1
|
Alvarado-Tapias E, Pose E, Gratacós-Ginès J, Clemente-Sánchez A, López-Pelayo H, Bataller R. Alcohol-associated liver disease: Natural history, management and novel targeted therapies. Clin Mol Hepatol 2025; 31:S112-S133. [PMID: 39481875 PMCID: PMC11925442 DOI: 10.3350/cmh.2024.0709] [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: 08/24/2024] [Accepted: 10/29/2024] [Indexed: 11/03/2024] Open
Abstract
Alcohol consumption is a leading cause of preventable morbidity and mortality worldwide and the primary cause of advanced liver disease. Alcohol use disorder is a chronic, frequently relapsing condition characterized by persistent alcohol consumption despite its negative consequences. Alcohol-associated liver disease (ALD) encompasses a series of stages, from fatty liver (steatosis) to inflammation (steatohepatitis), fibrosis, and, ultimately, liver cirrhosis and its complications. The development of ALD is complex, involving both genetic and environmental factors, yet the exact mechanisms at play remain unclear. Alcohol-associated hepatitis (AH), a severe form of ALD, presents with sudden jaundice and liver failure. Currently, there are no approved targeted therapies able to interfere in the pathogenesis of ALD to stop the progression of the disease, making alcohol abstinence the most effective way to improve prognosis across all stages of ALD. For patients with advanced ALD who do not respond to medical therapy, liver transplantation is the only option that can improve prognosis. Recently, AH has become an early indication for liver transplantation in non-responders to medical treatment, showing promising results in carefully selected patients. This review provides an update on the epidemiology, natural history, pathogenesis, and current treatments for ALD. A deeper insight into novel targeted therapies investigated for AH focusing on new pathophysiologically-based agents is also discussed, including anti-inflammatory and antioxidative stress drugs, gut-liver axis modulators, and hepatocyte regenerative molecules.
Collapse
Affiliation(s)
- Edilmar Alvarado-Tapias
- Department of Gastroenterology and Hepatology, Hospital of Santa Creu and Sant Pau, Autonomus University of Barcelona, Barcelona, Spain
- Centre for Biomedical Research in Liver and Digestive Diseases Network (CIBERehd), Madrid, Spain
| | - Elisa Pose
- Centre for Biomedical Research in Liver and Digestive Diseases Network (CIBERehd), Madrid, Spain
- Liver Unit, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Gratacós-Ginès
- Centre for Biomedical Research in Liver and Digestive Diseases Network (CIBERehd), Madrid, Spain
- Liver Unit, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ana Clemente-Sánchez
- Centre for Biomedical Research in Liver and Digestive Diseases Network (CIBERehd), Madrid, Spain
- Department of Gastroenterology and Hepatology, Hospital General Universitario Gregorio Marañón (IiSGM), Madrid, Spain
| | - Hugo López-Pelayo
- Addictions Unit, Psychiatry and Psychology Service, ICN, Hospital Clinic Barcelona, Barcelona; Health and Addictions Research Group, IDIBAPS, Barcelona, Spain
| | - Ramón Bataller
- Centre for Biomedical Research in Liver and Digestive Diseases Network (CIBERehd), Madrid, Spain
- Liver Unit, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
2
|
Coyne ES, Nie Y, Lee D, Pandovski S, Yang T, Zhou H, Rosahl TW, Carballo-Jane E, Abdurrachim D, Zhou Y, Hendra C, Ali AAB, Meyers S, Blumenschein W, Gongol B, Liu Y, Zhou Y, Talukdar S. Loss of mitochondrial amidoxime-reducing component 1 (mARC1) prevents disease progression by reducing fibrosis in multiple mouse models of chronic liver disease. Hepatol Commun 2025; 9:e0637. [PMID: 39927988 PMCID: PMC11809980 DOI: 10.1097/hc9.0000000000000637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/02/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease is a prevalent disease that affects nearly one-third of the global population. Recent genome-wide association studies revealed that a common missense variant in the gene encoding mitochondrial amidoxime reducing component 1 (mARC1) is associated with protection from metabolic dysfunction-associated steatotic liver disease, all-cause cirrhosis, and liver-related mortality suggesting a role for mARC1 in liver pathophysiology; however, little is known about its function in the liver. In this study, we aimed to evaluate the impact of mARC1 hepatoprotective variants on protein function, the effect of loss of mARC1 on cellular lipotoxic stress response, and the effect of global or hepatocyte-specific loss of mARC1 in various mouse models of metabolic dysfunction-associated steatohepatitis and liver fibrosis. METHODS AND RESULTS Expression and characterization of mARC1 hepatoprotective variants in cells and mouse liver revealed that the mARC1 p.A165T exhibited lower protein levels but maintained its mitochondrial localization. In cells, the knockdown of mARC1 improved cellular bioenergetics and decreased mitochondrial superoxide production in response to lipotoxic stress. Global genetic deletion and hepatocyte-specific knockdown of mARC1 in mice significantly reduced liver steatosis and fibrosis in multiple mouse models of metabolic dysfunction-associated steatohepatitis and liver fibrosis. Furthermore, RNA-seq analysis revealed that the pathways involved in extracellular matrix remodeling and collagen formation were downregulated in the liver, and the plasma lipidome was significantly altered in response to the loss of mARC1 in mice. CONCLUSIONS Overall, we have demonstrated that loss of mARC1 alters hepatocyte response to lipotoxic stress and protects mice from diet-induced MASH and liver fibrosis consistent with findings from human genetics.
