1
|
Nóvoa E, da Silva Lima N, Gonzalez-Rellan MJ, Chantada-Vazquez MD, Verheij J, Rodriguez A, Esquinas-Roman EM, Fondevila MF, Koning M, Fernandez U, Cabaleiro A, Parracho T, Iglesias-Moure J, Seoane S, Porteiro B, Escudero A, Senra A, Perez-Fernandez R, López M, Fidalgo M, Guallar D, Martinez-Chantar ML, Dieguez C, Varela-Rey M, Prevot V, Schwaninger M, Meijnikman A, Bravo SB, Frühbeck G, Nogueiras R. Mitochondrial antiviral signaling protein enhances MASLD progression through the ERK/TNFα/NFκβ pathway. Hepatology 2025; 81:1535-1552. [PMID: 38761407 PMCID: PMC11999095 DOI: 10.1097/hep.0000000000000930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 04/19/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND AND AIMS Mitochondrial antiviral signaling protein (MAVS) is a critical regulator that activates the host's innate immunity against RNA viruses, and its signaling pathway has been linked to the secretion of proinflammatory cytokines. However, the actions of MAVS on inflammatory pathways during the development of metabolic dysfunction-associated steatotic liver disease (MASLD) have been little studied. APPROACH AND RESULTS Liver proteomic analysis of mice with genetically manipulated hepatic p63, a transcription factor that induces liver steatosis, revealed MAVS as a target downstream of p63. MAVS was thus further evaluated in liver samples from patients and in animal models with MASLD. Genetic inhibition of MAVS was performed in hepatocyte cell lines, primary hepatocytes, spheroids, and mice. MAVS expression is induced in the liver of both animal models and people with MASLD as compared with those without liver disease. Using genetic knockdown of MAVS in adult mice ameliorates diet-induced MASLD. In vitro, silencing MAVS blunts oleic and palmitic acid-induced lipid content, while its overexpression increases the lipid load in hepatocytes. Inhibiting hepatic MAVS reduces circulating levels of the proinflammatory cytokine TNFα and the hepatic expression of both TNFα and NFκβ. Moreover, the inhibition of ERK abolished the activation of TNFα induced by MAVS. The posttranslational modification O -GlcNAcylation of MAVS is required to activate inflammation and to promote the high lipid content in hepatocytes. CONCLUSIONS MAVS is involved in the development of steatosis, and its inhibition in previously damaged hepatocytes can ameliorate MASLD.
Collapse
Affiliation(s)
- Eva Nóvoa
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), A Coruña, Spain
| | - Natália da Silva Lima
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Maria J. Gonzalez-Rellan
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Maria D.P. Chantada-Vazquez
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, A Coruña, Spain
| | - Joanne Verheij
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Amaia Rodriguez
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), A Coruña, Spain
- Department of Endocrinology & Nutrition, Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, IdiSNA, Navarra, Spain
| | - Eva M. Esquinas-Roman
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Marcos F. Fondevila
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Mirja Koning
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Uxia Fernandez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), A Coruña, Spain
| | - Alba Cabaleiro
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Tamara Parracho
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Jose Iglesias-Moure
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Samuel Seoane
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Begoña Porteiro
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Adriana Escudero
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Ana Senra
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Roman Perez-Fernandez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), A Coruña, Spain
| | - Miguel Fidalgo
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Diana Guallar
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Maria L. Martinez-Chantar
- Liver Disease Lab, BRTA CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain
| | - Carlos Dieguez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Endocrinology & Nutrition, Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, IdiSNA, Navarra, Spain
| | - Marta Varela-Rey
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, A Coruña, Spain
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, European Genomic Institute for Diabetes (EGID), Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Abraham Meijnikman
- Department of Internal and Experimental Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Susana B. Bravo
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), A Coruña, Spain
| | - Gema Frühbeck
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Department of Endocrinology & Nutrition, Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, IdiSNA, Navarra, Spain
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Endocrinology & Nutrition, Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, IdiSNA, Navarra, Spain
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| |
Collapse
|
2
|
Haag M, Winter S, Kemas AM, Tevini J, Feldman A, Eder SK, Felder TK, Datz C, Paulweber B, Liebisch G, Burk O, Lauschke VM, Aigner E, Schwab M. Circulating metabolite signatures indicate differential gut-liver crosstalk in lean and obese MASLD. JCI Insight 2025; 10:e180943. [PMID: 40100312 PMCID: PMC12016937 DOI: 10.1172/jci.insight.180943] [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: 03/11/2024] [Accepted: 03/07/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUNDAlterations in circulating metabolites have been described in obese metabolic dysfunction-associated steatotic liver disease (MASLD), but data on lean MASLD are lacking. We investigated serum metabolites, including microbial bile acids and short-chain fatty acids (SCFAs), and their association with lean and obese MASLD.METHODSSerum samples from 204 people of European descent were allocated to groups: lean healthy, lean MASLD, obese healthy, and obese MASLD (n = 47). Liquid chromatography-mass spectrometry-based metabolomics and linear model analysis were performed. MASLD prediction was assessed based on least absolute shrinkage and selection operator regression. Functional effects of altered molecules were verified in organotypic 3D primary human liver cultures.RESULTSLean MASLD was characterized by elevated isobutyrate, methionine sulfoxide, propionate, and phosphatidylcholines. Patients with obese MASLD had increased sarcosine and decreased lysine and asymmetric dimethylarginine. Using metabolites, sex, and BMI, MASLD versus healthy could be predicted with a median AUC of 86.5% and 85.6% in the lean and obese subgroups, respectively. Functional experiments in organotypic 3D primary human liver cultures showed propionate and isobutyrate induced lipid accumulation and altered expression of genes involved in lipid and glucose metabolism.CONCLUSIONLean MASLD is characterized by a distinct metabolite pattern related to amino acid metabolism, lipids, and SCFAs, while metabolic pathways of lipid accumulation are differentially activated by microbial metabolites. We highlight an important role of microbial metabolites in MASLD, with implications for predictive and mechanistic assessment of liver disease across weight categories.FUNDINGRobert Bosch Stiftung, Swedish Research Council (2021-02801, 2023-03015, 2024-03401), ERC Consolidator Grant 3DMASH (101170408), Ruth and Richard Julin Foundation for Gastroenterology (2021-00158), SciLifeLab and Wallenberg National Program for Data-Driven Life Science (WASPDDLS22:006), Novo Nordisk Foundation (NNF23OC0085944, NNF23OC0084420), PMU-FFF (E-18/28/148-FEL).
Collapse
Affiliation(s)
- Mathias Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Aurino M. Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Alexandra Feldman
- Obesity Research Unit, and
- Department of Internal Medicine I, Paracelsus Medical University, Salzburg, Austria
| | - Sebastian K. Eder
- Obesity Research Unit, and
- Department of Internal Medicine I, Paracelsus Medical University, Salzburg, Austria
| | | | - Christian Datz
- Obesity Research Unit, and
- Department of Internal Medicine, Hospital Oberndorf, Oberndorf, Austria
| | - Bernhard Paulweber
- Obesity Research Unit, and
- Department of Internal Medicine I, Paracelsus Medical University, Salzburg, Austria
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Oliver Burk
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
| | - Volker M. Lauschke
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Elmar Aigner
- Obesity Research Unit, and
- Department of Internal Medicine I, Paracelsus Medical University, Salzburg, Austria
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Departments of Clinical Pharmacology and of Biochemistry and Pharmacy, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Jain P, Jain A, Deshmukh R, Samal P, Satapathy T, Ajazuddin. Metabolic dysfunction-associated steatotic liver disease (MASLD): Exploring systemic impacts and innovative therapies. Clin Res Hepatol Gastroenterol 2025; 49:102584. [PMID: 40157567 DOI: 10.1016/j.clinre.2025.102584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/01/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), which includes the inflammatory subtype metabolic dysfunction-associated steatohepatitis, is a prominent cause of chronic liver disease with systemic effects. Insulin resistance, obesity, and dyslipidaemia produce MASLD in over 30 % of adults. It is a global health issue. From MASLD to MASH, hepatic inflammation and fibrosis grow, leading to cirrhosis, hepatocellular cancer, and extrahepatic complications such CVD, CKD, and sarcopenia. Effects of MASLD to MASH are mediated through mechanisms that include inflammation, oxidative stress, dysbiosis, and predisposition through genetic makeup. Advances in diagnostic nomenclature in the past few years have moved the emphasis away from NAFLD to MASLD, focusing on the metabolic etiology and away from the stigma of an alcoholic-related condition. Epidemiological data show a large geographical variability and increasing prevalence in younger populations, particularly in regions with high carbohydrate-rich diets and central adiposity. Lifestyle modification is considered as the main management of MASLD currently. This may include dietary intervention, exercise, and weight loss management. Pharmaceutical management is primarily aimed at metabolic dysfunction with promising findings for GLP-1 receptor agonists, pioglitazone and SGLT-2 inhibitors, which can correct both hepatic and systemic outcome. However, it still depends on well-integrated multidisciplinary care models by considering complex relationships between MASLD and its effects on extrahepatic organs. Determining complications at an early stage; developing precision medicine strategies; exploring new therapeutic targets will represent crucial factors in improving their outcomes. This review discuss the systemic nature of MASLD and calls for multiple collaborations to reduce its far-reaching health impacts and our quest for understanding its pathological mechanisms. Thus, collective efforts that are required to address MASLD are under the public health, clinical care, and research angles toward effectively containing its rapidly increasing burden.
Collapse
Affiliation(s)
- Parag Jain
- Department of Pharmacology, Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India, 490024.
| | - Akanksha Jain
- Department of Biotechnology, Bharti University, Durg, C.G., India
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, India, 281406
| | - Pradeep Samal
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Bilaspur, C.G., India
| | - Trilochan Satapathy
- Department of Pharmacy, Columbia Institute of Pharmaceutical Sciences, Raipur, C.G., India, 493111
| | - Ajazuddin
- Department of Pharmacology, Rungta College of Pharmaceutical Sciences and Research, Bhilai, C.G., India, 490024
| |
Collapse
|
4
|
Shipovskaya AA, Dudanova OP, Kurbatova IV. [Inflammatory cytokines, soluble interleukin-6 receptors, and fragmented cytokeratin-18 as indicators of non-alcoholic steatohepatitis]. TERAPEVT ARKH 2025; 97:115-120. [PMID: 40237746 DOI: 10.26442/00403660.2025.02.203123] [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: 03/03/2023] [Accepted: 07/17/2024] [Indexed: 04/18/2025]
Abstract
AIM To evaluate inflammatory cytokines, such as tumor necrosis factor α (TNF-α), interleukin (IL)-1β, 8, 6, soluble IL-6 receptors (sIL-6R) and fragmented cytokeratin-18 (FCK-18) as indicators of non-alcoholic steatohepatitis (NASH). MATERIALS AND METHODS 173 NASH patients aged 47.0±10.8 years were examined: 118 (68.2%) - men, 55 (31.8%) - women. The following markers were determined: TNF-α (Human TNFα Platinum ELISA, eBioscience, Austria), IL-1β, 8, 6 (Vector-Best, Russia), sIL-6R (Human sIL-6R ELISA, eBioscience, Austria), FCK-18 (TPS ELISA, Biotech, Sweden), insulin (Insulin TEST System, USA), HOMA-IR (Homeostasis Model Assessment of Insulin Resistance) and NAFLD fibrosis score (NFS) were calculated. RESULTS The highest level in NASH patients compared with healthy individuals was observed for IL-6 - 8.4±1.6 pg/ml versus 2.8±0.9 pg/ml (p=0.001), FCK-18 - 295.3±56.3 U/l versus 110.5±30.2 U/l (p=0.0001), then IL-8 - 17.3±6.7 pg/ml vs 7.6±1.9 pg/ml (p=0.003), TNF-α - 6.3±0.4 pg/ml versus 4.1±0.8 pg/ml (p=0.0001), sIL-6R - 151.5±21.2 ng/ml vs 95.9±12.5 ng/ml (p<0.05); IL-1β did not change - 5.3±1.4 pg/ml versus 4.7±1.5 pg/ml (p=0.3) respectively. FCK-18 showed the highest correlations with TNF-α (r=0.73), HOMA-IR (r=0.73), alanine aminotransferase (r=0.71), erythrocyte sedimentation rate (r=0.23), IL-6 (r=0.22); p<0.05. TNF-α correlated with FCK-18 (r=0.73), cholesterol (r=0.61), albumin (r=-0.42), fibrinogen (r=0.21), leukocyte count (r=0.21); p<0.05. IL-8 correlated with triglycerides (r=0.79) and HDL (r=-0.77), IL-6 - with NFS (r=0.63) and FCK-18 (r=0.22), rIL-6R - with aspartate aminotransferase (r=0.62); p<0.05. CONCLUSION TNF-α, IL-8, 6, sIL-6R and FCK-18 should be used as non-invasive biomarkers of NASH.
Collapse
Affiliation(s)
| | | | - I V Kurbatova
- Karelian Scientific Center of the Russian Academy of Sciences
| |
Collapse
|
5
|
El-Sehrawy AAMA, Rashid TA, Ullah MI, Uthirapathy S, Ganesan S, Singh A, Devi A, Joshi KK, Jasim AS, Kadhim AJ. Cutting edge: ferroptosis in metabolic dysfunction-associated steatotic liver disease (MASLD) pathogenesis and therapy. Funct Integr Genomics 2025; 25:71. [PMID: 40131513 DOI: 10.1007/s10142-025-01579-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
Ferroptosis denotes a distinct form of controlled cell death marked by substantial iron buildup and significant lipid peroxidation, playing a crucial role in several disease processes linked to cell death. Given the liver's essential functions in iron and lipid metabolism and its vulnerability to oxidative damage, more research has investigated the correlation between ferroptosis and numerous hepatic diseases, including metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD). NAFLD has arisen as a worldwide public health concern due to elevated morbidity and high death rates. The pathogenesis of MASLD remains incompletely elucidated. Recent data suggests that ferroptosis is crucial in the pathophysiology of MASLD; nevertheless, the specific processes by which ferroptosis influences MASLD remain unclear. The present review summarizes the molecular processes of ferroptosis and its intricate regulatory networks, outlines the differing impacts of ferroptosis at different stages of MASLD, and examines possible approaches targeting ferroptosis for the therapy of MASLD, suggesting a novel approach for its management.
