21 overexpression. We utilized both chronic Lieber-DeCarli and Gao-binge models of ethanol-induced liver injury to observe the therapeutic effect of FGF21-MSCs. Liver injury was phenotypically evaluated by performing biochemical methods, histology, and inflammatory cytokine levels. RESULTS Compared with MSCs alone, administration of MSCs overexpressing FGF21(FGF21-MSCs) treatment significantly enhanced the therapeutic effect of ALD in mice, as indicated by the alleviation of liver injury with reduced steatosis, inflammatory infiltration, oxidative stress, and hepatic apoptosis, and the promotion of liver regeneration. Mechanistically, FGF21 could facilitate the immunomodulatory function of MSCs on macrophages by setting metabolic commitment for oxidative phosphorylation, which enables macrophages to exhibit anti-inflammatory inclination. CONCLUSIONS Our data elucidate that MSC modification by FGF21 could enhance their therapeutic effect in ALD and may help in the exploration of effective MSCs-based cell therapies for the treatment of ALD.
Collapse
Affiliation(s)
- Qian Huai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Inflammation and Immune-mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Cheng Zhu
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Inflammation and Immune-mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Xu Zhang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Inflammation and Immune-mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Hanren Dai
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Inflammation and Immune-mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Xiaolei Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Inflammation and Immune-mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Inflammation and Immune-mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
Dumitru A, Matei E, Cozaru GC, Chisoi A, Alexandrescu L, Popescu RC, Butcaru MP, Dumitru E, Rugină S, Tocia C. Endotoxin Inflammatory Action on Cells by Dysregulated-Immunological-Barrier-Linked ROS-Apoptosis Mechanisms in Gut-Liver Axis. Int J Mol Sci 2024; 25:2472. [PMID: 38473721 DOI: 10.3390/ijms25052472] [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: 12/27/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
Our study highlighted the immune changes by pro-inflammatory biomarkers in the gut-liver-axis-linked ROS-cell death mechanisms in chronic and acute inflammations when gut cells are exposed to endotoxins in patients with hepatic cirrhosis or steatosis. In duodenal tissue samples, gut immune barrier dysfunction was analyzed by pro-inflammatory biomarker expressions, oxidative stress, and cell death by flow cytometry methods. A significant innate and adaptative immune system reaction was observed as result of persistent endotoxin action in gut cells in chronic inflammation tissue samples recovered from hepatic cirrhosis with the A-B child stage. Instead, in patients with C child stage of HC, the endotoxin tolerance was installed in cells, characterized by T lymphocyte silent activation and increased Th1 cytokines expression. Interesting mechanisms of ROS-cell death were observed in chronic and acute inflammation samples when gut cells were exposed to endotoxins and immune changes in the gut-liver axis. Late apoptosis represents the chronic response to injury induction by the gut immune barrier dysfunction, oxidative stress, and liver-dysregulated barrier. Meanwhile, necrosis represents an acute and severe reply to endotoxin action on gut cells when the immune system reacts to pro-inflammatory Th1 and Th2 cytokines releasing, offering protection against PAMPs/DAMPs by monocytes and T lymphocyte activation. Flow cytometric analysis of pro-inflammatory biomarkers linked to oxidative stress-cell death mechanisms shown in our study recommends laboratory techniques in diagnostic fields.
Collapse
Affiliation(s)
- Andrei Dumitru
- Gastroenterology Department, "Sf. Apostol Andrei" Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
- Medicine Faculty, "Ovidius" University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania
| | - Elena Matei
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, "Ovidius" University of Constanta, 145 Tomis Blvd., 900591 Constanta, Romania
| | - Georgeta Camelia Cozaru
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, "Ovidius" University of Constanta, 145 Tomis Blvd., 900591 Constanta, Romania
- Clinical Service of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
- Medical Sciences Academy, 1 I.C. Bratianu Street, 030167 Bucharest, Romania
| | - Anca Chisoi
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, "Ovidius" University of Constanta, 145 Tomis Blvd., 900591 Constanta, Romania
- Clinical Service of Pathology, "Sf. Apostol Andrei" Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
- Medical Sciences Academy, 1 I.C. Bratianu Street, 030167 Bucharest, Romania
| | - Luana Alexandrescu
- Gastroenterology Department, "Sf. Apostol Andrei" Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
- Medicine Faculty, "Ovidius" University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania
| | - Răzvan Cătălin Popescu
- Medicine Faculty, "Ovidius" University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania
| | - Mihaela Pundiche Butcaru
- Medicine Faculty, "Ovidius" University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania
| | - Eugen Dumitru
- Gastroenterology Department, "Sf. Apostol Andrei" Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
- Medicine Faculty, "Ovidius" University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, "Ovidius" University of Constanta, 145 Tomis Blvd., 900591 Constanta, Romania
- Academy of Romanian Scientist, 3 Ilfov Street, 050044 Bucharest, Romania
| | - Sorin Rugină
- Medicine Faculty, "Ovidius" University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania
- Academy of Romanian Scientist, 3 Ilfov Street, 050044 Bucharest, Romania
| | - Cristina Tocia
- Gastroenterology Department, "Sf. Apostol Andrei" Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
- Medicine Faculty, "Ovidius" University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania
| |
Collapse
|
8
|
Zhang W, Liu K, Ren GM, Wang Y, Wang T, Liu X, Li DX, Xiao Y, Chen X, Li YT, Zhan YQ, Xiang SS, Chen H, Gao HY, Zhao K, Yu M, Ge CH, Li CY, Ge ZQ, Yang XM, Yin RH. BRISC is required for optimal activation of NF-κB in Kupffer cells induced by LPS and contributes to acute liver injury. Cell Death Dis 2023; 14:743. [PMID: 37968261 PMCID: PMC10651896 DOI: 10.1038/s41419-023-06268-z] [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: 07/31/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/17/2023]
Abstract
BRISC (BRCC3 isopeptidase complex) is a deubiquitinating enzyme that has been linked with inflammatory processes, but its role in liver diseases and the underlying mechanism are unknown. Here, we investigated the pathophysiological role of BRISC in acute liver failure using a mice model induced by D-galactosamine (D-GalN) plus lipopolysaccharide (LPS). We found that the expression of BRISC components was dramatically increased in kupffer cells (KCs) upon LPS treatment in vitro or by the injection of LPS in D-GalN-sensitized mice. D-GalN plus LPS-induced liver damage and mortality in global BRISC-null mice were markedly attenuated, which was accompanied by impaired hepatocyte death and hepatic inflammation response. Constantly, treatment with thiolutin, a potent BRISC inhibitor, remarkably alleviated D-GalN/LPS-induced liver injury in mice. By using bone marrow-reconstituted chimeric mice and cell-specific BRISC-deficient mice, we demonstrated that KCs are the key effector cells responsible for protection against D-GalN/LPS-induced liver injury in BRISC-deficient mice. Mechanistically, we found that hepatic and circulating levels of TNF-α, IL-6, MCP-1, and IL-1β, as well as TNF-α- and MCP-1-producing KCs, in BRISC-deleted mice were dramatically decreased as early as 1 h after D-GalN/LPS challenge, which occurred prior to the elevation of the liver injury markers. Moreover, LPS-induced proinflammatory cytokines production in KCs was significantly diminished by BRISC deficiency in vitro, which was accompanied by potently attenuated NF-κB activation. Restoration of NF-κB activation by two small molecular activators of NF-κB p65 effectively reversed the suppression of cytokines production in ABRO1-deficient KCs by LPS. In conclusion, BRISC is required for optimal activation of NF-κB-mediated proinflammatory cytokines production in LPS-treated KCs and contributes to acute liver injury. This study opens the possibility to develop new strategies for the inhibition of KCs-driven inflammation in liver diseases.
Collapse
Affiliation(s)
- Wen Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, 300134, China
| | - Kai Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Guang-Ming Ren
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yu Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui Province, China
| | - Ting Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
| | - Xian Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Dong-Xu Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Yang Xiao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xu Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ya-Ting Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
| | - Yi-Qun Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Shen-Si Xiang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hui Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hui-Ying Gao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ke Zhao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Miao Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chang-Hui Ge
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zhi-Qiang Ge
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Xiao-Ming Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China.
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Rong-Hua Yin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
- Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
9
|
Chaudhry H, Sohal A, Iqbal H, Roytman M. Alcohol-related hepatitis: A review article. World J Gastroenterol 2023; 29:2551-2570. [PMID: 37213401 PMCID: PMC10198060 DOI: 10.3748/wjg.v29.i17.2551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/10/2023] [Accepted: 04/13/2023] [Indexed: 05/23/2023] Open
Abstract
Alcohol-related hepatitis (ARH) is a unique type of alcohol-associated liver disease characterized by acute liver inflammation caused by significant alcohol use. It ranges in severity from mild to severe and carries significant morbidity and mortality. The refinement of scoring systems has enhanced prognostication and guidance of clinical decision-making in the treatment of this complex disease. Although treatment focuses on supportive care, steroids have shown benefit in select circumstances. There has been a recent interest in this disease process, as coronavirus disease 2019 pandemic led to substantial rise in cases. Although much is known regarding the pathogenesis, prognosis remains grim due to limited treatment options. This article summarizes the epidemiology, genetics, pathogenesis, diagnosis and treatment of ARH.
Collapse
Affiliation(s)
- Hunza Chaudhry
- Department of Internal Medicine, University of California, San Francisco, Fresno, CA 93701, United States
| | - Aalam Sohal
- Department of Hepatology, Liver Institute Northwest, Seattle, WA 98105, United States
| | - Humzah Iqbal
- Department of Internal Medicine, University of California, San Francisco, Fresno, CA 93701, United States
| | - Marina Roytman
- Department of Gastroenterology and Hepatology, University of California, San Francisco, Fresno, CA 93701, United States
| |
Collapse
|
10
|
Zhao J, Jeong H, Yang D, Tian W, Kim JW, Woong Lim C, Kim B. Toll-like receptor-7 signaling in Kupffer cells exacerbates concanavalin A-induced liver injury in mice. Int Immunopharmacol 2023; 119:110238. [PMID: 37126986 DOI: 10.1016/j.intimp.2023.110238] [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: 12/08/2022] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
Concanavalin A (ConA) is a plant lectin that can induce immune-mediated liver damage. ConA induced liver damage animal model is a widely accepted model that can mimic clinical acute hepatitis and immune-mediated liver injury in humans. Toll-like receptor-7 (TLR7), a member of the TLR family, plays a key role in pathogen recognition and innate immune activation. The aim of this study was to examine the role of TLR7 in the pathogenesis of ConA-induced liver injury. Acute liver injury was induced by intravenous injection with ConA in WT (wild-type) and TLR7 knockout (KO) mice. Results showed that attenuated liver injury in TLR7-deficient mice, as indicated by increased survival rate, decreased aminotransferase levels, and reduced pathological lesions, was associated with decreased release of pro-inflammatory cytokines in livers. Consistently, significantly decreased proliferation of CD4+ T cell was detected in ConA-stimulated TLR7-deficient splenocytes, but not in CD3/CD28 stimulated TLR7-deficient CD4+ T cells. Moreover, TLR7 deficiency in KCs specifically suppressed the expression of TNF-α (tumor necrosis factor-α). Depletion of KCs abolished the detrimental role of TLR7 in ConA-induced liver injury. Taken together, these results demonstrate that TLR7 can regulate the expression of TNF-α in KCs, which is necessary for the full progression of ConA-induced liver injury.