Collapse
Affiliation(s)
- Erin S. Coyne
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Yilin Nie
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Darwin Lee
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Sentibel Pandovski
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Tiffany Yang
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Heather Zhou
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Thomas W. Rosahl
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Ester Carballo-Jane
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Desiree Abdurrachim
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Yongqi Zhou
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Christopher Hendra
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Asad Abu Bakar Ali
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Stacey Meyers
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Wendy Blumenschein
- Department of Quantitative Biosciences, Merck & Co. Inc., Rahway, New Jersey, USA
| | - Brendan Gongol
- Department of Data, AI & Genome Sciences, MSD, Singapore
| | - Yang Liu
- Department of Data, AI & Genome Sciences, MSD, Singapore
| | - Yingjiang Zhou
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| | - Saswata Talukdar
- Department of Cardiometabolic Disease, Merck & Co. Inc., South San Francisco, California, USA
| |
Collapse
|
3
|
Chen VL, Brady GF. Recent advances in MASLD genetics: Insights into disease mechanisms and the next frontiers in clinical application. Hepatol Commun 2025; 9:e0618. [PMID: 39774697 PMCID: PMC11717516 DOI: 10.1097/hc9.0000000000000618] [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: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 01/11/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease in the world and a growing cause of liver-related morbidity and mortality. Yet, at the same time, our understanding of the pathophysiology and genetic underpinnings of this increasingly common yet heterogeneous disease has increased dramatically over the last 2 decades, with the potential to lead to meaningful clinical interventions for patients. We have now seen the first pharmacologic therapy approved for the treatment of MASLD, and multiple other potential treatments are currently under investigation-including gene-targeted RNA therapies that directly extend from advances in MASLD genetics. Here we review recent advances in MASLD genetics, some of the key pathophysiologic insights that human genetics has provided, and the ways in which human genetics may inform our clinical practice in the field of MASLD in the near future.
Collapse
|
4
|
Giardoglou P, Gavra I, Amanatidou AI, Kalafati IP, Symianakis P, Kafyra M, Moulos P, Dedoussis GV. Development of a Polygenic Risk Score for Metabolic Dysfunction-Associated Steatotic Liver Disease Prediction in UK Biobank. Genes (Basel) 2024; 16:33. [PMID: 39858580 PMCID: PMC11765347 DOI: 10.3390/genes16010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the leading cause of liver-related morbidity and mortality. Although the invasive liver biopsy remains the golden standard for MASLD diagnosis, Magnetic Resonance Imaging-derived Proton Density Fat Fraction (MRI-PDFF) is an accurate, non-invasive method for the assessment of treatment response. This study aimed at developing a Polygenic Risk Score (PRS) to improve MRI-PDFF prediction using UK Biobank data to assess an individual's genetic liability to MASLD. METHODS We iteratively sequestered 10% of MRI-PDFF samples as a validation set and split the rest of each dataset into base and target partitions, containing GWAS summary statistics and raw genotype data, respectively. PRSice2 was deployed to derive PRS candidates. Based on the frequency of SNP appearances along the PRS candidates, we generated different SNP sets according to variable frequency cutoffs. By applying the PRSs to the validation set, we identified the optimal SNP set, which was then applied to a Greek nonalcoholic fatty liver disease (NAFLD) study. RESULTS Data from 3553 UK Biobank participants yielded 49 different SNP sets. After calculating the PRS on the validation set for every SNP set, an optimal PRS with 75 SNPs was selected (incremental R2 = 0.025, p-value = 0.00145). Interestingly, 43 SNPs were successfully mapped to MASLD-related known genes. The selected PRS could predict traits, like LDL cholesterol and diastolic blood pressure in the UK Biobank, as also disease outcome in the Greek NAFLD study. CONCLUSIONS Our findings provide strong evidence that PRS is a powerful prediction model for MASLD, while it can also be applied on populations of different ethnicity.