Collapse
Affiliation(s)
| | - Teeba Ammar Rashid
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq.
| | - Muhammad Ikram Ullah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, 72388, Aljouf, Saudi Arabia
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, 248002, Uttarakhand, India
- Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| | - Ahmed Salman Jasim
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 5100, Babylon, Iraq
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| |
Collapse
|
6
|
Song J, Wang H, Gao X, Yang F, Zhu X, Qiao G, Gan T, Tao J. The serum hepcidin and the hepcidin/ferritin ratio in NAFLD: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:62. [PMID: 39915727 PMCID: PMC11804044 DOI: 10.1186/s12876-025-03620-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a spectrum of chronic liver diseases characterized by hepatic steatosis exceeding 5% in the absence of alcohol and other liver-damaging factors. Clinical studies have identified a potential link between abnormal iron metabolism and the high incidence of NAFLD; however, the results from clinical trials remain inconsistent. This meta-analysis aims to compare serum hepcidin levels and the hepcidin/ferritin ratio between adults with NAFLD and those without to explore their potential relationship with NAFLD. METHODS A systematic search was conducted across the Web of Science platform, Cochrane, Scopus, Embase, and PubMed databases from their inception until December 18, 2024. The analysis primarily focused on serum hepcidin levels and the hepcidin/ferritin ratio. Observational studies comparing serum hepcidin levels and the hepcidin/ferritin ratio between individuals with NAFLD and control groups were included. A random-effects model was employed to calculate effect estimates, and outcomes were reported as standardized mean differences (SMD) with 95% confidence intervals (95% CI). RESULTS Following the systematic review, a total of 19 studies, comprising 2216 patients and 2125 controls, were included. The findings revealed a statistically significant difference in both hepcidin levels (SMD = 1.03, 95% CI: 0.49 to 1.56, p < 0.001) and the hepcidin/ferritin ratio (SMD = -1.13, 95% CI: -1.79 to -0.46, p < 0.001) between NAFLD and controls. Significant heterogeneity was observed across studies for both hepcidin (I² = 98.2%) and the hepcidin/ferritin ratio (I² = 93.3%), and the limited number of studies on hepcidin/ferritin were acknowledged as key limitations. Subgroup analysis revealed that patients with obesity exhibited higher levels of hepcidin (SMD = 1.12, 95% CI: 0.40 to 1.97) than overweight (SMD = 0.88, 95% CI: 0.05 to 1.72). Meta-regression analysis identified the hepcidin measurement method (p < 0.01), male-to-female ratio (p < 0.01), and study quality (p < 0.01) as significant moderators of the observed heterogeneity. CONCLUSION This meta-analysis revealed a significant association between hepcidin levels, the hepcidin/ferritin ratio and NAFLD in adults. Further investigations are needed to fully elucidate the role of these variables in iron metabolism and their potential impact on the diagnosis, prevention, and management of NAFLD.
Collapse
Affiliation(s)
- Jingmin Song
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Heqing Wang
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiaolian Gao
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, 430065, China.
- Hubei Shizhen Laboratory, Wuhan, 430065, China.
| | - Fen Yang
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Xinhong Zhu
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Guiyuan Qiao
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Ting Gan
- School of Nursing, Hubei University of Chinese Medicine, Wuhan, 430065, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Junxiu Tao
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China.
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China.
| |
Collapse
|
7
|
Akçiçek M, Dağ N. Evaluation of hepatic steatosis in obese children and adolescents using immune-inflammatory markers and shear wave elastography. J Ultrason 2025; 25:1-6. [PMID: 39882078 PMCID: PMC11774258 DOI: 10.15557/jou.2025.0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/27/2024] [Indexed: 01/31/2025] Open
Abstract
Aim To investigate the changes in liver stiffness and immune-inflammatory markers associated with obesity and the degree of hepatic steatosis in obese children and adolescents. Methods A total of 76 obese children and adolescents aged 6-18 years, with body mass index percentiles >95th, were included in the study. Patients with metabolic syndrome, diabetes mellitus, and chronic liver disease were excluded. A control group of 44 patients of healthy and normal-weight children was included. Laboratory values from the past month were analyzed using patient records. Shear wave elastography and ultrasound examinations were performed on a single device by the same experienced radiologist. Results The systemic immune-inflammation index and pan-immune inflammation values were significantly higher in obese patients with hepatic steatosis compared to obese patients without hepatic steatosis (p <0.001). Liver stiffness values were significantly higher in steatotic patients compared to nonsteatotic patients (p <0.001). A significant difference was observed between hepatic steatosis grades in terms of immune-inflammation index and pan-immune inflammation value values (p <0.001). There was a strong, positive, statistically significant correlation between liver stiffness and immune-inflammation index and pan-immune inflammation value (p <0.05). Conclusions Immune-inflammatory biomarkers and shear wave elastography may provide valuable insights into the diagnosis and follow-up of inflammation and fibrosis in the evaluation of hepatic steatosis in obese children and adolescents.
Collapse
Affiliation(s)
- Mehmet Akçiçek
- Radiology Department, Malatya Turgut Özal University, Faculty of Medicine, Malatya, Turkey
| | - Nurullah Dağ
- Radiology, Malatya Training and Research Hospital, Malatya, Turkey
| |
Collapse
|
8
|
Vajdi M, Hassanizadeh S, Hassanizadeh R, Bagherniya M. Curcumin supplementation effect on liver enzymes in patients with nonalcoholic fatty liver disease: a GRADE-assessed systematic review and dose-response meta-analysis of randomized controlled trials. Nutr Rev 2025; 83:1-12. [PMID: 38213188 DOI: 10.1093/nutrit/nuad166] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024] Open
Abstract
CONTEXT Clinical evidence from investigations of the effects of curcumin on liver enzymes in patients with nonalcoholic fatty liver disease (NAFLD) have led to inconsistent results. OBJECTIVE The aim of this systematic review and meta-analysis was to investigate the overall effects of curcumin and curcumin plus piperine supplementation on liver enzymes such as alanine aminotransferase (ALT), alkaline phosphatase (ALP), and aspartate aminotransferase (AST) in patients with NAFLD. DATA SOURCES The Scopus, Web of Science, PubMed, and Cochrane Library databases were searched from inception through July 2023, using search terms representing NAFLD and liver enzymes. Articles were screened independently by 2 researchers based on PICOS inclusion criteria. DATA EXTRACTION The following data were extracted: first author's name, study location, year of publication, mean age, study duration, study design, participants' sex, number of participants in each group, dose of curcumin supplementation, and ALT, ALP, and AST concentrations. Risk of bias was assessed using the Cochrane Collaboration's modified risk-of-bias tool. DATA ANALYSIS Fixed- or random-effects meta-analysis was performed to estimate the effects of curcumin on liver enzymes, considering heterogeneity across studies. The I2 and Cochran's Q tests were used to assess heterogeneity between studies. RESULTS Overall, 15 randomized controlled trials comprising 905 participants were eligible for this meta-analysis. Curcumin supplementation significantly reduced ALT (weighted mean difference [WMD], -4.10, 95%CI, -7.16 to -1.04) and AST (WMD, -3.27; 95%CI, -5.16 to -1.39), but not ALP (WMD, -0.49; 95%CI, -1.79 to 0.82). Curcumin plus piperine supplementation had no significant effect on ALT (WMD, -3.79; 95%CI, -13.30 to 5.72), and AST (WMD, -1.1; 95%CI, -3.32 to 1.09). CONCLUSIONS Curcumin supplementation improved AST and ALT levels compared with the control group. However, better-designed randomized controlled trials with larger sample sizes and of higher quality are needed to assess the effects of curcumin on ALP. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023448231.
Collapse
Affiliation(s)
- Mahdi Vajdi
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shirin Hassanizadeh
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Hassanizadeh
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Mohammad Bagherniya
- Department of Community Nutrition, School of Nutrition and Food Science, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
9
|
Hamed M, Abou Khalil NS, Alghriany AA, El-Din H. Sayed A. The protective effects of dietary microalgae against hematological, biochemical, and histopathological alterations in pyrogallol-intoxicated Clarias gariepinus. Heliyon 2024; 10:e40930. [PMID: 39759355 PMCID: PMC11699231 DOI: 10.1016/j.heliyon.2024.e40930] [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: 04/23/2024] [Revised: 11/05/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
Microalgae have well-established health benefits for farmed fish. Thus, this study aims to explore the potential protective effects of Spirulina platensis, Chlorella vulgaris, and Moringa oleifera against pyrogallol-induced hematological, hepatic, and renal biomarkers in African catfish (Clarias gariepinus), as well as the histopathological changes in the liver and kidney. Fish weighing 200 ± 25 g were divided into several groups: group 1 served as the control, group 2 was exposed to 10 mg/L of pyrogallol, and groups 3, 4, and 5 were exposed to the same concentration of pyrogallol, supplemented with S. platensis at 20 g/kg diet, C. vulgaris at 50 g/kg diet, and M. oleifera at 5 g/kg diet, respectively, for 15 days. Exposure to pyrogallol led to decreased packed cell volume (PCV) and lymphocyte count, but these effects were alleviated by microalgae interventions. C. vulgaris and M. oleifera equally restored PCV and increased lymphocyte counts. Supplementation with C. vulgaris and M. oleifera successfully normalized both neutrophil and eosinophil counts. Pyrogallol intoxication engenders an increase in glycemic status, but C. vulgaris and M. oleifera effectively mitigated this rise. Pyrogallol-exposed fish exhibited signs of renal dysfunction, with increased serum creatinine and total cholesterol levels. A significant decrease in both erythrocytic cellular and nuclear abnormalities was observed following supplementation with microalgae. C. vulgaris and M. oleifera showed promise in decreasing serum glucose and creatinine levels, and improving hematological parameters, while S. platensis exhibited limited efficacy in this regard. Exposure to pyrogallol led to a notable decrease in serum superoxide dismutase activity and total antioxidant capacity (TAC), accompanied by an increase in serum malondialdehyde (MDA) levels. Diets enriched with C. vulgaris and M. oleifera effectively restored these parameters to normal levels, whereas S. platensis did not induce significant changes. None of the microalgae improved TAC except for M. oleifera, which significantly enhanced it. MDA levels returned to control levels equally and significantly across all groups. Interleukin-6 levels did not exhibit significant differences between any of the groups. Collectively, the histopathological changes induced by pyrogallol were most prominently alleviated in the pyrogallol + C. vulgaris and pyrogallol + M. oleifera groups, and to a limited degree in the pyrogallol + S. platensis group. While the tested microalgae did not cause hepatic or renal dysfunction, they did lead to metabolic abnormalities. The incorporation of microalgae into the diet holds significant importance in mitigating the metabolic and histological toxicity of pyrogallol and should be considered in the formulation of fish feed.
Collapse
Affiliation(s)
- Mohamed Hamed
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Skip Bertman Drive, Baton Rouge, LA, 70803, USA
- Department of Zoology, Faculty of Science, Al-Azhar University (Assiut branch), Assiut, 71524, Egypt
| | - Nasser S. Abou Khalil
- Department of Medical Physiology, Faculty of Medicine, Assiut University, Assiut, Egypt
- Department of Animal Physiology and Biochemistry, Faculty of veterinary Medicine, Badr University, Assuit, Egypt
| | | | - Alaa El-Din H. Sayed
- Department of Zoology, Faculty of Science, Assiut University, Assiut, 71516, Egypt
- Molecular Biology Research & Studies Institute, Assiut University, 71516, Assiut, Egypt
| |
Collapse
|
10
|
Yao L, Chen T. A combined association of alanine aminotransferase, aspartate transaminase and bilirubin with sleep duration in aged 16-85 years (2005-2010). Medicine (Baltimore) 2024; 103:e40915. [PMID: 39654161 PMCID: PMC11630931 DOI: 10.1097/md.0000000000040915] [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: 08/29/2024] [Revised: 11/13/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
Sleep is a vital restorative process that plays a pivotal role in maintaining the delicate equilibrium of mental and physical well-being. Both short and long sleep duration are associated with a range of adverse health outcomes. Numerous studies have consistently demonstrated a robust association between sleep duration and liver disease. In this study, we conducted statistical tests and performed subgroup analyses to explore potential variations in this association across different contexts, aiming to elucidate the correlation between ALT, AST, and TB with sleep duration. This cross-sectional investigation utilized datasets from the National Health and Nutrition Examination Survey 2005 to 2010. Multivariate linear regression models were used to examine the linear association between ALT, AST, and TB with sleep duration. Test for interaction is commonly conducted using multivariabte models to assess statistically significant subgroup disparities. Fitted smoothied curves and threshold effect analyses were employed to depict nonlinear relationships. The study enrolled 17,491 participants aged 16 to 85 years who met the inclusion and exclusion criteria, with a mean age of the participants was 45.58 ± 19.94 years. Multivariate linear regression analysis showed a significant positive association between sleep duration and ALT [-0.23 (-0.45, -0.00) 0.0455] and AST[-0.20 (-0.38, -0.01) 0.0338] in Model 3. Using a two-segment linear regression model, we found an U-shaped relationship and significant inflection point between between ALT and AST with sleep duration. The present study unveiled a significant inverse correlation between sleep duration and levels of ALT and AST, while no significant association was observed with TB levels. Furthermore, variations in the optimal sleep duration for liver function recovery were identified across diverse populations, thereby offering valuable healthcare recommendations to public.
Collapse
Affiliation(s)
- Lishuai Yao
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Tiantian Chen
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Wu X, Song Y, Wu S. Relation of 91 Circulating Inflammatory Proteins to Nonalcoholic Fatty Liver Disease: A Two-Sample Mendelian Randomisation Study. J Cell Mol Med 2024; 28:e70322. [PMID: 39720899 DOI: 10.1111/jcmm.70322] [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: 03/25/2024] [Revised: 06/21/2024] [Accepted: 12/15/2024] [Indexed: 12/26/2024] Open
Abstract
Increasingly, emerging research evidence has demonstrated that nonalcoholic fatty liver disease (NAFLD) is a disease closely associated with systemic inflammation. However, the specific upstream inflammatory factors engaged in the pathogenesis of NAFLD remain unclear. Our study aimed to identify the inflammatory regulators causally associated with NAFLD pathogenesis through Mendelian randomisation. A two-sample Mendelian randomisation method was applied to analyse the causal association between 91 circulating inflammatory proteins and NAFLD. Data on circulating inflammatory proteins were derived from samples of European ancestry (14,824 samples) and NAFLD data were obtained from the FinnGen consortium (2025 cases and 284,826 controls). Instrumental variables were selected from the genetic variance and F-statistics were calculated to avoid bias. We adopted the random-effects inverse variance weighting (IVW) method as our primary analytical approach. Supplementary analyses were also implemented, including weighted median, MR-Egger and weighted mode. Moreover, we conducted pleiotropy and heterogeneity analyses to validate the accuracy of the findings. The application of Mendelian randomisation analysis identified four inflammatory factors that might be causally associated with NAFLD at the genetic level. Elevated levels of eotaxin (or = 1.27, 95% CI: 1.05-1.53, p = 0.014), osteoprotegerin (OPG) (or = 1.29, 1.03-1.60, p = 0.023) and TNFRSF9 (or = 1.32, 95% CI: 1.06-1.64, p = 0.014) may be causally related to an increasing risk of NAFLD. Conversely, heightened leukaemia inhibitory factor (LIF) levels (or = 0.63, 0.44-0.92, p = 0.016) were linked to a lower risk of NAFLD onset. There was no causal relationship between levels of other circulating inflammatory proteins and NAFLD. Our analysis uncovered four upstream inflammatory factors genetically associated with the pathogenesis of NAFLD. These results highlight the potential involvement of inflammation in NAFLD, which provides partial insights for further research in this field in the future.
Collapse
Affiliation(s)
- Xiaodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanhong Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Huang X, You D, An T, Zhao X, Jiang T, Huang Z. Glycyrrhizic acid attenuates the malignant biological properties of nonalcoholic fatty liver disease-related hepatocellular carcinoma. ENVIRONMENTAL TOXICOLOGY 2024; 39:4677-4688. [PMID: 38700384 DOI: 10.1002/tox.24295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024]
Abstract
Glycyrrhizic acid (GA) has effects on anti-hepatic fibrosis, anti-tumor and prevention from hepatocellular carcinoma (HCC) progression. Yet, the capacity of GA to ameliorate the advance of HCC pertinent to nonalcoholic fatty liver disease (NAFLD) remains to be clarified. We used the CCK-8 method to detect the optimal treatment concentration and time for L-02 cells, palmitic acid (PA)-induced L-02 cells and HepG2 cells, and selected 40 μM and 48 h to treat PA-induced L-02 cells and 60 μM for 24 h to treat HepG2 cells. Moreover, functional associations of HepG2 cells were elucidated through various assays. The results showed that GA demonstrated enhances lipid deposition and alleviates the inflammatory response in L-02 cells induced by palmitic acid. Simultaneously, we found that GA inhibits the proliferation, migration, and invasion while promoting apoptosis in HepG2 cells. In pursuit of constructing of HCC model rats, a combination of high-fat diets and diethylnitrosamine was utilized. The results showed that GA significantly decreased the liver index, body weight, liver weight, and the number of nodules in HCC model rats. Moreover, GA mitigated infiltration and heightened apoptosis in these rats. Mechanistically, GA notably attenuated the KKβ/NF-κB pathway in both HepG2 cells and the HCC model rats. In conclusion, GA functions as an inhibitor in the progression of NAFLD-related HCC cells, which might be relevant to the KKβ/NF-κB pathway. Therefore, GA is a potential drug for NAFLD-related HCC treatment.