Collapse
Affiliation(s)
- Jing Zhao
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People's Republic of China; Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea.
| | - Hyuneui Jeong
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea.
| | - Daram Yang
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea.
| | - Weishun Tian
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, Henan, People's Republic of China.
| | - Jong-Won Kim
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea.
| | - Chae Woong Lim
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea.
| | - Bumseok Kim
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk, Republic of Korea.
| |
Collapse
|
11
|
Zhang J, Zao X, Zhang J, Guo Z, Jin Q, Chen G, Gan D, Du H, Ye Y. Is it possible to intervene early cirrhosis by targeting toll-like receptors to rebalance the intestinal microbiome? Int Immunopharmacol 2023; 115:109627. [PMID: 36577151 DOI: 10.1016/j.intimp.2022.109627] [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: 11/01/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/27/2022]
Abstract
Cirrhosis is a progressive chronic liver disease caused by one or more causes and characterized by diffuse fibrosis, pseudolobules, and regenerated nodules. Once progression to hepatic decompensation, the function of the liver and other organs is impaired and almost impossible to reverse and recover, which often results in hospitalization, impaired quality of life, and high mortality. However, in the early stage of cirrhosis, there seems to be a possibility of cirrhosis reversal. The development of cirrhosis is related to the intestinal microbiota and activation of toll-like receptors (TLRs) pathways, which could regulate cell proliferation, apoptosis, expression of the hepatomitogen epiregulin, and liver inflammation. Targeting regulation of intestinal microbiota and TLRs pathways could affect the occurrence and development of cirrhosis and its complications. In this paper, we first reviewed the dynamic change of intestinal microbiota and TLRs during cirrhosis progression. And further discussed the interaction between them and potential therapeutic targets to reverse early staged cirrhosis.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Institute of Liver Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaobin Zao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Institute of Liver Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaying Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing, China
| | - Ziwei Guo
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qian Jin
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Guang Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Institute of Liver Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Da'nan Gan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Institute of Liver Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Hongbo Du
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Institute of Liver Diseases, Beijing University of Chinese Medicine, Beijing, China
| | - Yong'an Ye
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Institute of Liver Diseases, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
12
|
MAFLD and Celiac Disease in Children. Int J Mol Sci 2023; 24:ijms24021764. [PMID: 36675276 PMCID: PMC9866925 DOI: 10.3390/ijms24021764] [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: 12/31/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/18/2023] Open
Abstract
Celiac disease (CD) is an immune-mediated systemic disorder elicited by the ingestion of gluten whose clinical presentation ranges from the asymptomatic form to clinical patterns characterized by multiple systemic involvement. Although CD is a disease more frequently diagnosed in patients with symptoms of malabsorption such as diarrhea, steatorrhea, weight loss, or failure to thrive, the raised rate of overweight and obesity among general pediatric and adult populations has increased the possibility to diagnose celiac disease in obese patients as well. Consequently, it is not difficult to also find obesity-related disorders in patients with CD, including "metabolic associated fatty liver disease" (MAFLD). The exact mechanisms linking these two conditions are not yet known. The going assumption is that a gluten-free diet (GFD) plays a pivotal role in determining an altered metabolic profile because of the elevated content of sugars, proteins, saturated fats, and complex carbohydrates, and the higher glycemic index of gluten-free products than gluten-contained foods, predisposing individuals to the development of insulin resistance. However, recent evidence supports the hypothesis that alterations in one of the components of the so-called "gut-liver axis" might contribute to the increased afflux of toxic substances to the liver triggering the liver fat accumulation and to the subsequent hepatocellular damage. The aim of this paper was to describe the actual knowledge about the factors implicated in the pathogenesis of hepatic steatosis in pediatric patients with CD. The presented review allows us to conclude that the serological evaluations for CD with anti-transglutaminase antibodies, should be a part of the general workup in the asymptomatic patients with "non-alcoholic fatty liver disease" (NAFLD) when metabolic risk factors are not evident, and in the patients with steatohepatitis when other causes of liver disease are excluded.
Collapse
|
13
|
Liu Y, Liu X, Zhou W, Zhang J, Wu J, Guo S, Jia S, Wang H, Li J, Tan Y. Integrated bioinformatics analysis reveals potential mechanisms associated with intestinal flora intervention in nonalcoholic fatty liver disease. Medicine (Baltimore) 2022; 101:e30184. [PMID: 36086766 PMCID: PMC10980383 DOI: 10.1097/md.0000000000030184] [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: 12/10/2020] [Accepted: 07/07/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease that imposes a huge economic burden on global public health. And the gut-liver axis theory supports the therapeutic role of intestinal flora in the development and progression of NAFLD. To this end, we designed bioinformatics study on the relationship between intestinal flora disorder and NAFLD, to explore the possible molecular mechanism of intestinal flora interfering with NAFLD. METHODS Differentially expressed genes for NAFLD were obtained from the GEO database. And the disease genes for NAFLD and intestinal flora disorder were obtained from the disease databases. The protein-protein interaction network was established by string 11.0 database and visualized by Cytoscape 3.7.2 software. Cytoscape plug-in MCODE and cytoHubba were used to screen the potential genes of intestinal flora disorder and NAFLD, to obtain potential targets for intestinal flora to interfere in the occurrence and process of NAFLD. Enrichment analysis of potential targets was carried out using R 4.0.2 software. RESULTS The results showed that 7 targets might be the key genes for intestinal flora to interfere with NAFLD. CCL2, IL6, IL1B, and FOS are mainly related to the occurrence and development mechanism of NAFLD, while PTGS2, SPINK1, and C5AR1 are mainly related to the intervention of intestinal flora in the occurrence and development of NAFLD. The gene function is mainly reflected in basic biological processes, including the regulation of metabolic process, epithelial development, and immune influence. The pathway is mainly related to signal transduction, immune regulation, and physiological metabolism. The TNF signaling pathway, AGE-RAGE signaling pathway in diabetic activity, and NF-Kappa B signaling pathways are important pathways for intestinal flora to interfere with NAFLD. According to the analysis results, there is a certain correlation between intestinal flora disorder and NAFLD. CONCLUSION It is speculated that the mechanism by which intestinal flora may interfere with the occurrence and development of NAFLD is mainly related to inflammatory response and insulin resistance. Nevertheless, further research is needed to explore the specific molecular mechanisms.
Collapse
Affiliation(s)
- Yingying Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xinkui Liu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Zhou
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingyuan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Siyu Guo
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shanshan Jia
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Haojia Wang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jialin Li
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Tan
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Hasa E, Hartmann P, Schnabl B. Liver cirrhosis and immune dysfunction. Int Immunol 2022; 34:455-466. [PMID: 35792761 PMCID: PMC9447994 DOI: 10.1093/intimm/dxac030] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/27/2022] [Indexed: 01/05/2023] Open
Abstract
Cirrhosis is end-stage liver disease resulting from various etiologies and is a common cause of death worldwide. The progression from compensated to decompensated cirrhosis to acute-on-chronic liver failure (ACLF) is due to multiple factors, including continuation of alcohol use or continued exposure to other toxins, an imbalance of the gut microbiota (dysbiosis), increased gut permeability and a disrupted immune response. This disrupted immune response is also named cirrhosis-associated immune dysfunction, which is characterized by worsening systemic inflammation with concomitant immune paralysis, as liver disease deteriorates. This review highlights central immunologic events during the exacerbation of cirrhosis and characterizes the different immune cell populations involved therein.
Collapse
|
15
|
Almeida JI, Tenreiro MF, Martinez-Santamaria L, Guerrero-Aspizua S, Gisbert JP, Alves PM, Serra M, Baptista PM. Hallmarks of the human intestinal microbiome on liver maturation and function. J Hepatol 2022; 76:694-725. [PMID: 34715263 DOI: 10.1016/j.jhep.2021.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/17/2021] [Indexed: 12/18/2022]
Abstract
As one of the most metabolically complex systems in the body, the liver ensures multi-organ homeostasis and ultimately sustains life. Nevertheless, during early postnatal development, the liver is highly immature and takes about 2 years to acquire and develop almost all of its functions. Different events occurring at the environmental and cellular levels are thought to mediate hepatic maturation and function postnatally. The crosstalk between the liver, the gut and its microbiome has been well appreciated in the context of liver disease, but recent evidence suggests that the latter could also be critical for hepatic function under physiological conditions. The gut-liver crosstalk is thought to be mediated by a rich repertoire of microbial metabolites that can participate in a myriad of biological processes in hepatic sinusoids, from energy metabolism to tissue regeneration. Studies on germ-free animals have revealed the gut microbiome as a critical contributor in early hepatic programming, and this influence extends throughout life, mediating liver function and body homeostasis. In this seminar, we describe the microbial molecules that have a known effect on the liver and discuss how the gut microbiome and the liver evolve throughout life. We also provide insights on current and future strategies to target the gut microbiome in the context of hepatology research.
Collapse
Affiliation(s)
- Joana I Almeida
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Miguel F Tenreiro
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Lucía Martinez-Santamaria
- Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, ISCIII), Madrid, Spain; Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| | - Sara Guerrero-Aspizua
- Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, ISCIII), Madrid, Spain
| | - Javier P Gisbert
- Gastroenterology Department. Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid (UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Paula M Alves
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Margarida Serra
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Pedro M Baptista
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Fundación ARAID, Zaragoza, Spain.
| |
Collapse
|
16
|
Fan J, Sun J, Li T, Yan X, Jiang Y. Nuciferine prevents hepatic steatosis associated with improving intestinal mucosal integrity, mucus-related microbiota and inhibiting TLR4/MyD88/NF-κB pathway in high-fat induced rats. J Funct Foods 2022. [DOI: 10.1016/j.jff.2021.104859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
17
|
Li Q, Rempel JD, Yang J, Minuk GY. The Effects of Pathogen-Associated Molecular Patterns on Peripheral Blood Monocytes in Patients with Non-alcoholic Fatty Liver Disease. J Clin Exp Hepatol 2022; 12:808-817. [PMID: 35677503 PMCID: PMC9168738 DOI: 10.1016/j.jceh.2021.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/18/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Innate immune responses to gut-derived pathogen-associated molecular patterns (PAMPs) have been implicated in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Whether NAFLD patients have increased sensitivity to PAMP exposure has yet to be reported. METHODS Peripheral blood mononuclear cell (PBMC)/monocytes were exposed to lipopolysaccharide (LPS), Pam3CSK4, or BSA conjugated palmitate in vitro. Changes in toll-like receptors (TLR), cytokines, and chemokine receptors (CR) expressions were documented by flow cytometry and/or enzyme-linked immunoabsorbent assays (ELISAs). RESULTS TLR2 and TLR4 expression were similar at baseline and increased to a similar extent (TLR2) or remained unchanged (TLR4) following PAMP exposure in NAFLD and healthy control (HC) monocytes. Proinflammatory IL-1β and IL-6 levels were similar at baseline but increased in a concentration-dependent manner to a greater extent in NAFLD PBMCs. CCR1 and CCR2 expressions at baseline were similar and decreased to a similar extent in NAFLD and HC monocytes. The extent of PAMP-induced proinflammatory cytokine release correlated with evidence of hepatocyte injury (CK18M30 levels). DISCUSSION NAFLD patients have increased proinflammatory cytokine responses following exposure to PAMPs relative to HC subjects. This response is concentration-dependent and correlates with the extent of hepatic injury.