Collapse
Affiliation(s)
- Panagiota Giardoglou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Ioanna Gavra
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Athina I. Amanatidou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Ioanna Panagiota Kalafati
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
- Department of Nutrition and Dietetics, School of Physical Education, Sport Science and Dietetics, University of Thessaly, 42132 Trikala, Greece
| | - Panagiotis Symianakis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Maria Kafyra
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Panagiotis Moulos
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center ‘Alexander Fleming’, 16672 Vari, Greece;
| | - George V. Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| |
Collapse
|
5
|
Mullish BH, Thursz MR. Alcohol-associated liver disease: Emerging therapeutic strategies. Hepatology 2024; 80:1372-1389. [PMID: 38922808 DOI: 10.1097/hep.0000000000000986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
The large and growing burden of alcohol-associated liver disease-and the considerable burden of morbidity and mortality associated with it-has been a drive toward ongoing research into novel strategies for its treatment, with a particular focus upon alcohol-associated hepatitis (AH). Management of alcohol-use disorder forms the central pillar of alcohol-associated liver disease care, with evidence-based psychological and pharmacological approaches being well established, and certain models demonstrating improved clinical outcomes when hepatology and addiction services are co-located. Corticosteroids have previously been used somewhat indiscriminately in patients with severe AH, but effective tools now exist to assess early response (and limit futile ongoing exposure). Techniques to predict risk of corticosteroid-related infection are also available, although current clinical strategies to mitigate this risk are limited. A variety of novel therapeutic approaches to AH are at different phases of trials and evidence gathering, with some of the most promising signals related to cytokine manipulation, epigenetic modulation, and targeting of the gut microbiota (ie, by means of fecal microbiota transplant). While remaining an ongoing source of debate, early liver transplant in severe AH has grown in interest and acceptability over the past decade as evidence supporting its efficacy builds, in the process challenging paradigms about mandatory pretransplant sobriety periods. However, uncertainty remains regarding the optimal selection criteria, and whether liver transplant has a role for only a highly limited proportion of patients with AH or more widespread application. This review aims to provide an overview of this fast-moving field.
Collapse
Affiliation(s)
- Benjamin H Mullish
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
- Department of Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Mark R Thursz
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
- Department of Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
6
|
Raya Tonetti F, Eguileor A, Mrdjen M, Pathak V, Travers J, Nagy LE, Llorente C. Gut-liver axis: Recent concepts in pathophysiology in alcohol-associated liver disease. Hepatology 2024; 80:1342-1371. [PMID: 38691396 PMCID: PMC11801230 DOI: 10.1097/hep.0000000000000924] [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: 02/13/2024] [Accepted: 04/20/2024] [Indexed: 05/03/2024]
Abstract
The growing recognition of the role of the gut microbiome's impact on alcohol-associated diseases, especially in alcohol-associated liver disease, emphasizes the need to understand molecular mechanisms involved in governing organ-organ communication to identify novel avenues to combat alcohol-associated diseases. The gut-liver axis refers to the bidirectional communication and interaction between the gut and the liver. Intestinal microbiota plays a pivotal role in maintaining homeostasis within the gut-liver axis, and this axis plays a significant role in alcohol-associated liver disease. The intricate communication between intestine and liver involves communication between multiple cellular components in each organ that enable them to carry out their physiological functions. In this review, we focus on novel approaches to understanding how chronic alcohol exposure impacts the microbiome and individual cells within the liver and intestine, as well as the impact of ethanol on the molecular machinery required for intraorgan and interorgan communication.