Collapse
Affiliation(s)
- Xueqing Huang
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Dengwei You
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Tianzhi An
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xuya Zhao
- Department of Interventional Radiology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Tianpeng Jiang
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhi Huang
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
13
|
Lima RS, Belchior-Bezerra M, Silva de Oliveira D, Rocha RDS, Medeiros NI, Mattos RT, Dos Reis IC, Marques AS, Rosário PW, Calsolari MR, Correa-Oliveira R, Dutra WO, Moreira PR, Gomes JA. Obesity Influences T CD4 Lymphocytes Subsets Profiles in Children and Adolescent's Immune Response. J Nutr 2024; 154:3133-3143. [PMID: 39019165 DOI: 10.1016/j.tjnut.2024.07.006] [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: 03/21/2024] [Revised: 06/14/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND Evidence shows that CD4+ T cells are altered in obesity and play a significant role in the systemic inflammation in adults with the disease. OBJECTIVES Because the profile of these cells is poorly understood in the pediatric population, this study aims to investigate the profile of CD4+ T lymphocytes and the plasma levels of cytokines in this population. METHODS Using flow cytometry, we compared the expression profile of lymphocyte markers, master transcription factors, cytokines, and molecules involved in the regulation of the immune response in CD4+ T cells from children and adolescents with obesity (OB group, n = 20) with those with eutrophy group (EU group, n = 16). Plasma levels of cytokines in both groups were determined by cytometric bead array (CBA). RESULTS The OB group presents a lower frequency of CD3+ T cells, as well as a decreased frequency of CD4+ T cells expressing CD28, IL-4, and FOXP3, but an increased frequency of CD4+IL-17A+ cells compared with the EU group. The frequency of CD28 is increased in Th2 and Treg cells in the OB group, whereas CTLA-4 is decreased in all subpopulations compared with the EU group. Furthermore, Th2, Th17, and Treg profiles can differentiate the EU and OB groups. IL-10 plasma levels are reduced in the OB group and negatively correlated with adiposity and inflammatory parameters. CONCLUSIONS CD4+ T cells have an altered pattern of expression in children and adolescents with obesity, contributing to the inflammatory state and clinical characteristics of these patients.
Collapse
Affiliation(s)
- Rafael Silva Lima
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mayara Belchior-Bezerra
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniela Silva de Oliveira
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Nayara I Medeiros
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Imunologia Celular e Molecular, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ, Belo Horizonte, Brazil
| | - Rafael T Mattos
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabelle Camile Dos Reis
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aiessa Santos Marques
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pedro Ws Rosário
- Centro de Especialidades Médicas (CEM), Hospital Santa Casa, Belo Horizonte, Brazil
| | | | - Rodrigo Correa-Oliveira
- Imunologia Celular e Molecular, Instituto René Rachou, Fundação Oswaldo Cruz-FIOCRUZ, Belo Horizonte, Brazil
| | - Walderez O Dutra
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paula Rocha Moreira
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana As Gomes
- Laboratório de Biologia das Interações Celulares, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
14
|
Xia M, Varmazyad M, Pla-Palacín I, Gavlock DC, DeBiasio R, LaRocca G, Reese C, Florentino RM, Faccioli LAP, Brown JA, Vernetti LA, Schurdak M, Stern AM, Gough A, Behari J, Soto-Gutierrez A, Taylor DL, Miedel MT. Comparison of wild-type and high-risk PNPLA3 variants in a human biomimetic liver microphysiology system for metabolic dysfunction-associated steatotic liver disease precision therapy. Front Cell Dev Biol 2024; 12:1423936. [PMID: 39324073 PMCID: PMC11422722 DOI: 10.3389/fcell.2024.1423936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/16/2024] [Indexed: 09/27/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a worldwide health epidemic with a global occurrence of approximately 30%. The pathogenesis of MASLD is a complex, multisystem disorder driven by multiple factors, including genetics, lifestyle, and the environment. Patient heterogeneity presents challenges in developing MASLD therapeutics, creating patient cohorts for clinical trials, and optimizing therapeutic strategies for specific patient cohorts. Implementing pre-clinical experimental models for drug development creates a significant challenge as simple in vitro systems and animal models do not fully recapitulate critical steps in the pathogenesis and the complexity of MASLD progression. To address this, we implemented a precision medicine strategy that couples the use of our liver acinus microphysiology system (LAMPS) constructed with patient-derived primary cells. We investigated the MASLD-associated genetic variant patatin-like phospholipase domain-containing protein 3 (PNPLA3) rs738409 (I148M variant) in primary hepatocytes as it is associated with MASLD progression. We constructed the LAMPS with genotyped wild-type and variant PNPLA3 hepatocytes, together with key non-parenchymal cells, and quantified the reproducibility of the model. We altered media components to mimic blood chemistries, including insulin, glucose, free fatty acids, and immune-activating molecules to reflect normal fasting (NF), early metabolic syndrome (EMS), and late metabolic syndrome (LMS) conditions. Finally, we investigated the response to treatment with resmetirom, an approved drug for metabolic syndrome-associated steatohepatitis (MASH), the progressive form of MASLD. This study, using primary cells, serves as a benchmark for studies using "patient biomimetic twins" constructed with patient induced pluripotent stem cell (iPSC)-derived liver cells using a panel of reproducible metrics. We observed increased steatosis, immune activation, stellate cell activation, and secretion of pro-fibrotic markers in the PNPLA3 GG variant compared to the wild-type CC LAMPS, consistent with the clinical characterization of this variant. We also observed greater resmetirom efficacy in the PNPLA3 wild-type CC LAMPS compared to the GG variant in multiple MASLD metrics, including steatosis, stellate cell activation, and the secretion of pro-fibrotic markers. In conclusion, our study demonstrates the capability of the LAMPS platform for the development of MASLD precision therapeutics, enrichment of patient cohorts for clinical trials, and optimization of therapeutic strategies for patient subgroups with different clinical traits and disease stages.
Collapse
Affiliation(s)
- Mengying Xia
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mahboubeh Varmazyad
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Iris Pla-Palacín
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dillon C. Gavlock
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Richard DeBiasio
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Gregory LaRocca
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Celeste Reese
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rodrigo M. Florentino
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lanuza A. P. Faccioli
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jacquelyn A. Brown
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lawrence A. Vernetti
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mark Schurdak
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Andrew M. Stern
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Albert Gough
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jaideep Behari
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Alejandro Soto-Gutierrez
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Transcriptional Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| | - D. Lansing Taylor
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mark T. Miedel
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
15
|
Wang S, Sun D, Ye B, Xu G, Zou J. Dietary kelp meal improves serum antioxidants, intestinal immunity, and lipid metabolism in hybrid snakehead (Channa maculata ♀ × Channa argus ♂). JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:7464-7475. [PMID: 38733135 DOI: 10.1002/jsfa.13566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 03/28/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Dietary kelp possesses a variety of useful biological qualities but does not have a toxic effect on the host. In this study, we examine how kelp dietary supplementation enhances the serum biochemistry, intestinal immunity, and metabolism of hybrid snakehead. A total of 810 juvenile hybrid snakeheads (Channa maculata ♀ × Channa argus ♂), with an initial average weight of 11.4 ± 0.15 g, were allocated randomly to three treatment groups (three replicates per group). The fish were fed for 60 days with isonitrogenous and isolipidic diets. The groups were the control group (C) (20% high-gluten flour), the medium replacement group (MR) (10% high-gluten flour and 10% kelp meal), and the full replacement group (FR) (0% high-gluten flour and 15% kelp meal). RESULTS The results showed that dietary kelp increased the activity of serum antioxidant enzymes significantly and decreased the content of serum malondialdehyde (MDA) in hybrid snakeheads, with significant changes in the FR group (P < 0.05). The intestinal morphology results showed that dietary kelp helped to increase the specific surface area of intestinal villi, which was beneficial for intestinal digestion and absorption. According to transcriptome and quantitative real-time polymerase chain reaction (qRT-PCR) analysis, dietary kelp can improve the expression of intestinal immunity and metabolism-related pathways. Among them, immune-related genes MHC1 and HSPA1 were significantly up-regulated, and IGH, MHC2, and IL-8 were significantly down-regulated (P < 0.05). Lipid metabolism-related genes DGAT2, FABP2, RXRα, and PLPP1 were all significantly up-regulated (P < 0.05). CONCLUSION Dietary kelp can effectively improve the antioxidant function of hybrid snakeheads, improve intestinal morphology, reduce intestinal inflammation, and promote intestinal lipid synthesis and transportation, thereby improving intestinal immunity and metabolic functions. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Shaodan Wang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Di Sun
- Joint Laboratory of Guangdong province and Hong Kong region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Bin Ye
- Joint Laboratory of Guangdong province and Hong Kong region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| | - Guohuan Xu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jixing Zou
- Joint Laboratory of Guangdong province and Hong Kong region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China
| |
Collapse
|
16
|
Gao H, Peng X, Li N, Gou L, Xu T, Wang Y, Qin J, Liang H, Ma P, Li S, Wu J, Qin X, Xue B. Emerging role of liver-bone axis in osteoporosis. J Orthop Translat 2024; 48:217-231. [PMID: 39290849 PMCID: PMC11407911 DOI: 10.1016/j.jot.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/19/2024] [Accepted: 07/16/2024] [Indexed: 09/19/2024] Open
Abstract
Background Increasing attention to liver-bone crosstalk has spurred interest in targeted interventions for various forms of osteoporosis. Liver injury induced by different liver diseases can cause an imbalance in bone metabolism, indicating a novel regulatory paradigm between the liver and bone. However, the role of the liver-bone axis in both primary and secondary osteoporosis remains inadequately elucidated. Therefore, exploring the exact regulatory mechanisms of the liver-bone axis may offer innovative clinical approaches for treating diseases associated with the liver and bone. Methods Here, we summarize the latest research on the liver-bone axis by searching the PubMed and Web of Science databases and discuss the possible mechanism of the liver-bone axis in different types of osteoporosis. The literature directly reporting the regulatory role of the liver-bone axis in different types of osteoporosis from the PubMed and Web of Science databases has been included in the discussion of this review (including but not limited to the definition of the liver-bone axis, clinical studies, and basic research). In addition, articles discussing changes in bone metabolism caused by different etiologies of liver injury have also been included in the discussion of this review (including but not limited to clinical studies and basic research). Results Several endocrine factors (IGF-1, FGF21, hepcidin, vitamin D, osteocalcin, OPN, LCAT, Fetuin-A, PGs, BMP2/9, IL-1/6/17, and TNF-α) and key genes (SIRT2, ABCB4, ALDH2, TFR2, SPTBN1, ZNF687 and SREBP2) might be involved in the regulation of the liver-bone axis. In addition to the classic metabolic pathways involved in inflammation and oxidative stress, iron metabolism, cholesterol metabolism, lipid metabolism and immunometabolism mediated by the liver-bone axis require more research to elucidate the regulatory mechanisms involved in osteoporosis. Conclusion During primary and secondary osteoporosis, the liver-bone axis is responsible for liver and bone homeostasis via several hepatokines and osteokines as well as biochemical signaling. Combining multiomics technology and data mining technology could further advance our understanding of the liver-bone axis, providing new clinical strategies for managing liver and bone-related diseases.The translational potential of this article is as follows: Abnormal metabolism in the liver could seriously affect the metabolic imbalance of bone. This review summarizes the indispensable role of several endocrine factors and biochemical signaling pathways involved in the liver-bone axis and emphasizes the important role of liver metabolic homeostasis in the pathogenesis of osteoporosis, which provides novel potential directions for the prevention, diagnosis, and treatment of liver and bone-related diseases.
Collapse
Affiliation(s)
- Hongliang Gao
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
- Jiangsu Key Laboratory of Early Development and Chronic Disease Prevention in Children,Nanjing, Jiangsu,PR China
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
- Department of pathophysiology, Wannan Medical College, Wuhu, Anhui, PR China
| | - Xing Peng
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Ning Li
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Liming Gou
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
- Jiangsu Key Laboratory of Early Development and Chronic Disease Prevention in Children,Nanjing, Jiangsu,PR China
| | - Tao Xu
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Yuqi Wang
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Jian Qin
- Department of Orthoprdics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu , PR China
| | - Hui Liang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Peiqi Ma
- Medical Imaging Center, Fuyang People's Hospital, Fuyang, Anhui, PR China
| | - Shu Li
- Department of pathophysiology, Wannan Medical College, Wuhu, Anhui, PR China
| | - Jing Wu
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
- Jiangsu Key Laboratory of Early Development and Chronic Disease Prevention in Children,Nanjing, Jiangsu,PR China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Xihu Qin
- Department of General Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, PR China
| | - Bin Xue
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
- Jiangsu Key Laboratory of Early Development and Chronic Disease Prevention in Children,Nanjing, Jiangsu,PR China
- Core Laboratory, Department of Clinical Laboratory, Sir Run Run Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, PR China
- Department of General Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, PR China
| |
Collapse
|
17
|
Sandireddy R, Sakthivel S, Gupta P, Behari J, Tripathi M, Singh BK. Systemic impacts of metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction-associated steatohepatitis (MASH) on heart, muscle, and kidney related diseases. Front Cell Dev Biol 2024; 12:1433857. [PMID: 39086662 PMCID: PMC11289778 DOI: 10.3389/fcell.2024.1433857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is the most common liver disorder worldwide, with an estimated global prevalence of more than 31%. Metabolic dysfunction-associated steatohepatitis (MASH), formerly known as non-alcoholic steatohepatitis (NASH), is a progressive form of MASLD characterized by hepatic steatosis, inflammation, and fibrosis. This review aims to provide a comprehensive analysis of the extrahepatic manifestations of MASH, focusing on chronic diseases related to the cardiovascular, muscular, and renal systems. A systematic review of published studies and literature was conducted to summarize the findings related to the systemic impacts of MASLD and MASH. The review focused on the association of MASLD and MASH with metabolic comorbidities, cardiovascular mortality, sarcopenia, and chronic kidney disease. Mechanistic insights into the concept of lipotoxic inflammatory "spill over" from the MASH-affected liver were also explored. MASLD and MASH are highly associated (50%-80%) with other metabolic comorbidities such as impaired insulin response, type 2 diabetes, dyslipidemia, hypertriglyceridemia, and hypertension. Furthermore, more than 90% of obese patients with type 2 diabetes have MASH. Data suggest that in middle-aged individuals (especially those aged 45-54), MASLD is an independent risk factor for cardiovascular mortality, sarcopenia, and chronic kidney disease. The concept of lipotoxic inflammatory "spill over" from the MASH-affected liver plays a crucial role in mediating the systemic pathological effects observed. Understanding the multifaceted impact of MASH on the heart, muscle, and kidney is crucial for early detection and risk stratification. This knowledge is also timely for implementing comprehensive disease management strategies addressing multi-organ involvement in MASH pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Madhulika Tripathi
- Cardiovascular and Metabolic Disorders Research Program, Duke-NUS Medical School, Singapore, Singapore
| | - Brijesh Kumar Singh
- Cardiovascular and Metabolic Disorders Research Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
18
|
Yang X, Yao S, Jiang Q, Chen H, Liu S, Shen G, Xiang X, Chen L. Exploring the Regulatory Effect of Tegillarca granosa Polysaccharide on High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease in Mice Based on Intestinal Flora. Mol Nutr Food Res 2024; 68:e2300453. [PMID: 38389187 DOI: 10.1002/mnfr.202300453] [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: 07/03/2023] [Revised: 09/21/2023] [Indexed: 02/24/2024]
Abstract
To explore the potential mechanism of action of Tegillarca granosa polysaccharide (TGP) in treating nonalcoholic fatty liver disease (NAFLD), the study conducts in vivo experiments using male C57BL/6 mice fed a high-fat diet while administering TGP for 16 weeks. The study measures body weight, liver weight, serum biochemical markers, pathological histology, liver lipid accumulation, oxidative stress and inflammation-related factors, lipid synthesis and metabolism-related gene and protein expression, and the composition and abundance of intestinal flora. Additionally, short-chain fatty acid (SCFAs) content and the correlation between intestinal flora and environmental factors are measured. The results show that TGP effectively reduces excessive hepatic lipid accumulation, dyslipidemia, abnormal liver function, and steatosis in the mice with NAFLD. Moreover, TGP effectively regulates intestinal flora disorder, increases the diversity of intestinal flora, and affects the relative abundance of specific bacteria while also increasing the content of SCFAs. These findings provide a basis for exploring the regulatory effect of T. granosa polysaccharide on NAFLD based on intestinal flora and highlight its potential as a natural liver nutraceutical.