Collapse
Key Words
- ALT, alanine aminotransferase
- CM, culture medium
- CR, chemokine receptor
- ELISAs, enzyme-linked immunoabsorbent assays
- HC, healthy controls
- LPS, lipopolysaccharide
- NAFLD
- NAFLD, nonalcoholic fatty liver disease
- NASH
- PAMPs
- PAMPs, pathogen-associated molecular patterns
- PBMC, peripheral blood mononuclear cell
- Pal, palmitate
- Pam, Pam3CSK4
- TLR, toll-like receptor
- nonalcoholic fatty liver disease
- pathogen-associated molecular patterns
Collapse
Affiliation(s)
- Qian Li
- Section of Hepatology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Julia D. Rempel
- Section of Hepatology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jiaqi Yang
- Section of Hepatology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gerald Y. Minuk
- Section of Hepatology, University of Manitoba, Winnipeg, Manitoba, Canada
- Departments of Medicine, Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
- Address for correspondence: Dr. Gerald Y. Minuk, Morberg Family Chair in Hepatology, University of Manitoba, John Buhler Research Centre, 715 McDermot Ave., Winnipeg, MB R3E 3P4, Canada. Tel.: +(204) 789 3204; fax: +(204) 789 3987.
| |
Collapse
|
18
|
Dong H, Feng Y, Yang Y, Hu Y, Jia Y, Yang S, Zhao N, Zhao R. A Novel Function of Mitochondrial Phosphoenolpyruvate Carboxykinase as a Regulator of Inflammatory Response in Kupffer Cells. Front Cell Dev Biol 2022; 9:726931. [PMID: 34970539 PMCID: PMC8712867 DOI: 10.3389/fcell.2021.726931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/30/2021] [Indexed: 01/22/2023] Open
Abstract
Background: There has been a recent appreciation that some metabolic enzymes can profoundly influence the nature of the immune response produced in macrophages. However, the role of mitochondrial phosphoenolpyruvate carboxykinase (PCK2) in immune response remains unknown. This study aims to investigate the role of PCK2 in lipopolysaccharides (LPS)-induced activation in Kupffer cells. Methods: Inflammatory cytokines were determined by real-time quantitative reverse transcription-polymerase chain action (qRT-PCR) and flow cytometric analysis using a cytometric bead array. Western blotting and immunofluorescence staining were used to determine PCK2 expression and subcellular distribution under confocal laser microscopy. qRT-PCR, flow cytometry, and high-performance liquid chromatography (HPLC) were used to determine mitochondrial function. Pharmacological inhibition, knockdown, and overexpression of PCK2 were used to confirm its function. Co-immunoprecipitation (Co-IP) was performed to determine MAPK/NF-κB phosphorylation. Results: Inflammatory response was significantly increased in LPS-treated mice and Kupffer cells. During the inflammatory process, the protein level of PCK2 was significantly upregulated in Kupffer cells. Interestingly, the localization of PCK2 was mainly in cytosol rather than mitochondria after LPS stimulation. Gain-of-function and loss-of-function analyses found that PCK2 overexpression significantly upregulated the levels of inflammation markers, whereas PCK2 knockdown or inhibition significantly mitigated LPS-induced inflammatory response in Kupffer cells. Furthermore, PCK2 promoted protein phosphorylation of NF-κB and AKT/MAPK, the major signaling pathways, controlling inflammatory cascade activation. Conclusion: We identified a novel function of PCK2 in mediating LPS-induced inflammation and provided mechanistic insights into the regulation of inflammatory response in Kupffer cells. Therefore, PCK2 may serve as a novel therapeutic target for the regulation of Kupffer cells-mediated inflammatory responses.
Collapse
Affiliation(s)
- Haibo Dong
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Yue Feng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Yang Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Yun Hu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Yimin Jia
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Shu Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Nannan Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
19
|
Portincasa P, Bonfrate L, Khalil M, Angelis MD, Calabrese FM, D’Amato M, Wang DQH, Di Ciaula A. Intestinal Barrier and Permeability in Health, Obesity and NAFLD. Biomedicines 2021; 10:83. [PMID: 35052763 PMCID: PMC8773010 DOI: 10.3390/biomedicines10010083] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
The largest surface of the human body exposed to the external environment is the gut. At this level, the intestinal barrier includes luminal microbes, the mucin layer, gastrointestinal motility and secretion, enterocytes, immune cells, gut vascular barrier, and liver barrier. A healthy intestinal barrier is characterized by the selective permeability of nutrients, metabolites, water, and bacterial products, and processes are governed by cellular, neural, immune, and hormonal factors. Disrupted gut permeability (leaky gut syndrome) can represent a predisposing or aggravating condition in obesity and the metabolically associated liver steatosis (nonalcoholic fatty liver disease, NAFLD). In what follows, we describe the morphological-functional features of the intestinal barrier, the role of major modifiers of the intestinal barrier, and discuss the recent evidence pointing to the key role of intestinal permeability in obesity/NAFLD.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Mauro D’Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE-BRTA, 48160 Derio, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| |
Collapse
|
20
|
Vohra AH, Upadhyay KK, Joshi AS, Vyas HS, Thadani J, Devkar RV. Melatonin-primed ADMSCs elicit an efficacious therapeutic response in improving high-fat diet induced non-alcoholic fatty liver disease in C57BL/6J mice. EGYPTIAN LIVER JOURNAL 2021. [DOI: 10.1186/s43066-021-00157-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Stem cells are widely used for therapy including treatment of liver damage. Adipose-derived mesenchymal stem cells (ADMSCs) administered to treat fatty liver are known to improve liver function but their use is restricted due to a poor success rate. This study investigates efficacy of melatonin-primed ADMSCs (Mel. MSCs) in experimentally induced non-alcoholic fatty liver disease (NAFLD).
Results
MSCs treated with LPS showed prominent DCFDA fluorescence as compared to the untreated cells. Also, the JC-1 staining had accounted for higher intensity of green monomer and a weak fluorescence of red dimer indicating weaker mitochondrial membrane potential. But melatonin co-treatment could make necessary corrective changes as evidenced by reverse set of results. The overall cell survival was also found to be improved following melatonin treatment as evidenced by the MTT assay. Also, the antioxidant (Nrf2 and Ho-1) and anti-inflammatory genes (Il-4 and Il-10) showed a decrement in their mRNA levels following LPS treatment whereas the pro-inflammatory genes (Tnf-α, Il-6, Tlr-4, and Lbp) showed a reciprocal increment in the said group. Melatonin co-treatment accounted for an improved status of antioxidant and anti-inflammatory genes as evidenced by their mRNA levels. High-fat high-fructose diet (HFFD) fed C57BL/6J mice recorded higher serum AST and ALT levels and fatty manifestation in histology of liver along with lowered mRNA levels of antioxidant (Nrf2, Catalase, and Gss) genes and Hgf. These set of parameters showed a significant improvement in HFFD + Mel.MSC group.
Conclusion
A significant improvement in viability of MSCs was recorded due to lowered intracellular oxidative stress and improves mitochondrial membrane potential. Further, melatonin-primed MSCs accounted for a significant decrement in fatty manifestations in liver and an improved physiological status of NAFLD in HFFD fed C57BL/6J mice. Taken together, it is hypothesized that melatonin priming to MSCs prior to its use can significantly augment the success of stem cell therapy.
Collapse
|
21
|
Mehrabi M, Amini F, Mehrabi S. Kill and Clearance in HCC: An Approach Based on NK Cells and Macrophages. Front Oncol 2021; 11:693076. [PMID: 34557407 PMCID: PMC8453146 DOI: 10.3389/fonc.2021.693076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/23/2021] [Indexed: 11/15/2022] Open
Affiliation(s)
| | | | - Shima Mehrabi
- Internal Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
The Gut Microbiota-Derived Immune Response in Chronic Liver Disease. Int J Mol Sci 2021; 22:ijms22158309. [PMID: 34361075 PMCID: PMC8347749 DOI: 10.3390/ijms22158309] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
In chronic liver disease, the causative factor is important; however, recently, the intestinal microbiome has been associated with the progression of chronic liver disease and the occurrence of side effects. The immune system is affected by the metabolites of the microbiome, and diet is the primary regulator of the microbiota composition and function in the gut–liver axis. These metabolites can be used as therapeutic material, and postbiotics, in the future, can increase or decrease human immunity by modulating inflammation and immune reactions. Therefore, the excessive intake of nutrients and the lack of nutrition have important effects on immunity and inflammation. Evidence has been published indicating that microbiome-induced chronic inflammation and the consequent immune dysregulation affect the development of chronic liver disease. In this research paper, we discuss the overall trend of microbiome-derived substances related to immunity and the future research directions.
Collapse
|
23
|
Kessoku T, Kobayashi T, Tanaka K, Yamamoto A, Takahashi K, Iwaki M, Ozaki A, Kasai Y, Nogami A, Honda Y, Ogawa Y, Kato S, Imajo K, Higurashi T, Hosono K, Yoneda M, Usuda H, Wada K, Saito S, Nakajima A. The Role of Leaky Gut in Nonalcoholic Fatty Liver Disease: A Novel Therapeutic Target. Int J Mol Sci 2021; 22:ijms22158161. [PMID: 34360923 PMCID: PMC8347478 DOI: 10.3390/ijms22158161] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023] Open
Abstract
The liver directly accepts blood from the gut and is, therefore, exposed to intestinal bacteria. Recent studies have demonstrated a relationship between gut bacteria and nonalcoholic fatty liver disease (NAFLD). Approximately 10–20% of NAFLD patients develop nonalcoholic steatohepatitis (NASH), and endotoxins produced by Gram-negative bacilli may be involved in NAFLD pathogenesis. NAFLD hyperendotoxicemia has intestinal and hepatic factors. The intestinal factors include impaired intestinal barrier function (leaky gut syndrome) and dysbiosis due to increased abundance of ethanol-producing bacteria, which can change endogenous alcohol concentrations. The hepatic factors include hyperleptinemia, which is associated with an excessive response to endotoxins, leading to intrahepatic inflammation and fibrosis. Clinically, the relationship between gut bacteria and NAFLD has been targeted in some randomized controlled trials of probiotics and other agents, but the results have been inconsistent. A recent randomized, placebo-controlled study explored the utility of lubiprostone, a treatment for constipation, in restoring intestinal barrier function and improving the outcomes of NAFLD patients, marking a new phase in the development of novel therapies targeting the intestinal barrier. This review summarizes recent data from studies in animal models and randomized clinical trials on the role of the gut–liver axis in NAFLD pathogenesis and progression.