Collapse
Affiliation(s)
| | - Alvaro Eguileor
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Marko Mrdjen
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH
| | - Vai Pathak
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jared Travers
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH
- Department of Gastroenterology and Hepatology, University Hospital, Cleveland OH
| | - Laura E Nagy
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland OH
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
7
|
Palupi PD, Wei CY, Chou WH, Lin MR, Wan YJY, Chang WC. Dietary contributions in the genetic variation of liver fibrosis: a genome-wide association study of fibrosis-4 index in the liver fibrosis development. Cell Biosci 2024; 14:141. [PMID: 39578894 PMCID: PMC11583755 DOI: 10.1186/s13578-024-01321-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/04/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND The fibrosis-4 (FIB-4) index is a non-invasive method to assess the severity of liver fibrosis. The development of liver fibrosis is influenced by genetic predisposition and dietary factors. However, the modulating effect of dietary factors on the genetic susceptibility of liver fibrosis remains unclear. The study aims to investigate the role of dietary factors in modulating the genetic susceptibility of liver fibrosis. METHODS Here, we conducted a genome-wide association study (GWAS) of FIB-4 index-directed liver fibrosis risk, adjusted with diet, lifestyle factors, and hepatitis serological markers. The high (N = 1,476) and low (N = 36,735) liver fibrosis risk groups were defined with a FIB-4 > 2.67 and < 1.3, respectively. RESULTS The age-related FIB-4 variation showed subjects with a FIB-4 > 2.67 (3.8%), indicating high fibrosis risk, occurred predominantly among individuals above 60 years old. The multivariable analysis showed that tea intake is significantly associated with a reduced risk of liver fibrosis. The GWAS adjusted for sex, age, age2, dietary factors (tea and coffee consumption, vegetarian preference), lifestyle (alcohol consumption, physical activity), hepatitis serological markers (anti-HCV, HBsAg, HBeAg), and the top ten principal components indicated 25 genome-wide significant signals (p < 5 × 10- 8). Two variants (rs56293029 and rs9389269) were previously associated with the FIB-4 index in alcohol-related cirrhosis, while the 23 SNPs remaining were novel. The rs9399136 (HBS1L) is a protective variant, and rs9274407 (HLA-DQB1) is a risk variant, both contributing to liver fibrosis development. Our results showed that genetic factors play a major role in liver fibrosis, while dietary factors have minor effects on disease progression. Pathway analysis suggested the potential of immune response and hematopoietic systems function in the pathogenesis of liver disease. CONCLUSIONS The studies not only revealed the protective role of rs9399136 (HBS1L) and the risk effect of rs9274407 (HLA-DQB1) toward liver fibrosis in a Taiwanese population, but also demonstrated that individual consumption patterns, such as tea uptake, have a minor impact on liver fibrosis prevention. The pathway analysis from GWAS variants further indicated the importance of immune responses in the pathogenesis of liver fibrosis.
Collapse
Affiliation(s)
- Poppy Diah Palupi
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chun-Yu Wei
- Core Laboratory of Neoantigen Analysis for Personalized Cancer Vaccine, Office of R&D, Taipei Medical University, Taipei, 11031, Taiwan
| | - Wan-Hsuan Chou
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Min-Rou Lin
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Jui Yvonne Wan
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan.
- Master Program in Clinical Genomics and Proteomics, Taipei Medical University, Taipei, 11031, Taiwan.
- Integrative Research Center for Critical Care, Department of Pharmacy, Taipei Medical University-Wan-Fang Hospital, Taipei, 11696, Taiwan.
- Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, 11696, Taiwan.
| |
Collapse
|
8
|
Mignogna KM, Tatom Z, Macleod L, Sergi Z, Nguyen A, Michenkova M, Smith ML, Miles MF. Identification of novel genetic loci and candidate genes for progressive ethanol consumption in diversity outbred mice. Neuropsychopharmacology 2024; 49:1892-1904. [PMID: 38951586 PMCID: PMC11473901 DOI: 10.1038/s41386-024-01902-6] [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: 01/23/2024] [Revised: 04/26/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024]
Abstract
Mouse behavioral genetic mapping studies can identify genomic intervals modulating complex traits under well-controlled environmental conditions and have been used to study ethanol behaviors to aid in understanding genetic risk and the neurobiology of alcohol use disorder (AUD). However, historically such studies have produced large confidence intervals, thus complicating identification of potential causal candidate genes. Diversity Outbred (DO) mice offer the ability to perform high-resolution quantitative trait loci (QTL) mapping on a very genetically diverse background, thus facilitating identification of candidate genes. Here, we studied a population of 636 male DO mice with four weeks of intermittent ethanol access via a three-bottle choice procedure, producing a progressive ethanol consumption phenotype. QTL analysis identified 3 significant (Chrs 3, 4, and 12) and 13 suggestive loci for ethanol-drinking behaviors with narrow confidence intervals (1-4 Mbp for significant QTLs). Results suggested that genetic influences on initial versus progressive ethanol consumption were localized to different genomic intervals. A defined set of positional candidate genes were prioritized using haplotype analysis, identified coding polymorphisms, prefrontal cortex transcriptomics data, human GWAS data and prior rodent gene set data for ethanol or other misused substances. These candidates included Car8, the lone gene with a significant cis-eQTL within a Chr 4 QTL for week four ethanol consumption. These results represent the highest-resolution genetic mapping of ethanol consumption behaviors in mice to date, providing identification of novel loci and candidate genes for study in relation to the neurobiology of AUD.
Collapse
Affiliation(s)
- Kristin M Mignogna
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Zachary Tatom
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Lorna Macleod
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Zachary Sergi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Angel Nguyen
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Marie Michenkova
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Maren L Smith
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael F Miles
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA.