Collapse
Affiliation(s)
- Xingwen Yang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Shiwei Yao
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Qihong Jiang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Hui Chen
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Shulai Liu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Guoxin Shen
- Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, 310021, China
| | - Xingwei Xiang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, 310014, China
- Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Hangzhou, 310014, China
- National R&D Branch Center for Pelagic Aquatic Products Processing (Hangzhou), Hangzhou, 310014, China
| | - Lin Chen
- Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, 310021, China
| |
Collapse
|
19
|
Friedline RH, Noh HL, Suk S, Albusharif M, Dagdeviren S, Saengnipanthkul S, Kim B, Kim AM, Kim LH, Tauer LA, Baez Torres NM, Choi S, Kim BY, Rao SD, Kasina K, Sun C, Toles BJ, Zhou C, Li Z, Benoit VM, Patel PR, Zheng DXT, Inashima K, Beaverson A, Hu X, Tran DA, Muller W, Greiner DL, Mullen AC, Lee KW, Kim JK. IFNγ-IL12 axis regulates intercellular crosstalk in metabolic dysfunction-associated steatotic liver disease. Nat Commun 2024; 15:5506. [PMID: 38951527 PMCID: PMC11217362 DOI: 10.1038/s41467-024-49633-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 06/13/2024] [Indexed: 07/03/2024] Open
Abstract
Obesity is a major cause of metabolic dysfunction-associated steatohepatitis (MASH) and is characterized by inflammation and insulin resistance. Interferon-γ (IFNγ) is a pro-inflammatory cytokine elevated in obesity and modulating macrophage functions. Here, we show that male mice with loss of IFNγ signaling in myeloid cells (Lyz-IFNγR2-/-) are protected from diet-induced insulin resistance despite fatty liver. Obesity-mediated liver inflammation is also attenuated with reduced interleukin (IL)-12, a cytokine primarily released by macrophages, and IL-12 treatment in vivo causes insulin resistance by impairing hepatic insulin signaling. Following MASH diets, Lyz-IFNγR2-/- mice are rescued from developing liver fibrosis, which is associated with reduced fibroblast growth factor (FGF) 21 levels. These results indicate critical roles for IFNγ signaling in macrophages and their release of IL-12 in modulating obesity-mediated insulin resistance and fatty liver progression to MASH. In this work, we identify the IFNγ-IL12 axis in regulating intercellular crosstalk in the liver and as potential therapeutic targets to treat MASH.
Collapse
Affiliation(s)
- Randall H Friedline
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Hye Lim Noh
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sujin Suk
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- WCU Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Mahaa Albusharif
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Sezin Dagdeviren
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Suchaorn Saengnipanthkul
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Division of Nutrition, Department of Pediatrics, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Bukyung Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kosin University College of Medicine, Busan, Republic of Korea
| | - Allison M Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lauren H Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lauren A Tauer
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Natalie M Baez Torres
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Stephanie Choi
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Bo-Yeon Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Republic of Korea
| | - Suryateja D Rao
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kaushal Kasina
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Cheng Sun
- Division of Gastroenterology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Benjamin J Toles
- Division of Gastroenterology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Chan Zhou
- Division of Biostatistics and Health Services Research, Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Zixiu Li
- Division of Biostatistics and Health Services Research, Department of Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Vivian M Benoit
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Payal R Patel
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Doris X T Zheng
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kunikazu Inashima
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Annika Beaverson
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Xiaodi Hu
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Duy A Tran
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Werner Muller
- Division of Infection, Immunity & Respiratory Medicine, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Dale L Greiner
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alan C Mullen
- Division of Gastroenterology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ki Won Lee
- WCU Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- XO Center, Advanced Institutes of Convergence Technology, Seoul National University, Suwon, Republic of Korea
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- WCU Biomodulation Major, Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea.
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
20
|
Gong H, He Q, Zhu L, Feng Z, Sun M, Jiang J, Yuan X, Shen Y, Di J. Associations between systemic inflammation indicators and nonalcoholic fatty liver disease: evidence from a prospective study. Front Immunol 2024; 15:1389967. [PMID: 38979415 PMCID: PMC11228160 DOI: 10.3389/fimmu.2024.1389967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 06/10/2024] [Indexed: 07/10/2024] Open
Abstract
Background Although inflammation has been linked to nonalcoholic fatty liver disease (NAFLD), most studies have focused only on a single indicator, leading to inconsistent results. Therefore, a large prospective study that includes a variety of well-documented single and composite indicators of inflammation is needed. This study aimed to thoroughly investigate the potential associations between different systemic inflammatory indicators and NAFLD in the UK Biobank cohort. Methods After excluding ineligible participants, 378,139 individuals were included in the study. Associations between systemic inflammatory indicators and hepatic steatosis were assessed using multivariate logistic regression. The relationships between systemic inflammatory indicators and nonalcoholic fatty liver disease were analysed using Cox proportional hazards models, and nonlinear associations were investigated using restricted cubic splines. Results According to the cross-sectional analysis, systemic inflammatory indicators significantly correlated with hepatic steatosis. Over a median follow-up of 13.9 years, 4,145 individuals developed NAFLD. After sufficient adjustment for confounding factors, CRP levels were found to be nonlinearly positively associated with NAFLD risk (P<0.001), representing the strongest correlation among the tested relationships; lymphocyte count and the LMR showed an L-shaped correlation; monocyte count and neutrophil count showed a linear positive correlation (all P< 0.001); and the NLR, PLR, and SII showed a U-shaped correlation (all P<0.001). Conclusions Multiple systemic inflammatory indicators are strongly associated with the development of NAFLD, and aggressive systemic inflammation management may have a favourable impact on reducing the burden of NAFLD; further randomized controlled studies are needed.
Collapse
Affiliation(s)
- Hao Gong
- Infection Management Department, The First People's Hospital of Changzhou, Changzhou, China
| | - Qida He
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Lili Zhu
- Infection Management Department, The First People's Hospital of Changzhou, Changzhou, China
| | - Zhaolong Feng
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Mengtong Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jingting Jiang
- Tumor Biological Diagnosis and Treatment Center, The First People's Hospital of Changzhou, Changzhou, China
| | - Xiaofeng Yuan
- Department of Spine Surgery, The First People's Hospital of Changzhou, Changzhou, China
| | - Yueping Shen
- Department of Epidemiology and Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Jia Di
- Infection Management Department, The First People's Hospital of Changzhou, Changzhou, China
| |
Collapse
|
21
|
Kochumon S, Malik MZ, Sindhu S, Arefanian H, Jacob T, Bahman F, Nizam R, Hasan A, Thomas R, Al-Rashed F, Shenouda S, Wilson A, Albeloushi S, Almansour N, Alhamar G, Al Madhoun A, Alzaid F, Thanaraj TA, Koistinen HA, Tuomilehto J, Al-Mulla F, Ahmad R. Gut Dysbiosis Shaped by Cocoa Butter-Based Sucrose-Free HFD Leads to Steatohepatitis, and Insulin Resistance in Mice. Nutrients 2024; 16:1929. [PMID: 38931284 PMCID: PMC11207001 DOI: 10.3390/nu16121929] [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: 04/15/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND High-fat diets cause gut dysbiosis and promote triglyceride accumulation, obesity, gut permeability changes, inflammation, and insulin resistance. Both cocoa butter and fish oil are considered to be a part of healthy diets. However, their differential effects on gut microbiome perturbations in mice fed high concentrations of these fats, in the absence of sucrose, remains to be elucidated. The aim of the study was to test whether the sucrose-free cocoa butter-based high-fat diet (C-HFD) feeding in mice leads to gut dysbiosis that associates with a pathologic phenotype marked by hepatic steatosis, low-grade inflammation, perturbed glucose homeostasis, and insulin resistance, compared with control mice fed the fish oil based high-fat diet (F-HFD). RESULTS C57BL/6 mice (5-6 mice/group) were fed two types of high fat diets (C-HFD and F-HFD) for 24 weeks. No significant difference was found in the liver weight or total body weight between the two groups. The 16S rRNA sequencing of gut bacterial samples displayed gut dysbiosis in C-HFD group, with differentially-altered microbial diversity or relative abundances. Bacteroidetes, Firmicutes, and Proteobacteria were highly abundant in C-HFD group, while the Verrucomicrobia, Saccharibacteria (TM7), Actinobacteria, and Tenericutes were more abundant in F-HFD group. Other taxa in C-HFD group included the Bacteroides, Odoribacter, Sutterella, Firmicutes bacterium (AF12), Anaeroplasma, Roseburia, and Parabacteroides distasonis. An increased Firmicutes/Bacteroidetes (F/B) ratio in C-HFD group, compared with F-HFD group, indicated the gut dysbiosis. These gut bacterial changes in C-HFD group had predicted associations with fatty liver disease and with lipogenic, inflammatory, glucose metabolic, and insulin signaling pathways. Consistent with its microbiome shift, the C-HFD group showed hepatic inflammation and steatosis, high fasting blood glucose, insulin resistance, increased hepatic de novo lipogenesis (Acetyl CoA carboxylases 1 (Acaca), Fatty acid synthase (Fasn), Stearoyl-CoA desaturase-1 (Scd1), Elongation of long-chain fatty acids family member 6 (Elovl6), Peroxisome proliferator-activated receptor-gamma (Pparg) and cholesterol synthesis (β-(hydroxy β-methylglutaryl-CoA reductase (Hmgcr). Non-significant differences were observed regarding fatty acid uptake (Cluster of differentiation 36 (CD36), Fatty acid binding protein-1 (Fabp1) and efflux (ATP-binding cassette G1 (Abcg1), Microsomal TG transfer protein (Mttp) in C-HFD group, compared with F-HFD group. The C-HFD group also displayed increased gene expression of inflammatory markers including Tumor necrosis factor alpha (Tnfa), C-C motif chemokine ligand 2 (Ccl2), and Interleukin-12 (Il12), as well as a tendency for liver fibrosis. CONCLUSION These findings suggest that the sucrose-free C-HFD feeding in mice induces gut dysbiosis which associates with liver inflammation, steatosis, glucose intolerance and insulin resistance.
Collapse
Affiliation(s)
- Shihab Kochumon
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Md. Zubbair Malik
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Sardar Sindhu
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Hossein Arefanian
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Texy Jacob
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Fatemah Bahman
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Rasheeba Nizam
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Amal Hasan
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Reeby Thomas
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Fatema Al-Rashed
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Steve Shenouda
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Ajit Wilson
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Shaima Albeloushi
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Nourah Almansour
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Ghadeer Alhamar
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Ashraf Al Madhoun
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Fawaz Alzaid
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Thangavel Alphonse Thanaraj
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Heikki A. Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland;
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, P.O. Box 30, 00271 Helsinki, Finland;
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, P.O. Box 30, 00271 Helsinki, Finland;
- Department of Public Health, University of Helsinki, 00014 Helsinki, Finland
| | - Fahd Al-Mulla
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Rasheed Ahmad
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| |
Collapse
|
22
|
Xia M, Varmazyad M, Palacin IP, Gavlock DC, Debiasio R, LaRocca G, Reese C, Florentino R, Faccioli LAP, Brown JA, Vernetti LA, Schurdak ME, Stern AM, Gough A, Behari J, Soto-Gutierrez A, Taylor DL, Miedel M. Comparison of Wild-Type and High-risk PNPLA3 variants in a Human Biomimetic Liver Microphysiology System for Metabolic Dysfunction-associated Steatotic Liver Disease Precision Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590608. [PMID: 38712213 PMCID: PMC11071381 DOI: 10.1101/2024.04.22.590608] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a worldwide health epidemic with a global occurrence of approximately 30%. The pathogenesis of MASLD is a complex, multisystem disorder driven by multiple factors including genetics, lifestyle, and the environment. Patient heterogeneity presents challenges for developing MASLD therapeutics, creation of patient cohorts for clinical trials and optimization of therapeutic strategies for specific patient cohorts. Implementing pre-clinical experimental models for drug development creates a significant challenge as simple in vitro systems and animal models do not fully recapitulate critical steps in the pathogenesis and the complexity of MASLD progression. To address this, we implemented a precision medicine strategy that couples the use of our liver acinus microphysiology system (LAMPS) constructed with patient-derived primary cells. We investigated the MASLD-associated genetic variant PNPLA3 rs738409 (I148M variant) in primary hepatocytes, as it is associated with MASLD progression. We constructed LAMPS with genotyped wild type and variant PNPLA3 hepatocytes together with key non-parenchymal cells and quantified the reproducibility of the model. We altered media components to mimic blood chemistries, including insulin, glucose, free fatty acids, and immune activating molecules to reflect normal fasting (NF), early metabolic syndrome (EMS) and late metabolic syndrome (LMS) conditions. Finally, we investigated the response to treatment with resmetirom, an approved drug for metabolic syndrome-associated steatohepatitis (MASH), the progressive form of MASLD. This study using primary cells serves as a benchmark for studies using patient biomimetic twins constructed with patient iPSC-derived liver cells using a panel of reproducible metrics. We observed increased steatosis, immune activation, stellate cell activation and secretion of pro-fibrotic markers in the PNPLA3 GG variant compared to wild type CC LAMPS, consistent with the clinical characterization of this variant. We also observed greater resmetirom efficacy in PNPLA3 wild type CC LAMPS compared to the GG variant in multiple MASLD metrics including steatosis, stellate cell activation and the secretion of pro-fibrotic markers. In conclusion, our study demonstrates the capability of the LAMPS platform for the development of MASLD precision therapeutics, enrichment of patient cohorts for clinical trials, and optimization of therapeutic strategies for patient subgroups with different clinical traits and disease stages.