Collapse
Affiliation(s)
- Takaomi Kessoku
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
- Department of Palliative Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
- Correspondence: ; Tel.: +81-45-787-2640; Fax: +81-45-784-3546
| | - Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Kosuke Tanaka
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
- Department of Palliative Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Atsushi Yamamoto
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Kota Takahashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
- Department of Palliative Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Anna Ozaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Yuki Kasai
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Asako Nogami
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Yasushi Honda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
- Department of Palliative Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Yuji Ogawa
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Shingo Kato
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Kento Imajo
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Takuma Higurashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Kunihiro Hosono
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Haruki Usuda
- Department of Pharmacology, Shimane University Faculty of Medicine, 89-1 Enyacho, Izumo, Shimane 693-8501, Japan; (H.U.); (K.W.)
| | - Koichiro Wada
- Department of Pharmacology, Shimane University Faculty of Medicine, 89-1 Enyacho, Izumo, Shimane 693-8501, Japan; (H.U.); (K.W.)
| | - Satoru Saito
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan; (T.K.); (K.T.); (A.Y.); (K.T.); (M.I.); (A.O.); (Y.K.); (A.N.); (Y.H.); (Y.O.); (S.K.); (K.I.); (T.H.); (K.H.); (M.Y.); (S.S.); (A.N.)
| |
Collapse
|
24
|
Da-Huang-Fu-Zi-Tang Ameliorates Severe Acute Pancreatitis by Elevation of M2 Kupffer Cells in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5561216. [PMID: 34122596 PMCID: PMC8192177 DOI: 10.1155/2021/5561216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/08/2021] [Accepted: 05/27/2021] [Indexed: 11/18/2022]
Abstract
Introduction Severe acute pancreatitis (SAP) is a clinical emergency often accompanied by inflammatory response syndrome (SIRS), which eventually leads to acute lung injury and failure of other organs. The activation of liver Kupffer cells (KCs) plays a major role in the process of SIRS and multiorgan damage caused by SAP. Da-Huang-Fu-Zi-Tang (DHFZT), a traditional Chinese prescription, has been widely used for SAP in the clinic. The present study investigated the activation state of KCs in SAP and the potential mechanism of DHFZT. Methods A total of 24 Sprague Dawley rats were randomly assigned to four groups: SH (sham operation group + saline enema), SH-DHFZT (sham operation group + DHFZT enema), SAP (model group + saline enema), and SAP-DHFZT (model group + DHFZT enema). Blood samples were drawn from the abdominal aorta for measuring serum endotoxin, amylase, calcium ion, IL-1β, TNF-α, iNOS, and IL-10. Then, the pancreas, lung, liver, and ileum were harvested for histological observation, and the liver was used to detect the level of F4/80, CD86, and CD163 in KCs with immunohistochemistry and western blot. Results In the SAP group, the CD86+ KCs were significantly increased with a high level of IL-1β, TNF-α, and iNOS, and the organs were impaired. In the SAP-DHFZT group, CD163+ KCs were significantly increased with the high level of IL-10, and the damage to organs was alleviated. Conclusion These phenomena suggested that the SIRS and multiple organ damage in SAP might be related to the excessive activation of M1 KCs, and DHFZT might alleviate the SIRS by inducing the differentiation of KCs into the M2-type and promote the expression of anti-inflammatory factor IL-10.
Collapse
|
25
|
Kwon Y, Park C, Lee J, Park DH, Jeong S, Yun CH, Park OJ, Han SH. Regulation of Bone Cell Differentiation and Activation by Microbe-Associated Molecular Patterns. Int J Mol Sci 2021; 22:ijms22115805. [PMID: 34071605 PMCID: PMC8197933 DOI: 10.3390/ijms22115805] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota has emerged as an important regulator of bone homeostasis. In particular, the modulation of innate immunity and bone homeostasis is mediated through the interaction between microbe-associated molecular patterns (MAMPs) and the host pattern recognition receptors including Toll-like receptors and nucleotide-binding oligomerization domains. Pathogenic bacteria such as Porphyromonas gingivalis and Staphylococcus aureus tend to induce bone destruction and cause various inflammatory bone diseases including periodontal diseases, osteomyelitis, and septic arthritis. On the other hand, probiotic bacteria such as Lactobacillus and Bifidobacterium species can prevent bone loss. In addition, bacterial metabolites and various secretory molecules such as short chain fatty acids and cyclic nucleotides can also affect bone homeostasis. This review focuses on the regulation of osteoclast and osteoblast by MAMPs including cell wall components and secretory microbial molecules under in vitro and in vivo conditions. MAMPs could be used as potential molecular targets for treating bone-related diseases such as osteoporosis and periodontal diseases.
Collapse
Affiliation(s)
- Yeongkag Kwon
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea; (Y.K.); (C.P.); (J.L.); (D.H.P.); (S.J.)
| | - Chaeyeon Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea; (Y.K.); (C.P.); (J.L.); (D.H.P.); (S.J.)
| | - Jueun Lee
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea; (Y.K.); (C.P.); (J.L.); (D.H.P.); (S.J.)
| | - Dong Hyun Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea; (Y.K.); (C.P.); (J.L.); (D.H.P.); (S.J.)
| | - Sungho Jeong
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea; (Y.K.); (C.P.); (J.L.); (D.H.P.); (S.J.)
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea;
| | - Ok-Jin Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea; (Y.K.); (C.P.); (J.L.); (D.H.P.); (S.J.)
- Correspondence: (O.-J.P.); (S.H.H.); Tel.: +82-2-880-2312 (O.-J.P.); +82-2-880-2310 (S.H.H.)
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea; (Y.K.); (C.P.); (J.L.); (D.H.P.); (S.J.)
- Correspondence: (O.-J.P.); (S.H.H.); Tel.: +82-2-880-2312 (O.-J.P.); +82-2-880-2310 (S.H.H.)
| |
Collapse
|
26
|
Mega A, Marzi L, Kob M, Piccin A, Floreani A. Food and Nutrition in the Pathogenesis of Liver Damage. Nutrients 2021; 13:nu13041326. [PMID: 33923822 PMCID: PMC8073814 DOI: 10.3390/nu13041326] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 11/19/2022] Open
Abstract
The liver is an important organ and plays a key role in the regulation of metabolism and in the secretion, storage, and detoxification of endogenous and exogenous substances. The impact of food and nutrition on the pathophysiological mechanisms of liver injury represents a great controversy. Several environmental factors including food and micronutrients are involved in the pathogenesis of liver damage. Conversely, some xenobiotics and micronutrients have been recognized to have a protective effect in several liver diseases. This paper offers an overview of the current knowledge on the role of xenobiotics and micronutrients in liver damage.
Collapse
Affiliation(s)
- Andrea Mega
- Gastroenterology Department, Bolzano Regional Hospital, 39100 Bolzano, Italy;
- Correspondence:
| | - Luca Marzi
- Gastroenterology Department, Bolzano Regional Hospital, 39100 Bolzano, Italy;
| | - Michael Kob
- Dietetics and Clinical Nutrition Unit, Bolzano Regional Hospital, 39100 Bolzano, Italy;
| | - Andrea Piccin
- Northern Ireland Blood Transfusion Service, Belfast BT9 7TS, UK;
- Department of Internal Medicine V, Medical University of Innsbruck, A-6020 Innsbruck, Austria
- Department of Industrial Engineering, University of Trento, 38100 Trento, Italy
| | - Annarosa Floreani
- Scientific Institute for Research, Hospitalization and Healthcare, 37024 Negrar-Verona, Italy;
- Department Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy
| |
Collapse
|
27
|
Du HJ, Zhao SX, Zhao W, Fu N, Li WC, Qin XJ, Zhang YG, Nan YM, Zhao JM. Hepatic Macrophage activation and the LPS pathway in patients with different degrees of severity and histopathological patterns of drug induced liver injury. Histol Histopathol 2021; 36:653-662. [PMID: 33870482 DOI: 10.14670/hh-18-340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Inflammatory activation of hepatic macrophages plays a primary role in drug-induced liver injury (DILI). However, the exact mechanism underlying DILI remains unclear. METHODS A total of 328 DILI patients and 80 healthy individuals were prospectively enrolled in this study. The DILI patients were categorized into subgroups based on either disease severity or histopathological patterns. Plasma soluble CD163 (sCD163) and hepatic CD163 were examined to determine hepatic macrophage activation, and CD8, CD20, and MUM-1 were assessed to determine cellular immunity using immunohistochemistry. The lipopolysaccharide (LPS) pathway proteins [e.g. LPS, soluble CD14 (sCD14), and LPS-binding protein (LBP)] were measured using enzyme-linked immunosorbent assay. RESULTS Plasma sCD163 levels were nine-fold higher in DILI patients than in healthy controls at the baseline, but significantly decreased at the 4-week follow-up visit after treatment. The numbers of hepatic macrophages, B cells, and plasma cells were significantly higher in the liver tissues from DILI patients than those from healthy controls. Furthermore, the baseline levels of LPS pathway proteins in the DILI patients were significantly higher than those in the controls. Notably, these proteins significantly decreased at the 4-week follow-up visit but remained significantly higher than the levels for the controls. CONCLUSIONS Hepatic inflammation in DILI involves the activation of hepatic macrophages and cellular immunity, in which the LPS pathway likely plays a role, at least in part. As such, this study has improved our understanding of the pathological mechanisms for DILI and may facilitate the development of better treatments for patients with DILI.
Collapse
Affiliation(s)
- Hui-Juan Du
- Department of Traditional and Western Medical Hepatology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Su-Xian Zhao
- Department of Traditional and Western Medical Hepatology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wen Zhao
- Department of Traditional and Western Medical Hepatology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Na Fu
- Department of Traditional and Western Medical Hepatology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wen-Cong Li
- Department of Traditional and Western Medical Hepatology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiao-Jie Qin
- Department of Traditional and Western Medical Hepatology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yu-Guo Zhang
- Department of Traditional and Western Medical Hepatology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yue-Min Nan
- Department of Traditional and Western Medical Hepatology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Jing-Min Zhao
- Department of Pathology and Hepatology Institution, The Fifth Medical Center, General Hospital of PLA, Beijing, China
| |
Collapse
|
28
|
Fan Y, Li Y, Chu Y, Liu J, Cui L, Zhang D. Toll-Like Receptors Recognize Intestinal Microbes in Liver Cirrhosis. Front Immunol 2021; 12:608498. [PMID: 33708204 PMCID: PMC7940369 DOI: 10.3389/fimmu.2021.608498] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Liver cirrhosis is one major cause of mortality in the clinic, and treatment of this disease is an arduous task. The scenario will be even getting worse with increasing alcohol consumption and obesity in the current lifestyle. To date, we have no medicines to cure cirrhosis. Although many etiologies are associated with cirrhosis, abnormal intestinal microbe flora (termed dysbiosis) is a common feature in cirrhosis regardless of the causes. Toll-like receptors (TLRs), one evolutional conserved family of pattern recognition receptors in the innate immune systems, play a central role in maintaining the homeostasis of intestinal microbiota and inducing immune responses by recognizing both commensal and pathogenic microbes. Remarkably, recent studies found that correction of intestinal flora imbalance could change the progress of liver cirrhosis. Therefore, correction of intestinal dysbiosis and targeting TLRs can provide novel and promising strategies in the treatment of liver cirrhosis. Here we summarize the recent advances in the related topics. Investigating the relationship among innate immunity TLRs, intestinal flora disorders, and liver cirrhosis and exploring the underlying regulatory mechanisms will assuredly have a bright future for both basic and clinical research.