- VCU Alcohol Research Center, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
9
|
Adamus JP, Ruszczyńska A, Wyczałkowska-Tomasik A. Molybdenum's Role as an Essential Element in Enzymes Catabolizing Redox Reactions: A Review. Biomolecules 2024; 14:869. [PMID: 39062583 PMCID: PMC11275037 DOI: 10.3390/biom14070869] [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: 06/04/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Molybdenum (Mo) is an essential element for human life, acting as a cofactor in various enzymes crucial for metabolic homeostasis. This review provides a comprehensive insight into the latest advances in research on molybdenum-containing enzymes and their clinical significance. One of these enzymes is xanthine oxidase (XO), which plays a pivotal role in purine catabolism, generating reactive oxygen species (ROS) capable of inducing oxidative stress and subsequent organ dysfunction. Elevated XO activity is associated with liver pathologies such as non-alcoholic fatty liver disease (NAFLD) and hepatocellular carcinoma (HCC). Aldehyde oxidases (AOs) are also molybdenum-containing enzymes that, similar to XO, participate in drug metabolism, with notable roles in the oxidation of various substrates. However, beneath its apparent efficacy, AOs' inhibition may impact drug effectiveness and contribute to liver damage induced by hepatotoxins. Another notable molybdenum-enzyme is sulfite oxidase (SOX), which catalyzes the conversion of sulfite to sulfate, crucial for the degradation of sulfur-containing amino acids. Recent research highlights SOX's potential as a diagnostic marker for HCC, offering promising sensitivity and specificity in distinguishing cancerous lesions. The newest member of molybdenum-containing enzymes is mitochondrial amidoxime-reducing component (mARC), involved in drug metabolism and detoxification reactions. Emerging evidence suggests its involvement in liver pathologies such as HCC and NAFLD, indicating its potential as a therapeutic target. Overall, understanding the roles of molybdenum-containing enzymes in human physiology and disease pathology is essential for advancing diagnostic and therapeutic strategies for various health conditions, particularly those related to liver dysfunction. Further research into the molecular mechanisms underlying these enzymes' functions could lead to novel treatments and improved patient outcomes.
Collapse
Affiliation(s)
- Jakub Piotr Adamus
- Faculty of Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna Ruszczyńska
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, 02-089 Warsaw, Poland
| | | |
Collapse
|
10
|
Koshiol J, Krawczyk M. enHanCCing knowledge of genetic factors in primary liver tumor. Hepatology 2024; 80:11-13. [PMID: 38349672 DOI: 10.1097/hep.0000000000000771] [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: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 06/20/2024]
Affiliation(s)
- Jill Koshiol
- Division of Cancer Epidemiology and Genetics, Infections and Immunoepidemiology Branch, National Cancer Institute, Rockville, Maryland, USA
| | - Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
- Department of General, Laboratory of Metabolic Liver Diseases, Center for Preclinical Research, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
11
|
Hou W, Watson C, Cecconie T, Bolaki MN, Brady JJ, Lu Q, Gatto GJ, Day TA. Biochemical and functional characterization of the p.A165T missense variant of mitochondrial amidoxime-reducing component 1. J Biol Chem 2024; 300:107353. [PMID: 38723751 PMCID: PMC11190489 DOI: 10.1016/j.jbc.2024.107353] [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: 02/28/2024] [Revised: 04/25/2024] [Accepted: 05/03/2024] [Indexed: 06/07/2024] Open
Abstract
Recent genome-wide association studies have identified a missense variant p.A165T in mitochondrial amidoxime-reducing component 1 (mARC1) that is strongly associated with protection from all-cause cirrhosis and improved prognosis in nonalcoholic steatohepatitis. The precise mechanism of this protective effect is unknown. Substitution of alanine 165 with threonine is predicted to affect mARC1 protein stability and to have deleterious effects on its function. To investigate the mechanism, we have generated a knock-in mutant mARC1 A165T and a catalytically dead mutant C273A (as a control) in human hepatoma HepG2 cells, enabling characterization of protein subcellular distribution, stability, and biochemical functions of the mARC1 mutant protein expressed from its endogenous locus. Compared to WT mARC1, we found that the A165T mutant exhibits significant mislocalization outside of its traditional location anchored in the mitochondrial outer membrane and reduces protein stability, resulting in lower basal levels. We evaluated the involvement of the ubiquitin proteasome system in mARC1 A165T degradation and observed increased ubiquitination and faster degradation of the A165T variant. In addition, we have shown that HepG2 cells carrying the MTARC1 p.A165T variant exhibit lower N-reductive activity on exogenously added amidoxime substrates in vitro. The data from these biochemical and functional assays suggest a mechanism by which the MTARC1 p.A165T variant abrogates enzyme function which may contribute to its protective effect in liver disease.