Collapse
|
23
|
Zyśk B, Ostrowska L, Smarkusz-Zarzecka J, Orywal K, Mroczko B, Cwalina U. Evaluation of the Diagnostic Utility of Selected Serum Adipokines and Cytokines in Subjects with MASLD-A Pilot Study. Nutrients 2024; 16:1381. [PMID: 38732626 PMCID: PMC11085733 DOI: 10.3390/nu16091381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Excess adipose tissue, particularly of the visceral type, triggering chronic low-grade inflammation and altering its secretory profile, is a contributing factor to the initiation and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). This study aimed to compare the levels of selected adipokines and cytokines in individuals with normal weight and obesity, assessing their potential for diagnosing MASLD and establishing a cutoff point for body fat content associated with hepatic steatosis development. The research involved 99 participants categorized by body mass index and MASLD presence, undergoing body composition analysis, liver elastography, biochemical tests, and evaluation of adipokines and cytokines in serum. The results indicated elevated IL-6 (interleukin 6) serum levels in individuals with obesity with MASLD compared to the normal-weight group without MASLD. The multivariate regression analysis demonstrated a connection between hepatic steatosis and total adipose tissue content, VAT (visceral adipose tissue), VAT/SAT (subcutaneous adipose tissue) ratio, HOMA-IR (homeostasis model assessment of insulin resistance), IL-6, Il-1β (interleukin 1β), and MMP-2 (matrix metalloproteinase 2). Among the adipokines and cytokines examined in this study, interleukin 6 was the strongest predictor of MASLD regardless of gender. In addition, an association between the development of hepatic steatosis and higher serum IL-1β levels and higher adipose tissue was observed in women. However, further studies on a larger group of patients are needed to consider the use of these cytokines as markers of MASLD. The HOMA-IR index demonstrated potential diagnostic utility in identifying hepatic steatosis.
Collapse
Affiliation(s)
- Beata Zyśk
- Department of Dietetics and Clinical Nutrition, Medical University of Bialystok, Mieszka I 4B Street, 15-054 Bialystok, Poland; (L.O.)
| | - Lucyna Ostrowska
- Department of Dietetics and Clinical Nutrition, Medical University of Bialystok, Mieszka I 4B Street, 15-054 Bialystok, Poland; (L.O.)
| | - Joanna Smarkusz-Zarzecka
- Department of Dietetics and Clinical Nutrition, Medical University of Bialystok, Mieszka I 4B Street, 15-054 Bialystok, Poland; (L.O.)
| | - Karolina Orywal
- Department of Biochemical Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland (B.M.)
| | - Barbara Mroczko
- Department of Biochemical Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland (B.M.)
| | - Urszula Cwalina
- Department of Biostatistics and Medical Informatics, Medical University of Bialystok, Szpitalna 37 Street, 15-295 Bialystok, Poland
| |
Collapse
|
24
|
Odanga JJ, Anderson SM, Breathwaite EK, Presnell SC, LeCluyse EL, Chen J, Weaver JR. Characterization of diseased primary human hepatocytes in an all-human cell-based triculture system. Sci Rep 2024; 14:6772. [PMID: 38514705 PMCID: PMC10957907 DOI: 10.1038/s41598-024-57463-7] [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: 11/11/2023] [Accepted: 03/18/2024] [Indexed: 03/23/2024] Open
Abstract
Liver diseases, including NAFLD, are a growing worldwide health concern. Currently, there is a lack of suitable in vitro models that sustain basic primary human hepatocyte (PHH) morphology and functionality while supporting presentation of disease-associated phenotypic characteristics such as lipid accumulation and inflammasome activation. In TruVivo, an all-human triculture system (hTCS), basic metabolic functions were characterized in PHHs isolated from normal or diseased livers during two-weeks of culture. Decreases in albumin and urea levels and CYP3A4 activity were seen in diseased-origin PHHs compared to normal PHHs along with higher CYP2E1 expression. Positive expression of the macrophage markers CD68 and CD163 were seen in the diseased PHH preparations. Elevated levels of the pro-inflammatory cytokines IL-6 and MCP-1 and the fibrotic markers CK-18 and TGF-β were also measured. Gene expression of FASN, PCK1, and G6PC in the diseased PHHs was decreased compared to the normal PHHs. Further characterization revealed differences in lipogenesis and accumulation of intracellular lipids in normal and diseased PHHs when cultured with oleic acid and high glucose. TruVivo represents a promising new platform to study lipogenic mechanisms in normal and diseased populations due to the preservation of phenotypic differences over a prolonged culture period.
Collapse
Affiliation(s)
- Justin J Odanga
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Sharon M Anderson
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Erick K Breathwaite
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Sharon C Presnell
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Edward L LeCluyse
- Research and Development, LifeNet Health LifeSciences, 6 Davis Dr., Research Triangle Park, NC, USA
| | - Jingsong Chen
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA
| | - Jessica R Weaver
- Institute of Regenerative Med., LifeNet Health, 1864 Concert Dr., Virginia Beach, VA, USA.
- LifeSciences Product Development, LifeNet Health, 1864 Concert Drive, Virginia Beach, VA, 23453, USA.
| |
Collapse
|
25
|
Hwang SJ, Choi YJ, Wang JH, Son CG. Lactobacillus Casei-fermented Amomum Xanthioides Mitigates non-alcoholic fatty liver disease in a high-fat diet mice model. Biomed Pharmacother 2024; 172:116250. [PMID: 38320334 DOI: 10.1016/j.biopha.2024.116250] [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: 12/11/2023] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/08/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a substantial global health issue owing to its high prevalence and the lack of effective therapies. Fermentation of medicinal herbs has always been considered a feasible strategy for enhancing efficacy in treating various ailments. This study aimed to investigate the potential benefits of the Lactobacillus casei-fermented Amomum xanthioides (LAX) on NAFLD in a high-fat diet model. HFD-fed C57BL6/j mice were administered with 200 mg/kg of LAX or unfermented Amomum xanthioides (AX) or 100 mg/kg of metformin for 6 weeks from the 4th week. The 10-week HFD-induced alterations of hepatic lipid accumulation and hepatic inflammation were significantly attenuated by LAX dominantly (more than AX or metformin), which evidenced by pathohistological findings, lipid contents, inflammatory cytokines including tumor necrosis factor (TNF)-α, interleukin (IL)- 6 and IL-1β, oxidative parameters such as reactive oxygen species (ROS) and malondialdehyde (MDA), and molecular changes reversely between lipogenic proteins such as glycerol-3-phosphate acyltransferase (GPAM) and sterol regulatory element-binding protein (SREBP)- 1, and lipolytic proteins including peroxisome proliferator-activated receptor (PPAR-α) and AMP-activated kinase (AMPK)-α in the liver tissues. In addition, the abnormal serum lipid parameters (triglyceride, total cholesterol and LDL-cholesterol) notably ameliorated by LAX. In conclusion, these findings support the potential of LAX as a promising plant-derived remedy for NAFLD.
Collapse
Affiliation(s)
- Seung-Ju Hwang
- Institute of Bioscience & Integrative Medicine, Daejeon University, 75, Daedukdae-ro 176 bun-gil, Seo-gu, Daejeon 35235, the Republic of Korea; Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, 75, Daedukdae-ro 176 bun-gil, Seo-gu, Daejeon 35235, the Republic of Korea
| | - Yu-Jin Choi
- Institute of Bioscience & Integrative Medicine, Daejeon University, 75, Daedukdae-ro 176 bun-gil, Seo-gu, Daejeon 35235, the Republic of Korea; Department of Internal Medicine, College of Korean Medicine, Se-Myung University, Semyeong-ro 65, Jecheon-si, Chungcheongbuk-do, 27136, the Republic of Korea
| | - Jing-Hua Wang
- Institute of Bioscience & Integrative Medicine, Daejeon University, 75, Daedukdae-ro 176 bun-gil, Seo-gu, Daejeon 35235, the Republic of Korea; Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, 75, Daedukdae-ro 176 bun-gil, Seo-gu, Daejeon 35235, the Republic of Korea.
| | - Chang-Gue Son
- Institute of Bioscience & Integrative Medicine, Daejeon University, 75, Daedukdae-ro 176 bun-gil, Seo-gu, Daejeon 35235, the Republic of Korea; Liver and Immunology Research Center, Daejeon Oriental Hospital of Daejeon University, 75, Daedukdae-ro 176 bun-gil, Seo-gu, Daejeon 35235, the Republic of Korea.
| |
Collapse
|
26
|
Pezzino S, Luca T, Castorina M, Puleo S, Latteri S, Castorina S. Role of Perturbated Hemostasis in MASLD and Its Correlation with Adipokines. Life (Basel) 2024; 14:93. [PMID: 38255708 PMCID: PMC10820028 DOI: 10.3390/life14010093] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) continues to rise, making it one of the most prevalent chronic liver disorders. MASLD encompasses a range of liver pathologies, from simple steatosis to metabolic dysfunction-associated steatohepatitis (MASH) with inflammation, hepatocyte damage, and fibrosis. Interestingly, the liver exhibits close intercommunication with fatty tissue. In fact, adipose tissue could contribute to the etiology and advancement of MASLD, acting as an endocrine organ that releases several hormones and cytokines, with the adipokines assuming a pivotal role. The levels of adipokines in the blood are altered in people with MASLD, and recent research has shed light on the crucial role played by adipokines in regulating energy expenditure, inflammation, and fibrosis in MASLD. However, MASLD disease is a multifaceted condition that affects various aspects of health beyond liver function, including its impact on hemostasis. The alterations in coagulation mechanisms and endothelial and platelet functions may play a role in the increased vulnerability and severity of MASLD. Therefore, more attention is being given to imbalanced adipokines as causative agents in causing disturbances in hemostasis in MASLD. Metabolic inflammation and hepatic injury are fundamental components of MASLD, and the interrelation between these biological components and the hemostasis pathway is delineated by reciprocal influences, as well as the induction of alterations. Adipokines have the potential to serve as the shared elements within this complex interrelationship. The objective of this review is to thoroughly examine the existing scientific knowledge on the impairment of hemostasis in MASLD and its connection with adipokines, with the aim of enhancing our comprehension of the disease.
Collapse
Affiliation(s)
- Salvatore Pezzino
- Mediterranean Foundation “GB Morgagni”, 95125 Catania, Italy (M.C.); (S.C.)
| | - Tonia Luca
- Mediterranean Foundation “GB Morgagni”, 95125 Catania, Italy (M.C.); (S.C.)
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | | | - Stefano Puleo
- Mediterranean Foundation “GB Morgagni”, 95125 Catania, Italy (M.C.); (S.C.)
| | - Saverio Latteri
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | - Sergio Castorina
- Mediterranean Foundation “GB Morgagni”, 95125 Catania, Italy (M.C.); (S.C.)
- Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
27
|
Khalaf SE, Abdelfattah SN, Khaliefa AK, Daoud SA, Yahia E, Hasona NA. Expression of PVT-1 and miR-29a/29b as reliable biomarkers for liver cirrhosis and their correlation with the inflammatory biomarkers profile. Hum Exp Toxicol 2024; 43:9603271241251451. [PMID: 38685136 DOI: 10.1177/09603271241251451] [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] [Indexed: 05/02/2024]
Abstract
BACKGROUND & AIMS The liver is a vital organ responsible for numerous metabolic processes, which can be significantly impacted by long non-coding RNAs (lncRNAs) and microRNAs (miRNAs). These ribonucleic acid (RNA) molecules have been shown to play a crucial role in regulating gene expression, and their dysregulation has been implicated in numerous liver disorders. Our study aimed to investigate the diagnostic accuracy of plasmacytoma variant translocation-1 (PVT-1), microRNA-29a/29b (miR-29a/miR-29b), and inflammatory biomarkers [ interleukine-6 (IL-6), tumor necrosis factor-alpha (TNF-α), transforming growth factor-beta (TGF-β), and insulin growth factor-1 (IGF-1)] as diagnostic and prognostic biomarkers for liver cirrhosis. Therefore, understanding the mechanisms by which lncRNAs and miRNAs influence liver metabolism is of paramount importance in developing effective treatments for liver-related diseases. METHODS Serum samples were collected from 164 participants, comprising 114 cirrhotic patients with varying grades (35 grade I, 35 grade II, and 44 grade III) and 50 healthy controls. PVT-1 and miR-29a/miR-29b expression was analyzed by reverse transcription-quantitative polymerase chain reaction (RT-PCR), while the serum levels of inflammatory biomarkers were assessed using enzyme-linked immunosorbent assay (ELISA). RESULTS The study participants exhibited notable differences in PVT-1 and miR-29a/miR-29b expression. ROC analysis revealed excellent discriminative power for PVT-1 and miR-29a/miR-29b in distinguishing cirrhotic patients from healthy controls. CONCLUSION This study demonstrates the promising potential of PVT-1 and miR-29a/miR-29b as early diagnostic biomarkers for liver cirrhosis detection, requiring further validation in larger cohorts. Our findings also reinforce the diagnostic value of circulating inflammatory biomarkers (IL-6, TNF-α, TGF-β, and IGF-1) levels for liver cirrhosis screening.
Collapse
Affiliation(s)
- Shaza E Khalaf
- Department of Biochemistry, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | | | - Amal K Khaliefa
- Department of Biochemistry, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Sahar A Daoud
- Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
- Faculty of Medicine, Beni Suef National University, Beni-Suef, Egypt
| | - Enas Yahia
- Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
- Faculty of Medicine, Beni Suef National University, Beni-Suef, Egypt
| | - Nabil A Hasona
- Department of Biochemistry, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
- Biochemistry Department, Faculty of Science, Beni Suef National University, Beni-Suef, Egypt
| |
Collapse
|
28
|
Vrsaljko N, Gjurašin B, Papić N. COVID-19 severity and nonalcoholic fatty liver disease. MANAGEMENT, BODY SYSTEMS, AND CASE STUDIES IN COVID-19 2024:457-463. [DOI: 10.1016/b978-0-443-18703-2.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Kumari D, Gautam J, Sharma V, Gupta SK, Sarkar S, Jana P, Singhal V, Babele P, Kamboj P, Bajpai S, Tandon R, Kumar Y, Dikshit M. Effect of herbal extracts and Saroglitazar on high-fat diet-induced obesity, insulin resistance, dyslipidemia, and hepatic lipidome in C57BL/6J mice. Heliyon 2023; 9:e22051. [PMID: 38027691 PMCID: PMC10663915 DOI: 10.1016/j.heliyon.2023.e22051] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
We evaluated the effects of select herbal extracts (Tinospora cordifolia [TC], Tinospora cordifolia with Piper longum [TC + PL], Withania somnifera [WS], Glycyrrhiza glabra [GG], AYUSH-64 [AY-64], and Saroglitazar [S]) on various parameters in a diet-induced obesity mouse model. After 12 weeks of oral administration of the herbal extracts in high-fat diet (HFD)-fed C57BL/6J mice, we analyzed plasma biochemical parameters, insulin resistance (IR), liver histology, and the expression of inflammatory and fibrosis markers, along with hepatic lipidome. We also used a 3D hepatic spheroid model to assess their impact on profibrotic gene expression. Among the extracts, TC + PL showed a significant reduction in IR, liver weight, TNF-α, IL4, IL10 expression, and hepatic lipid levels (saturated triglycerides, ceramides, lysophosphocholines, acylcarnitines, diglycerides, and phosphatidylinositol levels). Saroglitazar reversed changes in body weight, IR, plasma triglycerides, glucose, insulin, and various hepatic lipid species (fatty acids, phospholipids, glycerophospholipids, sphingolipids, and triglycerides). With the exception of GG, Saroglitazar, and other extracts protected against palmitic acid-induced fibrosis marker gene expression in the 3D spheroids. TC + PL and Saroglitazar also effectively prevented HFD-induced insulin resistance, inflammation, and specific harmful lipid species in the liver.