Collapse
Affiliation(s)
- Yujing Fan
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunpeng Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanjie Chu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Liu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lin Cui
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dekai Zhang
- Center for Infectious and Inflammatory Diseases, Texas A&M University, Houston, TX, United States
| |
Collapse
|
29
|
Gül N, Grewal S, Bögels M, van der Bij GJ, Koppes MMA, Oosterling SJ, Fluitsma DM, Hoeben KA, Beelen RHJ, van Egmond M. Macrophages mediate colon carcinoma cell adhesion in the rat liver after exposure to lipopolysaccharide. Oncoimmunology 2021; 1:1517-1526. [PMID: 23264898 PMCID: PMC3525607 DOI: 10.4161/onci.22303] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The surgical resection of primary colorectal cancer is associated with an enhanced risk of liver metastases. Moreover, bacterial translocation or anastomic leakage during resection has been shown to correlate with a poor long-term surgical outcome, suggesting that bacterial products may contribute to the formation of metastases. Driven by these premises, we investigated the role of the bacterial product lipopolysaccharide (LPS) in the generation of liver metastases. Intraperitoneal injection of LPS led to enhanced tumor-cell adhesion to the rat liver as early as 1.5 h post-administration. Furthermore, a rapid loss of the expression of the tight junction protein zonula occludens-1 (ZO-1) was observed, suggesting that LPS disrupts the integrity of the microvasculature. LPS addition to endothelial-macrophage co-cultures damaged endothelial monolayers and caused the formation of intercellular gaps, which was accompanied by increased tumor-cell adhesion. These results suggest that macrophages are involved in the endothelial damage resulting from exposure to LPS. Interestingly, the expression levels of of ZO-1 were not affected by LPS treatment in rats in which liver macrophages had been depleted as well as in rats that had been treated with a reactive oxygen species (ROS) scavenger. In both settings, decreased tumor-cell adhesion was observed. Taken together, our findings indicate that LPS induces ROS release by macrophages, resulting in the damage of the vascular lining of the liver and hence allowing increased tumor-cell adherence. Thus, peri-operative treatments that prevent the activation of macrophages and—as a consequence—limit endothelial damage and tumor-cell adhesion may significantly improve the long-term outcome of cancer patients undergoing surgical tumor resection.
Collapse
Affiliation(s)
- Nuray Gül
- Department of Molecular Cell Biology and Immunology; VU University Medical Center; Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Tian X, Zhao H, Guo Z. Effects of Carvedilol on the Expression of TLR4 and its Downstream Signaling Pathway in the Liver Tissues of Rats with Cholestatic Liver Fibrosis. Curr Mol Med 2021; 20:708-716. [PMID: 32077825 DOI: 10.2174/1566524020666200220130705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 11/22/2022]
Abstract
Objectives:
This study was designed to investigate the effects of carvedilol
on the expression of TLR4 and its downstream signaling pathway in the liver tissues of
rats with cholestatic liver fibrosis and provide experimental evidence for clinical
treatment of liver fibrosis with carvedilol.
Methods:
A total of fifty male Sprague Dawley rats were randomly divided into five
groups (10 rats per group): sham operation (SHAM) control group, bile duct ligation
(BDL) model group, low-dose carvedilol treatment group (0.1mg·kg-1·d-1), medium-dose
carvedilol treatment group (1mg·kg-1·d-1), and high-dose carvedilol treatment group
(10mg·kg-1·d-1). Rat hepatic fibrosis model was established by applying BDL. Forty-eight
hours after the operation, carvedilol was administered twice a day. The blood and liver
were simultaneously collected under the aseptic condition for further detection in two
weeks after the operation. The alanine aminotransferase (ALT), aspartate
aminotransferase (AST), total bilirubin (TBil) and albumin (Alb) in serum were measured.
HE and Masson staining were used to determine hepatic fibrosis degree. Hydroxyproline
assay was employed to detect liver collagen synthesis. Western Blot was used to
measure the expression of TLR4, NF-κB p65 and β-arrestin2 protein. Quantitative
analysis of TLR4, MyD88, TNF-α and IL-6 mRNA was performed by Realtime-PCR.
Results:
Compared with the SHAM group, the BDL group showed obvious liver injury,
increased levels of inflammatory factors, and continued progression of liver fibrosis. The
above changes in the BDL group were alleviated in the carvedilol treatment groups. The
improvement effects augmented as dosages increased. In addition, compared with the
BDL group, the reduction of the expressions of TLR4, MyD88 and NF-κB p65 in liver
tissues and the increase of the expression of β -arrestin2 in the high-dose carvedilol
group were more significant.
Conclusions:
Carvedilol can reduce the release of inflammatory mediators by downregulating
TLR4 expression and inhibiting its downstream signaling pathway, thus
playing a potential therapeutic role in cholestatic liver fibrosis.
Collapse
Affiliation(s)
- Xiaopeng Tian
- Department of Gastroenterology, Xingtai People’s Hospital, Xingtai, Hebei 054000, China
| | - Huimin Zhao
- Department of Gastroenterology, Xingtai People’s Hospital, Xingtai, Hebei 054000, China
| | - Zengcai Guo
- Department of Gastroenterology, Xingtai People’s Hospital, Xingtai, Hebei 054000, China
| |
Collapse
|
31
|
Shao Q, Wu Y, Ji J, Xu T, Yu Q, Ma C, Liao X, Cheng F, Wang X. Interaction Mechanisms Between Major Depressive Disorder and Non-alcoholic Fatty Liver Disease. Front Psychiatry 2021; 12:711835. [PMID: 34966296 PMCID: PMC8710489 DOI: 10.3389/fpsyt.2021.711835] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Major depressive disorder (MDD), which is highly associated with non-alcoholic fatty liver disease (NAFLD), has complex pathogenic mechanisms. However, a limited number of studies have evaluated the mutual pathomechanisms involved in MDD and NAFLD development. Chronic stress-mediated elevations in glucocorticoid (GC) levels play an important role in the development of MDD-related NAFLD. Elevated GC levels can induce the release of inflammatory factors and changes in gut permeability. Elevated levels of inflammatory factors activate the hypothalamic-pituitary-adrenal (HPA) axis, which further increases the release of GC. At the same time, changes in gut permeability promote the release of inflammatory factors, which results in a vicious circle among the three, causing disease outbreaks. Even though the specific role of the thyroid hormone (TH) in this pathogenesis has not been fully established, it is highly correlated with MDD and NAFLD. Therefore, changing lifestyles and reducing psychological stress levels are necessary measures for preventing MDD-related NAFLD. Among them, GC inhibitors and receptor antagonists may be key in the alleviation of early and mid-term disease progression. However, combination medications may be important in late-stage diseases, but they are associated with various side effects. Traditional Chinese medicines have been shown to be potential therapeutic alternatives for such complex diseases.
Collapse
Affiliation(s)
- Qi Shao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yiping Wu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Ji
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tian Xu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qiaoyu Yu
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chongyang Ma
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xuejing Liao
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fafeng Cheng
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xueqian Wang
- College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Tian X, Zhao H, Guo Z. WITHDRAWN: Effects of carvedilol on expression of TLR4 and its downstream signaling pathway in liver tissue of rats with cholestatic liver fibrosisjaundice. REVISTA ESPANOLA DE ENFERMEDADES DIGESTIVAS 2020. [PMID: 33200614 DOI: 10.17235/reed.2020.6075/2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Ahead of Print article withdrawn by publisher. OBJECTIVES This study was designed to investigate the effects of carvedilol on the expression of TLR4 and its downstream signaling pathway in liver tissue of rats with cholestatic liver fibrosis, and provided experimental evidence for clinical treatment of liver fibrosis with carvedilol.? METHODS A total of fifty male Sprague Dawley rats were randomly divided into five groups (10 rats per group): sham surgery control group, bile duct ligation (BDL) model group, low-dose carvedilol treatment group (0.1mgkg-1d-1), medium-dose carvedilol treatment group (1mgkg-1d-1), high-dose carvedilol treatment group (10mgkg-1d-1). Rat hepatic fibrosis model was established by applying BDL. Forty-eight hours after the operation, carvedilol was administered twice a day. The blood and liver were simultaneously collected under the aseptic condition for further detection in two weeks after operation.? RESULTS Compared with the sham group, the BDL group showed obvious liver injury, increased levels of inflammatory factors, and continued progression of liver fibrosis. Carvedilol could alleviate the above changes. The improvement effects were augmenting as dosages increasing. In addition, compared with the BDL group, carvedilol can reduce the expressions of TLR4, MyD88 and NF-?B p65 in liver tissue and increase the expression of ?-arrestin2, and the effect in the high dose group was more obvious. CONCLUSIONS Carvedilol can reduce the release of inflammatory mediators by down-regulating TLR4 expression and inhibiting its downstream signaling pathway, thus playing a therapeutic role in cholestatic liver fibrosis.
Collapse
|
33
|
Cacicedo ML, Medina-Montano C, Kaps L, Kappel C, Gehring S, Bros M. Role of Liver-Mediated Tolerance in Nanoparticle-Based Tumor Therapy. Cells 2020; 9:E1985. [PMID: 32872352 PMCID: PMC7563539 DOI: 10.3390/cells9091985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/26/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
In the last decades, the use of nanocarriers for immunotherapeutic purposes has gained a lot of attention, especially in the field of tumor therapy. However, most types of nanocarriers accumulate strongly in the liver after systemic application. Due to the default tolerance-promoting role of liver non-parenchymal cells (NPCs), Kupffer cells (KCs), liver sinusoidal endothelial cells (LSECs), and hepatic stellate cells (HSCs), their potential role on the immunological outcome of systemic nano-vaccination approaches for therapy of tumors in the liver and in other organs needs to be considered. Concerning immunological functions, KCs have been the focus until now, but recent studies have elucidated an important role of LSECs and HSCs as well. Therefore, this review aims to summarize current knowledge on the employment of nanocarriers for immunotherapeutic therapy of liver diseases and the overall role of liver NPCs in the context of nano-vaccination approaches. With regard to the latter, we discuss strategies on how to address liver NPCs, aiming to exploit and modulate their immunological properties, and alternatively how to avoid unwanted engagement of nano-vaccines by liver NPCs for tumor therapy.
Collapse
Affiliation(s)
- Maximiliano L. Cacicedo
- Children’s Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.L.C.); (S.G.)