Collapse
Affiliation(s)
- Wangfang Hou
- Respiratory and Immunology Biology Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Christian Watson
- Respiratory and Immunology Biology Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Ted Cecconie
- MEDDesign-NCE-MD SPMB US, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | | | | | - Quinn Lu
- Respiratory and Immunology Biology Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Gregory J Gatto
- Respiratory and Immunology Biology Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA.
| | - Tovah A Day
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Nauffal V, Klarqvist MDR, Hill MC, Pace DF, Di Achille P, Choi SH, Rämö JT, Pirruccello JP, Singh P, Kany S, Hou C, Ng K, Philippakis AA, Batra P, Lubitz SA, Ellinor PT. Noninvasive assessment of organ-specific and shared pathways in multi-organ fibrosis using T1 mapping. Nat Med 2024; 30:1749-1760. [PMID: 38806679 DOI: 10.1038/s41591-024-03010-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/22/2024] [Indexed: 05/30/2024]
Abstract
Fibrotic diseases affect multiple organs and are associated with morbidity and mortality. To examine organ-specific and shared biologic mechanisms that underlie fibrosis in different organs, we developed machine learning models to quantify T1 time, a marker of interstitial fibrosis, in the liver, pancreas, heart and kidney among 43,881 UK Biobank participants who underwent magnetic resonance imaging. In phenome-wide association analyses, we demonstrate the association of increased organ-specific T1 time, reflecting increased interstitial fibrosis, with prevalent diseases across multiple organ systems. In genome-wide association analyses, we identified 27, 18, 11 and 10 independent genetic loci associated with liver, pancreas, myocardial and renal cortex T1 time, respectively. There was a modest genetic correlation between the examined organs. Several loci overlapped across the examined organs implicating genes involved in a myriad of biologic pathways including metal ion transport (SLC39A8, HFE and TMPRSS6), glucose metabolism (PCK2), blood group antigens (ABO and FUT2), immune function (BANK1 and PPP3CA), inflammation (NFKB1) and mitosis (CENPE). Finally, we found that an increasing number of organs with T1 time falling in the top quintile was associated with increased mortality in the population. Individuals with a high burden of fibrosis in ≥3 organs had a 3-fold increase in mortality compared to those with a low burden of fibrosis across all examined organs in multivariable-adjusted analysis (hazard ratio = 3.31, 95% confidence interval 1.77-6.19; P = 1.78 × 10-4). By leveraging machine learning to quantify T1 time across multiple organs at scale, we uncovered new organ-specific and shared biologic pathways underlying fibrosis that may provide therapeutic targets.
Collapse
Affiliation(s)
- Victor Nauffal
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Matthew C Hill
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Danielle F Pace
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paolo Di Achille
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joel T Rämö
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Pulkit Singh
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cody Hou
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenney Ng
- Center for Computational Health, IBM Research, Cambridge, MA, USA
| | - Anthony A Philippakis
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Puneet Batra
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven A Lubitz
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
13
|
Schwantes-An TH, Whitfield JB, Aithal GP, Atkinson SR, Bataller R, Botwin G, Chalasani NP, Cordell HJ, Daly AK, Darlay R, Day CP, Eyer F, Foroud T, Gawrieh S, Gleeson D, Goldman D, Haber PS, Jacquet JM, Lammert CS, Liang T, Liangpunsakul S, Masson S, Mathurin P, Moirand R, McQuillin A, Moreno C, Morgan MY, Mueller S, Müllhaupt B, Nagy LE, Nahon P, Nalpas B, Naveau S, Perney P, Pirmohamed M, Seitz HK, Soyka M, Stickel F, Thompson A, Thursz MR, Trépo E, Morgan TR, Seth D. A polygenic risk score for alcohol-associated cirrhosis among heavy drinkers with European ancestry. Hepatol Commun 2024; 8:e0431. [PMID: 38727677 PMCID: PMC11093576 DOI: 10.1097/hc9.0000000000000431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/01/2023] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Polygenic Risk Scores (PRS) based on results from genome-wide association studies offer the prospect of risk stratification for many common and complex diseases. We developed a PRS for alcohol-associated cirrhosis by comparing single-nucleotide polymorphisms among patients with alcohol-associated cirrhosis (ALC) versus drinkers who did not have evidence of liver fibrosis/cirrhosis. METHODS Using a data-driven approach, a PRS for ALC was generated using a meta-genome-wide association study of ALC (N=4305) and an independent cohort of heavy drinkers with ALC and without significant liver disease (N=3037). It was validated in 2 additional independent cohorts from the UK Biobank with diagnosed ALC (N=467) and high-risk drinking controls (N=8981) and participants in the Indiana Biobank Liver cohort with alcohol-associated liver disease (N=121) and controls without liver disease (N=3239). RESULTS A 20-single-nucleotide polymorphisms PRS for ALC (PRSALC) was generated that stratified risk for ALC comparing the top and bottom deciles of PRS in the 2 validation cohorts (ORs: 2.83 [95% CI: 1.82 -4.39] in UK Biobank; 4.40 [1.56 -12.44] in Indiana Biobank Liver cohort). Furthermore, PRSALC improved the prediction of ALC risk when added to the models of clinically known predictors of ALC risk. It also stratified the risk for metabolic dysfunction -associated steatotic liver disease -cirrhosis (3.94 [2.23 -6.95]) in the Indiana Biobank Liver cohort -based exploratory analysis. CONCLUSIONS PRSALC incorporates 20 single-nucleotide polymorphisms, predicts increased risk for ALC, and improves risk stratification for ALC compared with the models that only include clinical risk factors. This new score has the potential for early detection of heavy drinking patients who are at high risk for ALC.