Collapse
Affiliation(s)
- Deepika Kumari
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Jyoti Gautam
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Vipin Sharma
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Sonu Kumar Gupta
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Soumalya Sarkar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Pradipta Jana
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Vikas Singhal
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Prabhakar Babele
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Parul Kamboj
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Sneh Bajpai
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | | | - Yashwant Kumar
- Non-communicable Disease Centre, Translational Health Science and Technology Institute (THSTI), NCR Biotech Science Cluster, 3rd Milestone, Faridabad, 121001, Haryana, India
| | - Madhu Dikshit
- Central Drug Research Institute, Sitapur Rd, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India.
| |
Collapse
|
30
|
Pratim Das P, Medhi S. Role of inflammasomes and cytokines in immune dysfunction of liver cirrhosis. Cytokine 2023; 170:156347. [PMID: 37639845 DOI: 10.1016/j.cyto.2023.156347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/28/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Liver cirrhosis develops as a result of persistent inflammation and liver injury. The prolonged inflammation triggers the buildup of fibrous tissue and regenerative nodules within the liver, leading to the distortion of the hepatic vascular structure and impaired liver function. Cirrhosis disrupts the ability of liver function to maintain homeostasis and hepatic immunosurveillance which causes immunological dysfunction in the body. In pathological conditions, the production of cytokines in the liver is carefully regulated by various cells in response to tissue stimulation. Cytokines and inflammasomes are the key regulators and systematically contribute to the development of cirrhosis which involves an inflammatory response. However, the crosstalk role of different cytokines in the cirrhosis progression is poorly understood. Tumour necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), interleukin-6 (IL-6), and interferon-gamma (IFN-γ), among others, are proinflammatory cytokines that contribute to liver cell necrosis, which in turn causes the development of fibrosis. While IL-10 exhibits a potent anti-inflammatory effect on the liver by inhibiting immune cell activation and neutralizing pro-inflammatory cytokine activity. Inflammasomes have also been implicated in the profibrotic processes of liver cirrhosis, as well as the production of chemokines such as CCL2/MCP-1. It is evident that inflammasomes have a role in the proinflammatory response seen in chronic liver illnesses. In conclusion, cirrhosis significantly impacts the immune system, leading to immunological dysfunction and alterations in both innate and acquired immunity. Proinflammatory cytokines like TNF-α, IL-1β, IL-6, and IFNγ are upregulated in cirrhosis, contributing to liver cell necrosis and fibrosis development. Managing cytokine-mediated inflammation and fibrosis is a key therapeutic approach to alleviate portal hypertension and its associated liver complications. This review attempted to focus largely on the role of immune dysfunction mediated by different cytokines and inflammasomes involved in the progression, regulation and development of liver cirrhosis.
Collapse
Affiliation(s)
- Partha Pratim Das
- Dept. of Bioengineering & Technology, Gauhati University, Assam 781014, India
| | - Subhash Medhi
- Dept. of Bioengineering & Technology, Gauhati University, Assam 781014, India.
| |
Collapse
|
31
|
Xu Q, Feng M, Ren Y, Liu X, Gao H, Li Z, Su X, Wang Q, Wang Y. From NAFLD to HCC: Advances in noninvasive diagnosis. Biomed Pharmacother 2023; 165:115028. [PMID: 37331252 DOI: 10.1016/j.biopha.2023.115028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has gradually become one of the major liver health problems in the world. The dynamic course of the disease goes through steatosis, inflammation, fibrosis, and carcinoma. Before progressing to carcinoma, timely and effective intervention will make the condition better, which highlights the importance of early diagnosis. With the further study of the biological mechanism in the pathogenesis and progression of NAFLD, some potential biomarkers have been discovered, and the possibility of their clinical application is gradually being discussed. At the same time, the progress of imaging technology and the emergence of new materials and methods also provide more possibilities for the diagnosis of NAFLD. This article reviews the diagnostic markers and advanced diagnostic methods of NAFLD in recent years.
Collapse
Affiliation(s)
- Qinchen Xu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Maoxiao Feng
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Yidan Ren
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Xiaoyan Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Huiru Gao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Zigan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Xin Su
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Qin Wang
- Department of Anesthesiology, Qilu Hospital, Shandong University, 107 Wenhua Xi Road, Jinan 250012, China.
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China.
| |
Collapse
|
32
|
Vachliotis ID, Polyzos SA. The Role of Tumor Necrosis Factor-Alpha in the Pathogenesis and Treatment of Nonalcoholic Fatty Liver Disease. Curr Obes Rep 2023; 12:191-206. [PMID: 37407724 PMCID: PMC10482776 DOI: 10.1007/s13679-023-00519-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 07/07/2023]
Abstract
PURPOSE OF REVIEW To summarize experimental and clinical evidence on the association between tumor necrosis factor-α (TNF-α) and nonalcoholic fatty liver disease (NAFLD) and discuss potential treatment considerations. RECENT FINDINGS Experimental evidence suggests that TNF-α is a cytokine with a critical role in the pathogenesis of NAFLD. Although, the production of TNF-α may be an early event during the course of nonalcoholic fatty liver (NAFL), TNF-α may play a more substantial role in the pathogenesis of nonalcoholic steatohepatitis (NASH) and NAFLD-associated fibrosis. Moreover, TNF-α may potentiate hepatic insulin resistance, thus interconnecting inflammatory with metabolic signals and possibly contributing to the development of NAFLD-related comorbidities, including cardiovascular disease, hepatocellular carcinoma, and extra-hepatic malignancies. In clinical terms, TNF-α is probably associated with the severity of NAFLD; circulating TNF-α gradually increases from controls to patients with NAFL, and then, to patients with NASH. Given this potential association, various therapeutic interventions (obeticholic acid, peroxisome proliferator-activated receptors, sodium-glucose co-transporter 2 inhibitors, glucagon-like peptide-1 receptor agonists, probiotics, synbiotics, rifaximin, vitamin E, pentoxifylline, ursodeoxycholic acid, fibroblast growth factor-21, n-3 polyunsaturated fatty acids, statins, angiotensin receptor blockers) have been evaluated for their effect on TNF-α and NAFLD. Interestingly, anti-TNF biologics have shown favorable metabolic and hepatic effects, which may open a possible therapeutic window for the management of advanced NAFLD. The potential key pathogenic role of TNF-α in NAFLD warrants further investigation and may have important diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Ilias D. Vachliotis
- First Department of Pharmacology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Second Department of Internal Medicine, 424 General Military Hospital, Thessaloniki, Greece
| | - Stergios A. Polyzos
- First Department of Pharmacology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
33
|
Jaiswal V, Ang SP, Huang H, Momi NK, Hameed M, Naz S, Batra N, Ishak A, Doshi N, Gera A, Sharath M, Waleed MS, Raj N, Aguilera Alvarez VH. Association between nonalcoholic fatty liver disease and atrial fibrillation and other clinical outcomes: a meta-analysis. J Investig Med 2023; 71:591-602. [PMID: 37002665 DOI: 10.1177/10815589231164777] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
The association between nonalcoholic fatty liver disease (NAFLD) with cardiovascular and cerebrovascular outcomes, as well as their clinical impact, has yet to be established in the literature. This meta-analysis aims to evaluate the association between the NAFLD patients and the risk of atrial fibrillation (AF), heart failure (HF), stroke, cardiovascular mortality (CVM), and revascularization incidence. We performed a systematic literature search using PubMed, Embase, Scopus, and Cochrane libraries for relevant articles from inception until August 2022. A total of 12 cohort studies with 18,055,072 patients (2,938,753 NAFLD vs 15,116,319 non-NAFLD) were included in our analysis. The mean age of the NAFLD patients group and the non-NAFLD group was comparable (55.68 vs 55.87). The most common comorbidities among the NAFLD patients group included hypertension (38% vs 24%) and diabetes mellitus (14% vs 8%). The mean follow-up duration was 6.26 years. The likelihood of AF (risk ratio (RR), 1.42 (95% CI 1.19, 1.68), p < 0.001), HF (RR, 1.43(95% CI 1.03, 2.00), p < 0.001), stroke (RR, 1.26(95% CI 1.16, 1.36), p < 0.001), revascularization (RR, 4.06(95% CI 1.44, 11.46), p = 0.01), and CVM (RR, 3.10(95% CI 1.43, 6.73), p < 0.001) was significantly higher in the NAFLD patients group compared to that of the non-NAFLD group. However, all-cause mortality was comparable between both the groups of patients (RR, 1.30 (95% CI 0.63, 2.67), p = 0.48). In conclusion, the patients with NAFLD are at increased risk of AF, HF, and CVM.
Collapse
Affiliation(s)
- Vikash Jaiswal
- JCCR Cardiology, Varanasi, Uttar Pradesh, India
- Department of Research and Academic Affairs, Larkin Community Hospital, South Miami, Florida, USA
| | - Song Peng Ang
- Division of Internal Medicine, Rutgers Health/Community Medical Center, Toms River, NJ, USA
| | - Helen Huang
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Maha Hameed
- Department of Research and Academic Affairs, Larkin Community Hospital, South Miami, Florida, USA
| | - Sidra Naz
- The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Nitya Batra
- Department of Internal Medicine, Beaumont Hospital, Royal Oak, MI, USA
| | - Angela Ishak
- Department of Research and Academic Affairs, Larkin Community Hospital, South Miami, Florida, USA
| | - Neel Doshi
- Department of Medicine, Pravara Institute of Medical Science, Ahmednagar, Maharashtra, India
| | - Asmita Gera
- Department of Research and Academic Affairs, Larkin Community Hospital, South Miami, Florida, USA
| | - Medha Sharath
- Bangalore Medical College and Research Institute, Bengaluru, Karnataka, India
| | | | - Nishchita Raj
- JCCR Cardiology, Varanasi, Uttar Pradesh, India
- B.P Koirala Institute of Health Science, Dharan, Nepal
| | | |
Collapse
|
34
|
Park SJ, Garcia Diaz J, Um E, Hahn YS. Major roles of kupffer cells and macrophages in NAFLD development. Front Endocrinol (Lausanne) 2023; 14:1150118. [PMID: 37274349 PMCID: PMC10235620 DOI: 10.3389/fendo.2023.1150118] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an important public health problem with growing numbers of NAFLD patients worldwide. Pathological conditions are different in each stage of NAFLD due to various factors. Preclinical and clinical studies provide evidence for a crucial role of immune cells in NAFLD progression. Liver-resident macrophages, kupffer cells (KCs), and monocytes-derived macrophages are the key cell types involved in the progression of NAFLD, non-alcoholic steatohepatitis (NASH), and hepatocellular carcinoma (HCC). Their unique polarization contributes to the progression of NAFLD. KCs are phagocytes with self-renewal abilities and play a role in regulating and maintaining homeostasis. Upon liver damage, KCs are activated and colonized at the site of the damaged tissue. The secretion of inflammatory cytokines and chemokines by KCs play a pivotal role in initiating NAFLD pathogenesis. This review briefly describes the role of immune cells in the immune system in NAFLD, and focuses on the pathological role and molecular pathways of KCs and recruited macrophages. In addition, the relationship between macrophages and insulin resistance is described. Finally, the latest therapeutics that target KCs and macrophages are summarized for the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Soo-Jeung Park
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Josefina Garcia Diaz
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Eugene Um
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
| | - Young S. Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
35
|
Amorim R, Magalhães CC, Borges F, Oliveira PJ, Teixeira J. From Non-Alcoholic Fatty Liver to Hepatocellular Carcinoma: A Story of (Mal)Adapted Mitochondria. BIOLOGY 2023; 12:biology12040595. [PMID: 37106795 PMCID: PMC10135755 DOI: 10.3390/biology12040595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/30/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a global pandemic affecting 25% of the world's population and is a serious health and economic concern worldwide. NAFLD is mainly the result of unhealthy dietary habits combined with sedentary lifestyle, although some genetic contributions to NAFLD have been documented. NAFLD is characterized by the excessive accumulation of triglycerides (TGs) in hepatocytes and encompasses a spectrum of chronic liver abnormalities, ranging from simple steatosis (NAFL) to steatohepatitis (NASH), significant liver fibrosis, cirrhosis, and hepatocellular carcinoma. Although the molecular mechanisms that cause the progression of steatosis to severe liver damage are not fully understood, metabolic-dysfunction-associated fatty liver disease is strong evidence that mitochondrial dysfunction plays a significant role in the development and progression of NAFLD. Mitochondria are highly dynamic organelles that undergo functional and structural adaptations to meet the metabolic requirements of the cell. Alterations in nutrient availability or cellular energy needs can modify mitochondria formation through biogenesis or the opposite processes of fission and fusion and fragmentation. In NAFL, simple steatosis can be seen as an adaptive response to storing lipotoxic free fatty acids (FFAs) as inert TGs due to chronic perturbation in lipid metabolism and lipotoxic insults. However, when liver hepatocytes' adaptive mechanisms are overburdened, lipotoxicity occurs, contributing to reactive oxygen species (ROS) formation, mitochondrial dysfunction, and endoplasmic reticulum (ER) stress. Impaired mitochondrial fatty acid oxidation, reduction in mitochondrial quality, and disrupted mitochondrial function are associated with a decrease in the energy levels and impaired redox balance and negatively affect mitochondria hepatocyte tolerance towards damaging hits. However, the sequence of events underlying mitochondrial failure from steatosis to hepatocarcinoma is still yet to be fully clarified. This review provides an overview of our understanding of mitochondrial adaptation in initial NAFLD stages and highlights how hepatic mitochondrial dysfunction and heterogeneity contribute to disease pathophysiology progression, from steatosis to hepatocellular carcinoma. Improving our understanding of different aspects of hepatocytes' mitochondrial physiology in the context of disease development and progression is crucial to improving diagnosis, management, and therapy of NAFLD/NASH.