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (C.M.-M.); (C.K.)
| | - Leonard Kaps
- Department of Medicine, University Medical Center Mainz, I. Langenbeckstrasse 1, 55131 Mainz, Germany;
| | - Cinja Kappel
- Department of Dermatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (C.M.-M.); (C.K.)
| | - Stephan Gehring
- Children’s Hospital, University Medical Center, Langenbeckstrasse 1, 55131 Mainz, Germany; (M.L.C.); (S.G.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (C.M.-M.); (C.K.)
| |
Collapse
|
34
|
Di Ciaula A, Baj J, Garruti G, Celano G, De Angelis M, Wang HH, Di Palo DM, Bonfrate L, Wang DQH, Portincasa P. Liver Steatosis, Gut-Liver Axis, Microbiome and Environmental Factors. A Never-Ending Bidirectional Cross-Talk. J Clin Med 2020; 9:2648. [PMID: 32823983 PMCID: PMC7465294 DOI: 10.3390/jcm9082648] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/07/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023] Open
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing worldwide and parallels comorbidities such as obesity, metabolic syndrome, dyslipidemia, and diabetes. Recent studies describe the presence of NAFLD in non-obese individuals, with mechanisms partially independent from excessive caloric intake. Increasing evidences, in particular, point towards a close interaction between dietary and environmental factors (including food contaminants), gut, blood flow, and liver metabolism, with pathways involving intestinal permeability, the composition of gut microbiota, bacterial products, immunity, local, and systemic inflammation. These factors play a critical role in the maintenance of intestinal, liver, and metabolic homeostasis. An anomalous or imbalanced gut microbial composition may favor an increased intestinal permeability, predisposing to portal translocation of microorganisms, microbial products, and cell wall components. These components form microbial-associated molecular patterns (MAMPs) or pathogen-associated molecular patterns (PAMPs), with potentials to interact in the intestine lamina propria enriched in immune cells, and in the liver at the level of the immune cells, i.e., Kupffer cells and stellate cells. The resulting inflammatory environment ultimately leads to liver fibrosis with potentials to progression towards necrotic and fibrotic changes, cirrhosis. and hepatocellular carcinoma. By contrast, measures able to modulate the composition of gut microbiota and to preserve gut vascular barrier might prevent or reverse NAFLD.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| | - Jacek Baj
- Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Gabriella Garruti
- Section of Endocrinology, Department of Emergency and Organ Transplantations, University of Bari “Aldo Moro” Medical School, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Giuseppe Celano
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Maria De Angelis
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Helen H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (H.H.W.); (D.Q.-H.W.)
| | - Domenica Maria Di Palo
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
- Dipartimento di Scienze del Suolo, della Pianta e Degli Alimenti, Università degli Studi di Bari Aldo Moro, 70124 Bari, Italy; (G.C.); (M.D.A.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| | - David Q-H Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (H.H.W.); (D.Q.-H.W.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (A.D.C.); (D.M.D.P.); (L.B.)
| |
Collapse
|
35
|
Yang X, Lu D, Zhuo J, Lin Z, Yang M, Xu X. The Gut-liver Axis in Immune Remodeling: New insight into Liver Diseases. Int J Biol Sci 2020; 16:2357-2366. [PMID: 32760203 PMCID: PMC7378637 DOI: 10.7150/ijbs.46405] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota consists of a dynamic multispecies community of bacteria, fungi, archaea, and protozoans, playing a fundamental role in the induction, training, and function of the host immune system. The liver is anatomically and physiologically linked to the gut microbiota via enterohepatic circulation, specifically receiving intestine-derived blood through the portal vein. The gut microbiota is crucial for maintaining immune homeostasis of the gut-liver axis. A shift in gut microbiota composition can result in activation of the mucosal immune response causing homeostasis imbalance. This imbalance results in translocation of bacteria and migration of immune cells to the liver, which is related to inflammation-mediated liver injury and tumor progression. In this review, we outline the role of the gut microbiota in modulating host immunity and summarize novel findings and recent advances in immune-based therapeutics associated with the gut-liver axis. Moving forward, a deep understanding of the microbiome-immune-liver axis will provide insight into the basic mechanisms of gut microbiota dysbiosis affecting liver diseases.
Collapse
Affiliation(s)
- Xinyu Yang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Di Lu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jianyong Zhuo
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Zuyuan Lin
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Modan Yang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, China.,NHFPC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| |
Collapse
|
36
|
Li C, Chen Y, Yuan X, He L, Li X, Huang S, Hou S, Liang J. Vitexin ameliorates chronic stress plub high fat diet-induced nonalcoholic fatty liver disease by inhibiting inflammation. Eur J Pharmacol 2020; 882:173264. [PMID: 32544504 DOI: 10.1016/j.ejphar.2020.173264] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022]
Abstract
Evidences showed that chronic stress (CS) can aggravate the situation of nonalcoholic fatty liver disease (NAFLD). Vitexin is one of the major components in hawthorn, which is widely used to reduce blood lipid. This study was aimed to explore the therapeutic effects and potential mechanisms of vitexin on chronic stress mice with high-fat diet (CSHFD). The results showed that 5-week vitexin administration (40 mg/kg, i.g.) could obviously reduce hepatic fat deposition, alleviate lipid metabolism, and inhibit liver inflammation in CSHFD mice. In addition, vitexin significantly reduced hepatic macrophage infiltration, obviously down-regulated the mRNA and protein expressions of hepatic SREBP-1c, FAS, ACC. Moreover, we also found that vitexin treatment could significantly inhibit the expressions of TLR4/NF-κB signaling in CSHFD mice. This results suggested that vitexin could ameliorate chronic stress combined with high-fat diet induced NAFLD, and its mechanisms is closely related to inhibit TLR4/NF-κB signaling and reduce fatty acid synthesis proteins.
Collapse
Affiliation(s)
- Chujie Li
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangzhou, 510006, PR China
| | - Yonger Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Xin Yuan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangzhou, 510006, PR China
| | - Lian He
- Guangdong Food and Drug Vocational College, Guangzhou, 510520, Guangdong, PR China
| | - Xiaojun Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Song Huang
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangzhou, 510006, PR China
| | - Shaozhen Hou
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangzhou, 510006, PR China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China.
| | - Jian Liang
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangzhou, 510006, PR China.
| |
Collapse
|
37
|
|
38
|
Giuffrè M, Campigotto M, Campisciano G, Comar M, Crocè LS. A story of liver and gut microbes: how does the intestinal flora affect liver disease? A review of the literature. Am J Physiol Gastrointest Liver Physiol 2020; 318:G889-G906. [PMID: 32146836 DOI: 10.1152/ajpgi.00161.2019] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Each individual is endowed with a unique gut microbiota (GM) footprint that mediates numerous host-related physiological functions, such as nutrient metabolism, maintenance of the structural integrity of the gut mucosal barrier, immunomodulation, and protection against microbial pathogens. Because of increased scientific interest in the GM, its central role in the pathophysiology of many intestinal and extraintestinal conditions has been recognized. Given the close relationship between the gastrointestinal tract and the liver, many pathological processes have been investigated in the light of a microbial-centered hypothesis of hepatic damage. In this review we introduce to neophytes the vast world of gut microbes, including prevalent bacterial distribution in healthy individuals, how the microbiota is commonly analyzed, and the current knowledge of the role of GM in liver disease pathophysiology. Also, we highlight the potentials and downsides of GM-based therapy.
Collapse
Affiliation(s)
- Mauro Giuffrè
- Dipartimento Universitario Clinico di Scienze Mediche Chirurgiche e della Salute, Università degli Studi di Trieste, Italy
| | - Michele Campigotto
- Dipartimento Universitario Clinico di Scienze Mediche Chirurgiche e della Salute, Università degli Studi di Trieste, Italy
| | - Giuseppina Campisciano
- Istituto di Ricovero e Cura a Carattere Scientifico Materno Infantile Burlo Garofolo, Trieste, Italy
| | - Manola Comar
- Dipartimento Universitario Clinico di Scienze Mediche Chirurgiche e della Salute, Università degli Studi di Trieste, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico Materno Infantile Burlo Garofolo, Trieste, Italy
| | - Lory Saveria Crocè
- Dipartimento Universitario Clinico di Scienze Mediche Chirurgiche e della Salute, Università degli Studi di Trieste, Italy.,Clinica Patologie del Fegato, Azienda Sanitaria Universitaria Integrata di Trieste, Italy.,Fondazione Italiana Fegato, Trieste, Italy
| |
Collapse
|
39
|
BMP-9 Modulates the Hepatic Responses to LPS. Cells 2020; 9:cells9030617. [PMID: 32143367 PMCID: PMC7140468 DOI: 10.3390/cells9030617] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/25/2020] [Accepted: 03/02/2020] [Indexed: 12/11/2022] Open
Abstract
It was previously shown that Bone Morphogenetic Protein (BMP)-9 is constitutively produced and secreted by hepatic stellate cells (HSC). Upon acute liver damage, BMP-9 expression is transiently down-regulated and blocking BMP-9 under conditions of chronic damage ameliorated liver fibrogenesis in C57BL/6 mice. Thereby, BMP-9 acted as a pro-fibrogenic cytokine in the liver but without directly activating isolated HSC in vitro. Lipopolysaccharide (LPS), an endotoxin derived from the membrane of Gram-negative bacteria in the gut, is known to be essential in the pathogenesis of diverse kinds of liver diseases. The aim of the present project was therefore to investigate how high levels of BMP-9 in the context of LPS signalling might result in enhanced liver damage. For this purpose, we stimulated human liver sinusoidal endothelial cells (LSEC) with LPS and incubated primary human liver myofibroblasts (MF) with the conditioned medium of these cells. We found that LPS led to the secretion of factors from LSEC that upregulate BMP-9 expression in MF. At least one of these BMP-9 enhancing factors was defined to be IL-6. High BMP-9 in turn, especially in combination with LPS stimulation, induced the expression of certain capillarization markers in LSEC and enhanced the LPS-mediated induction of pro-inflammatory cytokines in primary human macrophages. In LSEC, pre-treatment with BMP-9 reduced the LPS-mediated activation of the NfkB pathway, whereas in macrophages, LPS partially inhibited the BMP-9/Smad-1 signaling cascade. In vivo, in mice, BMP-9 led to the enhanced presence of F4/80-positive cells in the liver and it modulated the LPS-mediated regulation of inflammatory mediators. In summary, our data point to BMP-9 being a complex and highly dynamic modulator of hepatic responses to LPS: Initial effects of LPS on LSEC led to the upregulation of BMP-9 in MF but sustained high levels of BMP-9 in turn promote pro-inflammatory reactions of macrophages. Thereby, the spatial and timely fine-tuned presence (or absence) of BMP-9 is needed for efficient wound-healing responses in the liver.