Collapse
Affiliation(s)
- Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis IN, USA
| | - John B. Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Queensland 4029, Australia
| | - Guruprasad P. Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Stephen R. Atkinson
- Department of Metabolism, Digestion & Reproduction, Imperial College London, UK
| | - Ramon Bataller
- Center for Liver Diseases, University of Pittsburgh Medical Center, 3471 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Greg Botwin
- Department of Veterans Affairs, VA Long Beach Healthcare System, 5901 East Seventh Street, Long Beach, CA 90822, USA
- F. Widjaja Family Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California CA 90048, USA
| | - Naga P. Chalasani
- Department of Medicine, Indiana University, Indianapolis, IN 46202-5175, USA
| | - Heather J. Cordell
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Ann K. Daly
- Faculty of Medical Sciences, Newcastle University Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Rebecca Darlay
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Christopher P. Day
- Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Florian Eyer
- Division of Clinical Toxicology, Department of Internal Medicine 2, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis IN, USA
| | - Samer Gawrieh
- Department of Medicine, Indiana University, Indianapolis, IN 46202-5175, USA
| | - Dermot Gleeson
- Liver Unit, Sheffield Teaching Hospitals, AO Floor Robert Hadfield Building, Northern General Hospital, Sheffield S5 7AU, UK
| | - David Goldman
- Office of the Clinical Director and Laboratory of Neurogenetics, NIAAA, Bethesda, MD 20952, USA
| | - Paul S. Haber
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Sydney Local Health District, Missenden Road, Camperdown, NSW 2050, Australia
- Faculty of Medicine and Health, the University of Sydney, Sydney, NSW 2006, Australia
| | | | - Craig S. Lammert
- Department of Medicine, Indiana University, Indianapolis, IN 46202-5175, USA
| | - Tiebing Liang
- Department of Medicine, Indiana University, Indianapolis, IN 46202-5175, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University and Roudebush Veterans Administration Medical Center, Indianapolis, USA
| | - Steven Masson
- Faculty of Medical Sciences, Newcastle University Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Philippe Mathurin
- CHRU de Lille, Hôpital Claude Huriez, Rue M. Polonovski CS 70001, 59 037 Lille Cedex, France
| | - Romain Moirand
- Univ Rennes, INRA, INSERM, CHU Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
| | - Andrew McQuillin
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College London, London WC1E 6DE, UK
| | - Christophe Moreno
- CUB Hôpital Erasme, Université Libre de Bruxelles, clinique d’Hépatologie, Brussels, Belgium; Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Marsha Y. Morgan
- UCL Institute for Liver & Digestive Health, Division of Medicine, Royal Free Campus, University College London, London NW3 2PF, UK
| | - Sebastian Mueller
- Department of Internal Medicine, Salem Medical Center and Center for Alcohol Research, University of Heidelberg, Zeppelinstraße 11-33, 69121 Heidelberg, Germany
| | - Beat Müllhaupt
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, CH-8901 Zurich, Switzerland
| | - Laura E. Nagy
- Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio, OH 44195, USA
| | - Pierre Nahon
- Service d'Hépatologie, APHP Hôpital Avicenne et Université Paris 13, Bobigny, France
- University Paris 13, Bobigny, France
- Inserm U1162 Génomique fonctionnelle des tumeurs solides, Paris, France
| | - Bertrand Nalpas
- Service Addictologie, CHRU Caremeau, 30029 Nîmes, France
- DISC, Inserm, 75013 Paris, France
| | - Sylvie Naveau
- Hôpital Antoine-Béclère, 157 Rue de la Porte de Trivaux, 92140 Clamart, France
| | - Pascal Perney
- Hôpital Universitaire Caremeau, Place du Pr. Robert Debre, 30029 Nîmes, France
| | - Munir Pirmohamed
- MRC Centre for Drug Safety Science, Liverpool Centre for Alcohol Research, University of Liverpool, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, and Liverpool Health Partners, Liverpool, L69 3GL, UK
| | - Helmut K. Seitz
- Department of Internal Medicine, Salem Medical Center and Center for Alcohol Research, University of Heidelberg, Zeppelinstraße 11-33, 69121 Heidelberg, Germany
| | - Michael Soyka
- Psychiatric Hospital University of Munich, Nussbaumsstr.7, 80336 Munich, Germany
| | - Felix Stickel
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, CH-8901 Zurich, Switzerland
| | - Andrew Thompson
- MRC Centre for Drug Safety Science, Liverpool Centre for Alcohol Research, University of Liverpool, The Royal Liverpool and Broadgreen University Hospitals NHS Trust, and Liverpool Health Partners, Liverpool, L69 3GL, UK
- Health Analytics, Lane Clark & Peacock LLP, London, UK