Collapse
Affiliation(s)
- Ricardo Amorim
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Carina C Magalhães
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Fernanda Borges
- CIQUP-IMS/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, 4169-007 Porto, Portugal
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - José Teixeira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
36
|
Bertran L, Adalid L, Vilaró-Blay M, Barrientos-Riosalido A, Aguilar C, Martínez S, Sabench F, del Castillo D, Porras JA, Alibalic A, Richart C, Auguet T. Expression of STING in Women with Morbid Obesity and Nonalcoholic Fatty Liver Disease. Metabolites 2023; 13:metabo13040496. [PMID: 37110154 PMCID: PMC10146769 DOI: 10.3390/metabo13040496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic hepatic disease. Although mostly benign, this disease can evolve into nonalcoholic steatohepatitis (NASH). The stimulator of interferon genes (STING) plays an important role in the immune response against stressed cells, but this protein may also be involved in liver lipogenesis and microbiota composition. In this study, the role of STING in NAFLD was evaluated by RT–qPCR to analyze STING mRNA abundance and by immunohistochemical analysis to evaluate protein expression in liver biopsies from a cohort composed of 69 women with morbid obesity classified according to their liver involvement (normal liver, n = 27; simple steatosis (SS), n = 26; NASH, n = 16). The results showed that STING mRNA expression in the liver increases with the occurrence of NAFLD, specifically in the SS stage in which the degree of steatosis is mild or moderate. Protein analysis corroborated these results. Positive correlations were observed among hepatic STING mRNA abundance and gamma-glutamyl transferase and alkaline phosphatase levels, hepatic Toll-like receptor 9 expression and some circulating microbiota-derived bile acids. In conclusion, STING may be involved in the outcome and progression of NAFLD and may be related to hepatic lipid metabolism. However, further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Laia Bertran
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Laia Adalid
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona, Mallafré Guasch, 4, 43007 Tarragona, Spain
| | - Mercè Vilaró-Blay
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Andrea Barrientos-Riosalido
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Carmen Aguilar
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Salomé Martínez
- Servei Anatomia Patològica, Hospital Universitari Joan XXIII Tarragona, Mallafré Guasch, 4, 43007 Tarragona, Spain
| | - Fàtima Sabench
- Servei de Cirurgia i Anestèsia, Hospital Sant Joan de Reus, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204 Reus, Spain
| | - Daniel del Castillo
- Servei de Cirurgia i Anestèsia, Hospital Sant Joan de Reus, Departament de Medicina i Cirurgia, Universitat Rovira i Virgili (URV), IISPV, Avinguda Doctor Josep Laporte, 2, 43204 Reus, Spain
| | - José Antonio Porras
- Servei de Medicina Interna, Hospital Universitari Joan XXIII Tarragona, Mallafré Guash, 4, 43007 Tarragona, Spain
| | - Ajla Alibalic
- Servei de Medicina Interna, Hospital Universitari Joan XXIII Tarragona, Mallafré Guash, 4, 43007 Tarragona, Spain
| | - Cristóbal Richart
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
| | - Teresa Auguet
- Grup de Recerca GEMMAIR (AGAUR)—Medicina Aplicada (URV), Departament de Medicina i Cirurgia, Institut d’Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), 43007 Tarragona, Spain
- Servei de Medicina Interna, Hospital Universitari Joan XXIII Tarragona, Mallafré Guash, 4, 43007 Tarragona, Spain
- Correspondence: ; Tel.: +34-977-29-58-33
| |
Collapse
|
37
|
Lu B, Wang D, Xie D, Wu C, Sun M. 20(S)-Protopanaxatriol ameliorates MAFLD by inhibiting NLRP3 inflammasome. Eur J Pharmacol 2023; 940:175468. [PMID: 36566009 DOI: 10.1016/j.ejphar.2022.175468] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/03/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Metabolic associated fatty liver disease (MAFLD) is one of the most common chronic liver diseases and may develop into non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and even hepatocellular carcinoma, which has threatened human health. Although NLRP3 inflammasome is widely recognized in the pathogenesis of MAFLD, there are currently no drugs targeting NLRP3 inflammasome approved by regulatory agencies. Panax ginseng and its main saponin components have been used to regulate inflammatory and metabolic disorders. Notably, 20(S)-protopanaxatriol (PPT) is an active metabolite of protopanaxatriol saponins with prominent anti-inflammatory activity. However, the mechanism by which PPT ameliorates MAFLD has not been fully elucidated. Therefore, this study explored the efficacy and mechanism of PPT in treating MAFLD based on the inhibition of NLRP3 inflammasome activation. First, we screened potential NLRP3 inflammasome blockers from protopanaxadiol saponins in mouse primary bone marrow-derived macrophages (BMDMs) stimulated by LPS and different inflammasome inducers. Second, LPS-primed mouse BMDMs, mouse primary hepatocytes, mouse primary Kupffer cells and human peripheral blood mononuclear cells (PBMCs) stimulated by cholesterol and ATP were used to evaluate the effect of PPT in inhibiting NLRP3 inflammasome. Finally, MCD-induced mouse MAFLD were established to verify the therapeutic effect of PPT by inhibiting NLRP3 inflammasome. Our results showed that PPT of ginseng saponins significantly inhibited NLRP3 inflammasome activation in multiple primary cells, suppressed systemic inflammation, restored liver function, and attenuated liver inflammation as well as fibrosis in MCD--induced mouse MAFLD. Collectively, protopanaxatriol saponins metabolite PPT, may serve as a potent therapeutic agent for MAFLD by inhibiting NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Bingjie Lu
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Dan Wang
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Dong Xie
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Chao Wu
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Mingyu Sun
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
38
|
Fotschki B, Ognik K, Fotschki J, Napiórkowska D, Cholewińska E, Krauze M, Juśkiewicz J. Chromium Nanoparticles Together with a Switch Away from High-Fat/Low-Fiber Dietary Habits Enhances the Pro-Healthy Regulation of Liver Lipid Metabolism and Inflammation in Obese Rats. Int J Mol Sci 2023; 24:ijms24032940. [PMID: 36769261 PMCID: PMC9918060 DOI: 10.3390/ijms24032940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
The study on Wistar rats was conducted to investigate the effects of a pharmacologically relevant dose 0.3 mg/kg body weight of chromium supplementation (commonly used picolinate or novel form as nanoparticles) and switching away from obesogenic dietary habits on the parameters of lipid metabolism, inflammation, and oxidative stress in liver and plasma. Favorable effects related to dietary changes from the obesogenic diet were considerably enhanced when the diet was supplemented with chromium nanoparticles. This combination exerted the strongest fat content and cholesterol reduction in the liver. Moreover, in this group, a favorable antioxidative effect was observed through GSH/GSSG elevation in the liver as well as ALT activity reduction in the plasma and IL-6 levels in the liver. The molecular mechanisms associated with regulating lipid metabolism, oxidative stress and inflammation might be related to lower expression of HIF-1α, COX-2, and LOX-1 and upregulation of PPARα in the liver. Supplementation with chromium nanoparticles without changes in the obesogenic diet also favorably affected lipid metabolism and oxidative stress in the liver; however, the examined effects were moderate. In conclusion, the favorable effects of switching from an obesogenic to a balanced diet on hepatic lipid metabolism, oxidative stress, and inflammation induced by an obesogenic diet might be enhanced by supplementation with chromium nanoparticles.
Collapse
Affiliation(s)
- Bartosz Fotschki
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10 Tuwima Street, 10-718 Olsztyn, Poland
| | - Katarzyna Ognik
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, 13 Akademicka Street, 20-950 Lublin, Poland
| | - Joanna Fotschki
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10 Tuwima Street, 10-718 Olsztyn, Poland
| | - Dorota Napiórkowska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10 Tuwima Street, 10-718 Olsztyn, Poland
| | - Ewelina Cholewińska
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, 13 Akademicka Street, 20-950 Lublin, Poland
| | - Magdalena Krauze
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, 13 Akademicka Street, 20-950 Lublin, Poland
| | - Jerzy Juśkiewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10 Tuwima Street, 10-718 Olsztyn, Poland
- Correspondence:
| |
Collapse
|
39
|
gp130 Activates Mitochondrial Dynamics for Hepatocyte Survival in a Model of Steatohepatitis. Biomedicines 2023; 11:biomedicines11020396. [PMID: 36830933 PMCID: PMC9953457 DOI: 10.3390/biomedicines11020396] [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: 12/22/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
Obesity is the main cause of metabolic complications. Fatty liver infiltration is a companion of obesity. NAFLD is associated with impaired energy metabolism with an excess of nutrients. Mitochondrial dynamics are important for the regulation of energy balance, which regulates mitochondrial function, apoptosis, and mitophagy. The aim of this study was to investigate the effect of gp130 on the components of mitochondrial dynamics in a cellular model of steatohepatitis. Addition of IL-6/gp130 contributed to an increase in the percentage of live cells and a decrease in the percentage of dead and apoptotic cells. Addition of IL-6/gp130 increased the expression of NF-kB1 gene and mitochondrial dynamics markers (MFN2 and TFAM) in HepG2 with tBHP/Oleic. Addition of IL-6 or gp130 reduced the expression of cytoprotector genes (HSF1 and HSP70) in HepG2 cell cultures with tBHP/Oleic. Increased mitochondrial dynamics gene activity protected against HepG2 cell death in the steatohepatitis model. Trans-signaling resulted in increased TFAM and MAPLC3B, and decreased DNM1L gene expression in HepG2 with tBHP/Oleic.
Collapse
|
40
|
Overview of Cellular and Soluble Mediators in Systemic Inflammation Associated with Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2023; 24:ijms24032313. [PMID: 36768637 PMCID: PMC9916753 DOI: 10.3390/ijms24032313] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently the most prevalent chronic liver disease in Western countries, affecting approximately 25% of the adult population. This condition encompasses a spectrum of liver diseases characterized by abnormal accumulation of fat in liver tissue (non-alcoholic fatty liver, NAFL) that can progress to non-alcoholic steatohepatitis (NASH), characterized by the presence of liver inflammation and damage. The latter form often coexists with liver fibrosis which, in turn, may progress to a state of cirrhosis and, potentially, hepatocarcinoma, both irreversible processes that often lead to the patient's death and/or the need for liver transplantation. Along with the high associated economic burden, the high mortality rate among NAFLD patients raises interest, not only in the search for novel therapeutic approaches, but also in early diagnosis and prevention to reduce the incidence of NAFLD-related complications. In this line, an exhaustive characterization of the immune status of patients with NAFLD is mandatory. Herein, we attempted to gather and compare the current and relevant scientific evidence on this matter, mainly on human reports. We addressed the current knowledge related to circulating cellular and soluble mediators, particularly platelets, different leukocyte subsets and relevant inflammatory soluble mediators.
Collapse
|
41
|
Wang D, Wang Q, Zuo Z, Dong Z, He J, Ye X, Tang H, Zou J. Koumine induces apoptosis in Cyprinus carpio liver cells by regulating JAK-STAT and p53 signaling pathways. FISH & SHELLFISH IMMUNOLOGY 2023; 132:108475. [PMID: 36496140 DOI: 10.1016/j.fsi.2022.108475] [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: 05/14/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
Koumine is an alkaloid with significant anti-anxiety, anticancer cell proliferation, and analgesic activities, and our previous studies have shown that koumine can be used as an immunostimulant in aquaculture, but the molecular mechanism of its effect remains unclear. We fed a basal diet with 0, 0.2, 2, and 20 mg/kg koumine to C. carpio for 10 weeks, and comprehensive studies of the histological and biochemical parameters and transcriptomes of the four groups were performed. Histological results indicated that the number of apoptotic cells in the liver increased with increasing koumine concentration. Compared with those of the control group, the malondialdehyde, superoxide dismutase, catalase, acid phosphatase, alkaline phosphatase, and lactate dehydrogenase levels of the treatment group increased to varying degrees. In total, 100.11 GB of clean data, 4774 DEGs, and 138 differentially expressed genes were obtained from the transcriptome data. Differentially expressed genes were classified into 187 signalling pathways, and the circadian rhythm signalling pathway, the JAK-STAT signalling pathway, the p53 signalling pathway and the PPAR signalling pathway were the top enriched pathways. The qRT-PCR results confirmed that the key genes ifnar1, socs3l, epoa, ghra, cMyc, mcl-1, shisa4, and gtse1 involved in balancing cell proliferation and apoptosis were enriched in these pathways. We discovered that the JAK-STAT and p53 pathways are important targets of koumine. Such information contributes to a better understanding of the potential mechanism by which koumine regulates hepatic immunity as well as to lays the theoretical foundation for its application.
Collapse
Affiliation(s)
- Dongjie Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qiujie Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Zhiheng Zuo
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Zaijie Dong
- Key Laboratory of Freshwater Fisheries and Germplasm Resources Utilization, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi, 214081, China.
| | - Jiayang He
- College of Environmental Science and Engineering, Guilin University of Technology, Guilin, 541004, China
| | - Xiangchen Ye
- Aquatic Species Introduction and Breeding Centre of Guangxi Zhuang Autonomous Region, Nanning, 530000, China
| | - Huijuan Tang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jixing Zou
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
42
|
Demiröz Taşolar S, Çiftçi N. Role of pan immune inflammatory value in the evaluation of hepatosteatosis in children and adolescents with obesity. J Pediatr Endocrinol Metab 2022; 35:1481-1486. [PMID: 36284505 DOI: 10.1515/jpem-2022-0494] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/07/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Inflammation is a feature of non-alcoholic fatty liver disease progression and plays an important role in hepatic steatosis and fibrosis. Since there are no studies in the literature showing the relationship between hepatosteatosis with the systemic immune-inflammation index (SII) and pan-immune inflammation value (PIV), we aimed to evaluate the relationship between these biomarkers and hepatosteatosis in childhood. METHODS We included 133 consecutive obese children and adolescents aged 6-18 years into this single-center, retrospective, and cross-sectional study. Anthropometric, physical examination, radiological and laboratory data were obtained and recorded from the file records of each case. RESULTS When we grouped the patient population according to the grade of hepatosteatosis, there was a statistically significant difference between the groups in terms of SII and PIV values (p<0.05, for both). In the analyzes performed to identify independent predictors of hepatosteatosis pubertal status (p=0.019) and PIV value (p<0.001) were found to be significant as independent predictors. Moreover, in the analysis performed to predict severity of hepatic steatosis, regression analysis was performed by dividing the groups into groups with and without severe adiposity. As a result of this analysis, HOMA-IR (p=0.019) and PIV value (p=0.028) were found to be significant in the prediction of severe hepatic adiposity. CONCLUSIONS Our findings showed that increased PIV levels were associated with the presence and severity of hepatic steatosis, but not with SII.
Collapse
Affiliation(s)
- Sevgi Demiröz Taşolar
- Department of Pediatric Radiology, Malatya Training and Research Hospital, Malatya, Turkiye
| | - Nurdan Çiftçi
- Department of Pediatric Endocrinology, Malatya Training and Research Hospital, Malatya, Turkiye
| |
Collapse
|
43
|
Qiu T, Hu W, Rao Z, Fang T. The molecular basis of the associations between non-alcoholic fatty liver disease and colorectal cancer. Front Genet 2022; 13:1007337. [PMID: 36568397 PMCID: PMC9780501 DOI: 10.3389/fgene.2022.1007337] [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: 07/30/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022] Open
Abstract
Background: Given the ongoing research on non-alcoholic fatty liver disease (NAFLD) and colorectal cancer (CRC), the number of studies suggesting a strong link between NAFLD and CRC is on the rise, while its underlying pathological mechanisms remain uncertain. This study aims to explore the shared genes and mechanisms and to reveal the molecular basis of the association between CRC and NAFLD through bioinformatics approaches. Methods: The Gene Expression Omnibus (GEO) dataset GSE89632 is downloaded for NAFLD cases and healthy controls. Additionally, the GSE4107 and GSE9348 datasets are obtained for CRC cases and healthy controls. Differentially expressed genes (DEGs) are obtained for NAFLD and CRC datasets, as well as shared genes between the two disorders. GO and KEGG enrichment analyses are further conducted. Subsequently, the STRING database and Cytoscape software are utilized to establish the PPI network and identify the hub genes. Then, co-expression analysis is performed using GeneMANIA. Subsequently, ROC curves and external datasets validation were applied to further screen the candidate markers. Finally, NetworkAnalyst is available as a means to construct a miRNA-gene regulatory network. Results: Under the threshold of FDR ≤ 0.01, 147 common genes are obtained in NAFLD and CRC. Categorization of GO functions shows that DEGs are predominantly enriched in "response to organic substance", "cellular response to chemical stimulus", and "response to external stimulus". The predominant KEGG pathways in DEGs are the "IL-17 signaling pathway", the "TNF signaling pathway", "Viral protein interaction with cytokine and cytokine receptor", "Cytokine-cytokine receptor interaction", and the "Toll-like receptor signaling pathway". Additionally, MYC, IL1B, FOS, CXCL8, PTGS2, MMP9, JUN, and IL6 are identified as hub genes by the evaluation of 7 algorithms. With the construction of miRNA-gene networks, 2 miRNAs, including miR-106a-5p, and miR-204-5p are predicted to be potential key miRNAs. Conclusion: This study identifies possible hub genes acting in the co-morbidity of NAFLD and CRC and discovers the interaction of miRNAs and hub genes, providing a novel understanding of the molecular basis for the relevance of CRC and NAFLD, thus contributing to the development of new therapeutic strategies to combat NAFLD and CRC.