Collapse
|
40
|
Lokugamage MP, Gan Z, Zurla C, Levin J, Islam FZ, Kalathoor S, Sato M, Sago CD, Santangelo PJ, Dahlman JE. Mild Innate Immune Activation Overrides Efficient Nanoparticle-Mediated RNA Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1904905. [PMID: 31743531 PMCID: PMC7029413 DOI: 10.1002/adma.201904905] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/18/2019] [Indexed: 04/14/2023]
Abstract
Clinical mRNA delivery remains challenging, in large part because how physiology alters delivery in vivo remains underexplored. For example, mRNA delivered by lipid nanoparticles (LNPs) is being considered to treat inflammation, but whether inflammation itself changes delivery remains understudied. Relationships between immunity, endocytosis, and mRNA translation lead to hypothesize that toll-like receptor 4 (TLR4) activation reduced LNP-mediated mRNA delivery. Therefore, LNP uptake, endosomal escape, and mRNA translation with and without TLR4 activation are quantified. In vivo DNA barcoding is used to discover a novel LNP that delivers mRNA to Kupffer cells at clinical doses; unlike most LNPs, this LNP does not preferentially target hepatocytes. TLR4 activation blocks mRNA translation in all tested cell types, without reducing LNP uptake; inhibiting TLR4 or its downstream effector protein kinase R improved delivery. The discrepant effects of TLR4 on i) LNP uptake and ii) translation suggests TLR4 activation can "override" LNP targeting, even after mRNA is delivered into target cells. Given near-future clinical trials using mRNA to modulate inflammation, this highlights the need to understand inflammatory signaling in on- and off-target cells. More generally, this suggests an LNP which delivers mRNA to one inflammatory disease may not deliver mRNA to another.
Collapse
Affiliation(s)
- Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| | - Zubao Gan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| | - Chiara Zurla
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| | - Joel Levin
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| | - Fatima Z Islam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| | - Sujay Kalathoor
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| | - Manaka Sato
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| | - Cory D Sago
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, 30332, USA
| |
Collapse
|
41
|
Ito T, Nakamura K, Kageyama S, Korayem IM, Hirao H, Kadono K, Aziz J, Younan S, DiNorcia J, Agopian VG, Yersiz H, Farmer DG, Busuttil RW, Kupiec-Weglinski JW, Kaldas FM. Impact of Rifaximin Therapy on Ischemia/Reperfusion Injury in Liver Transplantation: A Propensity Score-Matched Analysis. Liver Transpl 2019; 25:1778-1789. [PMID: 31509643 PMCID: PMC6887108 DOI: 10.1002/lt.25633] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/13/2019] [Indexed: 12/22/2022]
Abstract
Intestinal microbiota is thought to play an important role in hepatic ischemia/reperfusion injury (IRI) after liver transplantation (LT). Rifaximin, a nonabsorbable antibiotic used to treat encephalopathy, exhibits antibacterial activity within the gut. We report the first study examining the impact of pre-LT rifaximin use on reducing hepatic IRI and inflammatory cell infiltration after LT. This retrospective single-center study included adult LT recipients from January 2013 through June 2016. Patients were divided into 2 groups based on duration of rifaximin use before LT: rifaximin group (≥28 days) and control group (none or <28 days). Patients receiving other antibiotics within 28 days of LT and re-LTs were excluded. Outcomes and messenger RNA (mRNA) expression in the graft were compared by 1:1 propensity score-matching and multivariate analyses. On 1:1 matching (n = 39/group), rifaximin patients had lower postoperative serum transaminase levels and lower early allograft dysfunction (EAD; 10.3% versus 33.3%; P = 0.014). Of the matched patients, 8 patients (n = 4/group) had postreperfusion liver biopsies (approximately 2 hours after reperfusion) available for mRNA analysis. Hepatic expression of CD86 (macrophage marker) and cathepsin G (neutrophil marker) was significantly lower in rifaximin patients than controls (P < 0.05). The multivariate analysis included 458 patients. Rifaximin treatment <28 days was identified as an independent risk factor EAD in all patients and those with high Model for End-Stage Liver Disease (MELD) score (MELD ≥35; n = 230). In conclusion, the propensity score-matched and multivariate analyses suggest a therapeutic role of rifaximin in reducing EAD. Pre-LT rifaximin administration exerted a protective function against early liver injury, potentially by suppressing inflammatory cell activation in the graft.
Collapse
Affiliation(s)
- Takahiro Ito
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Kojiro Nakamura
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA,Department of Surgery, Kyoto University, Kyoto,
Japan
| | - Shoichi Kageyama
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Islam M. Korayem
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA,Hepato-Pancreato-Biliary Surgery Unit, Department of
Surgery, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Hirofumi Hirao
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Kentaro Kadono
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Justine Aziz
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Stephanie Younan
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Joseph DiNorcia
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Vatche G. Agopian
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Hasan Yersiz
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Douglas G. Farmer
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Ronald W. Busuttil
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Jerzy W. Kupiec-Weglinski
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| | - Fady M. Kaldas
- The Dumont-UCLA Transplantation Center, Division of Liver
and Pancreas Transplantation, Department of Surgery, David Geffen School of
Medicine, University of California, Los Angeles, CA
| |
Collapse
|
42
|
Challan SB, Marzook FA, Massoud A. Synthesis of radioiodinated carnosine for hepatotoxicity imaging induced by carbon tetrachloride and its biological assessment in rats. RADIOCHIM ACTA 2019. [DOI: 10.1515/ract-2019-3162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Abstract
The imaging of organs is very important in the field of diagnosis especially in case of liver diseases. In the present work, carnosine was successfully labeled with iodine-131 at room temperature in acidic medium using chloramine-T (Ch-T) as moderate oxidizing agent. The parameters affecting labeling of carnosine such as amount of oxidizing agent, amount of substrate, pH value of the reaction mixture, reaction temperature and reaction time, were investigated. The best conditions for formation of 131I-carnosine (131I-CAR) complex were 40 μg of chloramine-T (Ch-T), 75 μg of carnosine, pH 4 and 45 min reaction time at room temperature. The radiochemical yield for 131I-CAR complex was (91 ± 0.11) % at optimum conditions and the labeled complex was stable for 2 h after labeling process. Biodistribution study was achieved using three groups of rats (normal, treated by inactive carnosine and hepatotoxicity rats induced by CCl4). Hepatotoxicity of liver was evaluated using different biochemical markers such as ALT, AST and ALK.P. The 131I-CAR complex showed selective bio-localization in stomach and liver and its selectivity increases in acquired hepatotoxicity. The biological distribution indicates that the suitability of 131I-CAR as a potential hepatotoxicity imaging to detect hepatitis and medical prognosis.
Collapse
Affiliation(s)
- Safaa B. Challan
- Chemistry Unite of Cyclotron, Nuclear Research Center, Egyptian Atomic Energy Authority , P.O. Code 13759 , Cairo , Egypt
| | - Fawzy A. Marzook
- Department of Labeled Compounds , Hot Labs Center, Egyptian Atomic Energy Authority , P.O. Code 13759 , Cairo , Egypt
| | - Ayman Massoud
- Chemistry Unite of Cyclotron, Nuclear Research Center, Nuclear Chemistry Department, Egyptian Atomic Energy Authority , P.O. Code 13759 , Cairo , Egypt , E-mail:
| |
Collapse
|
43
|
Nicoletti A, Ponziani FR, Biolato M, Valenza V, Marrone G, Sganga G, Gasbarrini A, Miele L, Grieco A. Intestinal permeability in the pathogenesis of liver damage: From non-alcoholic fatty liver disease to liver transplantation. World J Gastroenterol 2019; 25:4814-4834. [PMID: 31543676 PMCID: PMC6737313 DOI: 10.3748/wjg.v25.i33.4814] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/04/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
The intimate connection and the strict mutual cooperation between the gut and the liver realizes a functional entity called gut-liver axis. The integrity of intestinal barrier is crucial for the maintenance of liver homeostasis. In this mutual relationship, the liver acts as a second firewall towards potentially harmful substances translocated from the gut, and is, in turn, is implicated in the regulation of the barrier. Increasing evidence has highlighted the relevance of increased intestinal permeability and consequent bacterial translocation in the development of liver damage. In particular, in patients with non-alcoholic fatty liver disease recent hypotheses are considering intestinal permeability impairment, diet and gut dysbiosis as the primary pathogenic trigger. In advanced liver disease, intestinal permeability is enhanced by portal hypertension. The clinical consequence is an increased bacterial translocation that further worsens liver damage. Furthermore, this pathogenic mechanism is implicated in most of liver cirrhosis complications, such as spontaneous bacterial peritonitis, hepatorenal syndrome, portal vein thrombosis, hepatic encephalopathy, and hepatocellular carcinoma. After liver transplantation, the decrease in portal pressure should determine beneficial effects on the gut-liver axis, although are incompletely understood data on the modifications of the intestinal permeability and gut microbiota composition are still lacking. How the modulation of the intestinal permeability could prevent the initiation and progression of liver disease is still an uncovered area, which deserves further attention.
Collapse
Affiliation(s)
- Alberto Nicoletti
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Francesca Romana Ponziani
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Marco Biolato
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Venanzio Valenza
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Giuseppe Marrone
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Gabriele Sganga
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Antonio Gasbarrini
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Luca Miele
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Antonio Grieco
- Fondazione Policlinico Universitario A Gemelli IRCCS, Rome 00168, Italy
- Università Cattolica del Sacro Cuore, Rome 00168, Italy
| |
Collapse
|
44
|
Iacob S, Iacob DG. Infectious Threats, the Intestinal Barrier, and Its Trojan Horse: Dysbiosis. Front Microbiol 2019; 10:1676. [PMID: 31447793 PMCID: PMC6692454 DOI: 10.3389/fmicb.2019.01676] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
The ecosystem of the gut microbiota consists of diverse intestinal species with multiple metabolic and immunologic activities and it is closely connected with the intestinal epithelia and mucosal immune response, with which it builds a complex barrier against intestinal pathogenic bacteria. The microbiota ensures the integrity of the gut barrier through multiple mechanisms, either by releasing antibacterial molecules (bacteriocins) and anti-inflammatory short-chain fatty acids or by activating essential cell receptors for the immune response. Experimental studies have confirmed the role of the intestinal microbiota in the epigenetic modulation of the gut barrier through posttranslational histone modifications and regulatory mechanisms induced by epithelial miRNA in the epithelial lumen. Any quantitative or functional changes of the intestinal microbiota, referred to as dysbiosis, alter the immune response, decrease epithelial permeability and destabilize intestinal homeostasis. Consequently, the overgrowth of pathobionts (Staphylococcus, Pseudomonas, and Escherichia coli) favors intestinal translocations with Gram negative bacteria or their endotoxins and could trigger sepsis, septic shock, secondary peritonitis, or various intestinal infections. Intestinal infections also induce epithelial lesions and perpetuate the risk of bacterial translocation and dysbiosis through epithelial ischemia and pro-inflammatory cytokines. Furthermore, the decline of protective anaerobic bacteria (Bifidobacterium and Lactobacillus) and inadequate release of immune modulators (such as butyrate) affects the release of antimicrobial peptides, de-represses microbial virulence factors and alters the innate immune response. As a result, intestinal germs modulate liver pathology and represent a common etiology of infections in HIV immunosuppressed patients. Antibiotic and antiretroviral treatments also promote intestinal dysbiosis, followed by the selection of resistant germs which could later become a source of infections. The current article addresses the strong correlations between the intestinal barrier and the microbiota and discusses the role of dysbiosis in destabilizing the intestinal barrier and promoting infectious diseases.