Collapse
Affiliation(s)
- Ting Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Weitao Hu
- Department of Rheumatology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zilan Rao
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Taiyong Fang
- Department of Gastroenterology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China,*Correspondence: Taiyong Fang,
| |
Collapse
|
44
|
Monirujjaman M, Bathe OF, Mazurak VC. Dietary EPA+DHA Mitigate Hepatic Toxicity and Modify the Oxylipin Profile in an Animal Model of Colorectal Cancer Treated with Chemotherapy. Cancers (Basel) 2022; 14:cancers14225703. [PMID: 36428795 PMCID: PMC9688617 DOI: 10.3390/cancers14225703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Irinotecan (CPT-11) and 5-fluorouracil (5-FU) are commonly used to treat metastatic colorectal cancer, but chemotherapy-associated steatosis/steatohepatitis (CASSH) frequently accompanies their use. The objective of this study was to determine effect of CPT-11+5-FU on liver toxicity, liver oxylipins, and cytokines, and to explore whether these alterations could be modified by dietary eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) in the form of fish oil (EPA+DHA). Tumor-bearing animals were administered CPT-11+5-FU and maintained on a control diet or a diet containing EPA+DHA (2.3 g/100 g). Livers were collected one week after chemotherapy for the analysis of oxylipins, cytokines, and markers of liver pathology (oxidized glutathione, GSSH; 4-hydroxynonenal, 4-HNE, and type-I collagen fiber). Dietary EPA+DHA prevented the chemotherapy-induced increases in liver GSSH (p < 0.011) and 4-HNE (p < 0.006). Compared with the tumor-bearing animals, ten oxylipins were altered (three/ten n-6 oxylipins were elevated while seven/ten n-3 oxylipins were reduced) following chemotherapy. Reductions in the n-3 fatty-acid-derived oxylipins that were evident following chemotherapy were restored by dietary EPA+DHA. Liver TNF-α, IL-6 and IL-10 were elevated (p < 0.05) following chemotherapy; dietary EPA+DHA reduced IL-6 (p = 0.09) and eotaxin (p = 0.007) levels. Chemotherapy-induced liver injury results in distinct alterations in oxylipins and cytokines, and dietary EPA+DHA attenuates these pathophysiological effects.
Collapse
Affiliation(s)
- Md Monirujjaman
- Division of Human Nutrition, Department of Agricultural Food and Nutritional Science, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Oliver F. Bathe
- Department of Surgery and Oncology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Vera C. Mazurak
- Division of Human Nutrition, Department of Agricultural Food and Nutritional Science, Li Ka Shing Centre for Health Research Innovation, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Correspondence: ; Tel.: +1-780-492-8048
| |
Collapse
|
45
|
Yang TY, Yu MH, Wu YL, Hong CC, Chen CS, Chan KC, Wang CJ. Mulberry Leaf ( Morus alba L.) Extracts and Its Chlorogenic Acid Isomer Component Improve Glucolipotoxicity-Induced Hepatic Lipid Accumulation via Downregulating miR-34a and Decreased Inflammation. Nutrients 2022; 14:nu14224808. [PMID: 36432495 PMCID: PMC9695749 DOI: 10.3390/nu14224808] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Mulberry leaf (Morus alba L.) is used as a traditional medicine and potential health food to treat various metabolic diseases, such as hypertension, diabetes, and hyperlipidemia. However, we sought the mechanisms by which functional components of mulberry leaves mediate diabetic steatohepatitis. We applied an in vitro model of HepG2 cells induced by glucolipotoxicity and evaluated the effects of MLE and its major components nCGA, Crp, and CGA. The results showed that MLE and nCGA reduced liver fat accumulation by inhibiting SREBP-1/FASN, SREBP-2/HMG-CoAR, and activating PPARα/CPT-1. Additionally, MLE and nCGA decreased inflammatory responses associated with NF-κB, TNF-α, and IL-6 to alleviate steatohepatitis. Furthermore, we showed that MLE and nCGA exerted anti-glucolipotoxicity effects by downregulating miR-34a, thus activating SIRT1/AMPK signaling, and subsequently suppressing hepatic lipid accumulation.
Collapse
Affiliation(s)
- Tsung-Yuan Yang
- Department of Internal Medicine, Chung-Shan Medical University Hospital, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
- School of Medicine, Institute of Medicine, Chung-Shan Medical University, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
| | - Meng-Hsun Yu
- Department of Health Industry Technology Management, Chung Shan Medical University, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
- Department of Nutrition, Chung Shan Medical University, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
| | - Yi-Liang Wu
- Division of Cardiovascular Surgery, Surgical Department, Chung Shan Medical University Hospital, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
- Department of Surgery, School of Medicine, Chung-Shan Medical University, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
| | - Ching-Chun Hong
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
| | - Chin-Shuh Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
| | - Kuei-Chuan Chan
- Department of Internal Medicine, Chung-Shan Medical University Hospital, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
- School of Medicine, Institute of Medicine, Chung-Shan Medical University, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
- Correspondence: (K.-C.C.); (C.-J.W.); Tel.: +886-4-247-30022 (ext. 34704) (K.-C.C. & C.-J.W.)
| | - Chau-Jong Wang
- Department of Health Industry Technology Management, Chung Shan Medical University, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, No. 110, Section 1, Jianguo N. Road, Taichung 402, Taiwan
- Correspondence: (K.-C.C.); (C.-J.W.); Tel.: +886-4-247-30022 (ext. 34704) (K.-C.C. & C.-J.W.)
| |
Collapse
|
46
|
Xie R, Xiao M, Li L, Ma N, Liu M, Huang X, Liu Q, Zhang Y. Association between SII and hepatic steatosis and liver fibrosis: A population-based study. Front Immunol 2022; 13:925690. [PMID: 36189280 PMCID: PMC9520084 DOI: 10.3389/fimmu.2022.925690] [Citation(s) in RCA: 126] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/24/2022] [Indexed: 12/26/2022] Open
Abstract
Background The systemic immune-inflammation index (SII) is a novel marker of inflammation, and hepatic steatosis and fibrosis are associated with inflammation. This study aimed to investigate the possible relationship between SII and hepatic steatosis and fibrosis. Methods The datasets from the National Health and Nutrition Examination Survey (NHANES) 2017–2020 were used in a cross-sectional investigation. Multivariate linear regression models were used to examine the linear connection between SII and controlled attenuation parameter (CAP) and liver stiffness measurement (LSM). Fitted smoothing curves and threshold effect analysis were used to describe the nonlinear relationship. Results This population-based study included a total of 6,792 adults aged 18–80 years. In a multivariate linear regression analysis, a significant positive association between SII and CAP was shown [0.006 (0.001, 0.010)]. This positive association in a subgroup analysis was maintained in men [0.011 (0.004, 0.018)] but not in women. Furthermore, the association between SII and CAP was nonlinear; using a two-segment linear regression model, we found an inverted U-shaped relationship between SII and CAP with an inflection point of 687.059 (1,000 cells/µl). The results of the multiple regression analysis showed that the relationship between SII and LSM was not significant (P = 0.263). Conclusions Our findings imply that increased SII levels are linked to hepatic steatosis, but SII is not linked to liver fibrosis. To confirm our findings, more large-scale prospective investigations are needed.
Collapse
Affiliation(s)
- Ruijie Xie
- Department of Hand and Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Mengde Xiao
- Department of Medical Records Management Center, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Lihong Li
- Department of General Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Nengqian Ma
- Department of General Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Mingjiang Liu
- Department of Hand and Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiongjie Huang
- Department of Hand and Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Qianlong Liu
- Department of Hand and Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Ya Zhang
- Department of General Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, China
- *Correspondence: Ya Zhang,
| |
Collapse
|
47
|
Giri S, Sundaram S. Decompensation in Patients With Advanced NAFLD: More Questions That Need Answering. Clin Gastroenterol Hepatol 2022; 20:2154-2155. [PMID: 34752963 DOI: 10.1016/j.cgh.2021.10.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 02/07/2023]
Affiliation(s)
- Suprabhat Giri
- Department of Gastroenterology, Seth GS Medical College and KEM Hospital, Mumbai, India
| | - Sridhar Sundaram
- Department of Digestive Diseases and Clinical Nutrition, TATA Memorial Hospital, Mumbai, India
| |
Collapse
|
48
|
Cao J, Hua L, Zhang S, Tang J, Ke F, Wu Z, Xue G. Serum interleukin-38 levels correlated with insulin resistance, liver injury and lipids in non-alcoholic fatty liver disease. Lipids Health Dis 2022; 21:70. [PMID: 35948957 PMCID: PMC9364532 DOI: 10.1186/s12944-022-01676-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
Background Insulin resistance, liver injury and dyslipidemia are reported in non-alcoholic fat liver disease (NAFLD) patients. Interleukin (IL)-38 may take part in the pathophysiology of insulin resistance. Nevertheless, the function of IL-38 in NAFLD is unknown. Herein, we determined whether serum IL-38 level might be utilised as a biochemical marker for diagnosing NAFLD. Methods NAFLD patients and healthy participants (n = 91 each) were enrolled. Circulating serum IL-38 levels were detected using enzyme-linked immunosorbent assay. Other metabolic and inflammatory indices related to NAFLD were also assessed. Results Patients with NAFLD had higher serum IL-38 levels than healthy individuals. Significantly higher serum IL-38 levels were found in patients with severe and moderate NAFLD than in patients with mild NAFLD. IL-38 showed a significant correlation with parameters of insulin resistance, inflammation, and liver enzyme in NAFLD cases. Anthropometric, insulin resistance, inflammatory parameters, lipids and frequency of NAFLD showed significant differences among the serum IL-38 level tertiles. Participants in the 2nd and 3rd tertiles of serum IL-38 levels had a greater risk of NAFLD than those in the 1st tertile. Furthermore, IL-38 ROC curve showed a high area under ROC with 0.861. Conclusions It is possible for serum IL-38 to be a biomarker for NAFLD.
Collapse
Affiliation(s)
- Jun Cao
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiujiang University, 17# Lufeng Road, Jiujiang, 332000, Jiangxi Province, China
| | - Lin Hua
- Department of Clinical Laboratory, Jiujiang NO.1 People's Hospital, 48# The South of Taling Road, Jiujiang, 332000, Jiangxi Province, China
| | - Shipei Zhang
- Department of Clinical Laboratory, Jiujiang NO.1 People's Hospital, 48# The South of Taling Road, Jiujiang, 332000, Jiangxi Province, China
| | - Jinping Tang
- Department of Clinical Laboratory, Jiujiang NO.1 People's Hospital, 48# The South of Taling Road, Jiujiang, 332000, Jiangxi Province, China
| | - Fan Ke
- Department of Endocrinology, Jiujiang NO.1 People's Hospital, 48# The South of Taling Road, Jiujiang, 332000, Jiangxi Province, China
| | - Zhouhuan Wu
- Department of pharmacology, School of Medicine, Jiujiang University, 17# Lufeng Road, Jiujiang, 332000, Jiangxi Province, China
| | - Guohui Xue
- Department of Clinical Laboratory, Jiujiang NO.1 People's Hospital, 48# The South of Taling Road, Jiujiang, 332000, Jiangxi Province, China.
| |
Collapse
|
49
|
Kim JE, Kim E, Lee JW. TM4SF5-Mediated Regulation of Hepatocyte Transporters during Metabolic Liver Diseases. Int J Mol Sci 2022; 23:ijms23158387. [PMID: 35955521 PMCID: PMC9369364 DOI: 10.3390/ijms23158387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is found in up to 30% of the world’s population and can lead to hepatocellular carcinoma (HCC), which has a poor 5-year relative survival rate of less than 40%. Clinical therapeutic strategies are not very successful. The co-occurrence of metabolic disorders and inflammatory environments during the development of steatohepatitis thus needs to be more specifically diagnosed and treated to prevent fatal HCC development. To improve diagnostic and therapeutic strategies, the identification of molecules and/or pathways responsible for the initiation and progression of chronic liver disease has been explored in many studies, but further study is still required. Transmembrane 4 L six family member 5 (TM4SF5) has been observed to play roles in the regulation of metabolic functions and activities in hepatocytes using in vitro cell and in vivo animal models without or with TM4SF5 expression in addition to clinical liver tissue samples. TM4SF5 is present on the membranes of different organelles or vesicles and cooperates with transporters for fatty acids, amino acids, and monocarbohydrates, thus regulating nutrient uptake into hepatocytes and metabolism and leading to phenotypes of chronic liver diseases. In addition, TM4SF5 can remodel the immune environment by interacting with immune cells during TM4SF5-mediated chronic liver diseases. Because TM4SF5 may act as an NAFLD biomarker, this review summarizes crosstalk between TM4SF5 and nutrient transporters in hepatocytes, which is related to chronic liver diseases.
Collapse
|
50
|
Vachher M, Bansal S, Kumar B, Yadav S, Arora T, Wali NM, Burman A. Contribution of organokines in the development of NAFLD/NASH associated hepatocellular carcinoma. J Cell Biochem 2022; 123:1553-1584. [PMID: 35818831 DOI: 10.1002/jcb.30252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022]
Abstract
Globally the incidence of hepatocellular carcinoma (HCC) is on an upsurge. Evidence is accumulating that liver disorders like nonalcoholic fatty liver disease (NAFLD) and its more progressive form nonalcoholic steatohepatitis (NASH) are associated with increased risk of developing HCC. NAFLD has a prevalence of about 25% and 50%-90% in obese population. With the growing burden of obesity epidemic worldwide, HCC presents a major healthcare burden. While cirrhosis is one of the major risk factors of HCC, available literature suggests that NAFLD/NASH associated HCC also develops in minimum or noncirrhotic livers. Therefore, there is an urgent need to understand the pathogenesis and risk factors associated with NAFLD and NASH related HCC that would help in early diagnosis and favorable prognosis of HCC secondary to NAFLD. Adipokines, hepatokines and myokines are factors secreted by adipocytes, hepatocytes and myocytes, respectively, playing essential roles in cellular homeostasis, energy balance and metabolism with autocrine, paracrine and endocrine effects. In this review, we endeavor to focus on the role of these organokines in the pathogenesis of NAFLD/NASH and its progression to HCC to augment the understanding of the factors stimulating hepatocytes to acquire a malignant phenotype. This shall aid in the development of novel therapeutic strategies and tools for early diagnosis of NAFLD/NASH and HCC.
Collapse
Affiliation(s)
- Meenakshi Vachher
- Department of Biochemistry, Institute of Home Economics, University of Delhi, Delhi, India
| | - Savita Bansal
- Department of Biochemistry, Institute of Home Economics, University of Delhi, Delhi, India
| | - Bhupender Kumar
- Department of Biochemistry, Institute of Home Economics, University of Delhi, Delhi, India
| | - Sandeep Yadav
- Department of Biochemistry, Institute of Home Economics, University of Delhi, Delhi, India
| | - Taruna Arora
- Department of Biochemistry, Institute of Home Economics, University of Delhi, Delhi, India
| | - Nalini Moza Wali
- Department of Biochemistry, Institute of Home Economics, University of Delhi, Delhi, India
| | - Archana Burman
- Department of Biochemistry, Institute of Home Economics, University of Delhi, Delhi, India
| |
Collapse
|