Collapse
Affiliation(s)
- Simona Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Balş", Bucharest, Romania
| | - Diana Gabriela Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
45
|
Kucherenko AM, Moroz LV, Bevz TI, Bulavenko VI, Antypkin YG, Berezenko VS, Dyba MB, Pampukha VM, Gorodna OV, Livshits LA. Investigation of rs11536889 + 3725G/C Polymorphism of the TLR4 Gene in Patients with Autoimmune and Chronic Viral Hepatitis C. CYTOL GENET+ 2019. [DOI: 10.3103/s0095452719040078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
46
|
Li YT, Ye JZ, Lv LX, Xu H, Yang LY, Jiang XW, Wu WR, Shi D, Fang DQ, Bian XY, Wang KC, Wang QQ, Xie JJ, Lu YM, Li LJ. Pretreatment With Bacillus cereus Preserves Against D-Galactosamine-Induced Liver Injury in a Rat Model. Front Microbiol 2019; 10:1751. [PMID: 31417535 PMCID: PMC6685349 DOI: 10.3389/fmicb.2019.01751] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022] Open
Abstract
Bacillus cereus (B. cereus) functions as a probiotic in animals, but the underlying mechanisms remain unclear. We aim to evaluate the protective effects and definite mechanism by which orally administered B. cereus prevents D-galactosamine (D-GalN)-induced liver injury in rats. Twenty-one Sprague–Dawley rats were equally assigned into three groups (N = 7 animals per group). B. cereus ATCC11778 (2 × 109 colony-forming units/ml) was administered to the B. cereus group via gavage, and phosphate-buffered saline was administered to the positive control (PC) and negative control (NC) groups for 2 weeks. The PC and B. cereus groups received 1.1 g/kg D-GalN via an intraperitoneal injection to induce liver injury. The blood, terminal ileum, liver, kidney and mesenteric lymph nodes (MLNs) were collected for histological examinations and to evaluate bacterial translocation. Liver function was also determined. Fecal samples were collected for deep sequencing of the 16S rRNA on an Illumina MiSeq platform. B. cereus significantly attenuated D-GalN-induced liver injury and improved serum alanine aminotransferase (ALT) and serum cholinesterase levels (P < 0.05 and P < 0.01, respectively). B. cereus modulated cytokine secretion, as indicated by the elevated levels of the anti-inflammatory cytokine interleukin-10 (IL-10) in both the liver and plasma (P < 0.05 and P < 0.01, respectively) and the substantially decreased levels of the cytokine IL-13 in the liver (P < 0.05). Pretreatment with B. cereus attenuated anoxygenic bacterial translocation in the veins (P < 0.05) and liver (P < 0.05) and upregulated the expression of the tight junction protein 1. The gut microbiota from the B. cereus group clustered separately from that of the PC group, with an increase in species of the Ruminococcaceae and Peptococcaceae families and a decrease in those of the Parabacteroides, Paraprevotella, and Desulfovibrio families. The potential probiotic B. cereus attenuated liver injury by restoring the gut flora balance and enhancing the intestinal barrier function.
Collapse
Affiliation(s)
- Ya-Ting Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Jian-Zhong Ye
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Long-Xian Lv
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Hong Xu
- Department of Orthopedics, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou, China
| | - Li-Ya Yang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xian-Wan Jiang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Wen-Rui Wu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Ding Shi
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Dai-Qiong Fang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xiao-Yuan Bian
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Kai-Cen Wang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Qiang-Qiang Wang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Jiao-Jiao Xie
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Yan-Meng Lu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Lan-Juan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
47
|
Shi L, Zheng X, Fan Y, Yang X, Li A, Qian J. The contribution of miR-122 to the innate immunity by regulating toll-like receptor 4 in hepatoma cells. BMC Gastroenterol 2019; 19:130. [PMID: 31340754 PMCID: PMC6657172 DOI: 10.1186/s12876-019-1048-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a kind of malignancies to impact human health. It has been reported that aberrant toll-like receptor (TLR) signaling may contribute to the development and progression of HCC, especially TLR4. MiR-122, which extensively involved in hepatitis virus infection and the apoptosis of hepatoma cells, might be decreased in HCC patients livers. The hypothesis of this study was whether miR-122 plays a role in inflammatory pathways through regulating TLR4 expression in hepatoma cells. METHODS The expression of miR-122 in the tissues of HCC patients compared to controls in TCGA datasets was analyzed. The relationship between miR-122 and TLR4 was detected in HCC cell lines by increasing/decreasing miR-122 expression. The target of miR-122 on TLR4 was confirmed by luciferase reporter assays. The proliferation of HCC cells and production of proinflammatory cytokines were measured with miR-122 upregulation and inhibition. RESULTS We found that the expression of miR-122 was decreased in HCC tissues and showed the diagnostic capacity for HCC in TCGA datasets. MiR-122 and TLR4 expression have negative correlation in normal liver cells and HCC cells. Upregulation of miR-122 significantly inhibited TLR4 expression in hepatoma cells, including in hepatoma cells with the induction of LPS, while knocking down miR-122 increased TLR4 expression. By screening potential miR-122 targets among TLR4, we found that there was a putative miR-122 target in TLR4 3'UTR. Mutations in the nt1603-nt1609 region of TLR4 3'UTR abandoned the impact of miR-122 on TLR4 expression. Over-expression/down-expression of miR-122 could influence the proliferation and the expression of natural immune factors. CONCLUSIONS MiR-122 might target TLR4 and regulate host innate immunity in hepatoma cells, which revealed a new molecular mechanism of miR-122 on the regulation of innate immunity.
Collapse
Affiliation(s)
- Liyu Shi
- Department of Microbiology, Harbin Medical University, No. 194, Xuefu Road, Harbin, 150081, Heilongjiang province, China.,Wu Lien-Teh institutes, Harbin Medical University, Harbin, Heilongjiang province, China
| | - Xiaoqiu Zheng
- Department of Microbiology, Harbin Medical University, No. 194, Xuefu Road, Harbin, 150081, Heilongjiang province, China.,Wu Lien-Teh institutes, Harbin Medical University, Harbin, Heilongjiang province, China
| | - Yuzhuo Fan
- Department of Microbiology, Harbin Medical University, No. 194, Xuefu Road, Harbin, 150081, Heilongjiang province, China
| | - Xiaolan Yang
- Department of Microbiology, Harbin Medical University, No. 194, Xuefu Road, Harbin, 150081, Heilongjiang province, China.,Wu Lien-Teh institutes, Harbin Medical University, Harbin, Heilongjiang province, China
| | - Aimei Li
- Department of Microbiology, Harbin Medical University, No. 194, Xuefu Road, Harbin, 150081, Heilongjiang province, China.,Wu Lien-Teh institutes, Harbin Medical University, Harbin, Heilongjiang province, China
| | - Jun Qian
- Department of Microbiology, Harbin Medical University, No. 194, Xuefu Road, Harbin, 150081, Heilongjiang province, China. .,Wu Lien-Teh institutes, Harbin Medical University, Harbin, Heilongjiang province, China.
| |
Collapse
|
48
|
Qiu H, Mao D, Tang N, Long F, Zhang R, Wang M, Shi Q, Li J, Jiang Q, Chen Y, Wang X. The underlying mechanisms of Jie-Du-Hua-Yu granule for protecting rat liver failure. Drug Des Devel Ther 2019; 13:589-600. [PMID: 30809090 PMCID: PMC6376884 DOI: 10.2147/dddt.s180969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Jie-Du-Hua-Yu (JDHY) granule is a combination of six traditional Chinese medicines with known therapeutic effect in treating acute liver failure (ALF). The aim of this study was to investigate the amelioration efficacy of JDHY in lipopolysaccharide/D-galactosamine (LPS/D-GalN)-induced ALF in rat and explore the possible molecular mechanism underlying the therapeutic efficacy. MATERIALS AND METHODS The efficacy of JDHY was determined by assessing hepatic pathology and function in LPS and D-GalN challenged Wistar rat. We also evaluated the effect of JDHY on LPS-induced Kupffer cells by measuring inflammatory cytokines and determining the phenotypic function. By means of bioinformatics analysis of liver tissue and validation in Kupffer cells, we identified possible pathways involved in the pharmacologic action of mechanism of JDHY. RESULTS JDHY could attenuate LPS-induced liver injury in rat by inhibiting apoptosis and increasing hepatic activity. In vitro study showed that JDHY could decrease the production of proinflammatory cytokines (tumor necrosis factor-α, IL6, and interferon-γ), increase anti-inflammatory cytokines (IL10, IL13), and promote cell survival and proliferation, possibly due to inhibition of IκB/nuclear factor-κB (NF-κB) signaling pathway and expression of CD14 and CXCL2, which was consistent with the findings from bioinformatics analysis. CONCLUSION Our results revealed that JDHY protected against LPS-induced liver damage both in vitro and in vivo, by inhibiting the NF-κB-mediated inflammatory pathway, indicating its potential function to treat liver diseases.
Collapse
Affiliation(s)
- Hua Qiu
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| | - Dewen Mao
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| | - Nong Tang
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi 530200, China,
| | - Fuli Long
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| | - Rongzhen Zhang
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| | - Minggang Wang
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| | - Qinglan Shi
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| | - Jiahuan Li
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| | - Qin Jiang
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| | - Yueqiao Chen
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| | - Xiufeng Wang
- Department of Liver Disease, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, Guangxi 530023, China,
| |
Collapse
|
49
|
Interplay between early-life malnutrition, epigenetic modulation of the immune function and liver diseases. Nutr Res Rev 2019; 32:128-145. [PMID: 30707092 DOI: 10.1017/s0954422418000239] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Early-life nutrition plays a critical role in fetal growth and development. Food intake absence and excess are the two main types of energy malnutrition that predispose to the appearance of diseases in adulthood, according to the hypothesis of 'developmental origins of health and disease'. Epidemiological data have shown an association between early-life malnutrition and the metabolic syndrome in later life. Evidence has also demonstrated that nutrition during this period of life can affect the development of the immune system through epigenetic mechanisms. Thus, epigenetics has an essential role in the complex interplay between environmental factors and genetics. Altogether, this leads to the inflammatory response that is commonly seen in non-alcoholic fatty liver disease (NAFLD), the hepatic manifestation of the metabolic syndrome. In conjunction, DNA methylation, covalent modification of histones and the expression of non-coding RNA are the epigenetic phenomena that affect inflammatory processes in the context of NAFLD. Here, we highlight current understanding of the mechanisms underlying developmental programming of NAFLD linked to epigenetic modulation of the immune system and environmental factors, such as malnutrition.
Collapse
|
50
|
Human Toll-Like Receptor 4 (hTLR4): Structural and functional dynamics in cancer. Int J Biol Macromol 2019; 122:425-451. [DOI: 10.1016/j.ijbiomac.2018.10.142] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/10/2018] [Accepted: 10/18/2018] [Indexed: 12/23/2022]
|