| | - Mark R. Thursz
- Department of Metabolism, Digestion & Reproduction, Imperial College London, UK
| | - Eric Trépo
- CUB Hôpital Erasme, Université Libre de Bruxelles, clinique d’Hépatologie, Brussels, Belgium; Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Timothy R. Morgan
- Department of Medicine, University of California, Irvine, USA
- Department of Veterans Affairs, VA Long Beach Healthcare System, 5901 East Seventh Street, Long Beach, CA 90822, USA
| | - Devanshi Seth
- Edith Collins Centre (Translational Research in Alcohol Drugs and Toxicology), Sydney Local Health District, Missenden Road, Camperdown, NSW 2050, Australia
- Faculty of Medicine and Health, the University of Sydney, Sydney, NSW 2006, Australia
- Centenary Institute of Cancer Medicine and Cell Biology, the University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
14
|
Dutta T, Sasidharan K, Ciociola E, Pennisi G, Noto FR, Kovooru L, Kroon T, Lindblom A, Du Y, Pirmoradian M, Wallin S, Mancina RM, Lindén D, Romeo S. Mitochondrial amidoxime-reducing component 1 p.Ala165Thr increases protein degradation mediated by the proteasome. Liver Int 2024; 44:1219-1232. [PMID: 38375985 DOI: 10.1111/liv.15857] [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: 12/08/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/21/2024]
Abstract
OBJECTIVE Metabolic dysfunction-associated steatotic liver disease (MASLD) is a global health concern with no effective and specific drug treatment available. The rs2642438 minor allele in mitochondrial amidoxime-reducing component 1 (MARC1) results in an aminoacidic substitution (p.Ala165Thr) and associates with protection against MASLD. However, the mechanisms behind this protective effect are unknown. In this study, we examined the consequences of this aminoacidic substitution on protein stability and subcellular localization. METHODS We overexpressed the human MARC1 A165 (wild-type) or 165T (mutant) in vivo in mice and in vitro in human hepatoma cells (HepG2 and HuH-7), generated several mutants at position 165 by in situ mutagenesis and then examined protein levels. We also generated HepG2 cells stably overexpressing MARC1 A165 or 165T to test the effect of this substitution on MARC1 subcellular localization. RESULTS MARC1 165T overexpression resulted in lower protein levels than A165 both in vivo and in vitro. Similarly, any mutant at position 165 showed lower protein levels compared to the wild-type protein. We showed that the 165T mutant protein is polyubiquitinated and its degradation is accelerated through lysine-48 ubiquitin-mediated proteasomal degradation. We also showed that the 165T substitution does not affect the MARC1 subcellular localization. CONCLUSIONS This study shows that alanine at position 165 in MARC1 is crucial for protein stability, and the threonine substitution at this position leads to a hypomorphic protein variant due to lower protein levels. Our result supports the notion that lowering hepatic MARC1 protein level may be a successful therapeutic strategy for treating MASLD.
Collapse
Affiliation(s)
- Tanmoy Dutta
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Kavitha Sasidharan
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Ester Ciociola
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Grazia Pennisi
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
- Section of Gastroenterology and Hepatology, Dipartimento Di Promozione Della Salute, Materno Infantile, Medicina Interna e Specialistica Di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Francesca R Noto
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
- Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Lohitesh Kovooru
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Tobias Kroon
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Lindblom
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Yue Du
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Mohammad Pirmoradian
- Translational Science and Experimental Medicine, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Simonetta Wallin
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rosellina M Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Lindén
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Division of Endocrinology, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
- Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
15
|
Mukhopadhyay B, Marietta C, Shen PH, Oiseni A, Mirshahi F, Mazzu M, Hodgkinson C, Winkler E, Yuan Q, Miranda D, Kunos G, Sanyal AJ, Goldman D. A patient-based iPSC-derived hepatocyte model of alcohol-associated cirrhosis reveals bioenergetic insights into disease pathogenesis. Nat Commun 2024; 15:2869. [PMID: 38693144 PMCID: PMC11063145 DOI: 10.1038/s41467-024-47085-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: 05/28/2023] [Accepted: 03/15/2024] [Indexed: 05/03/2024] Open
Abstract
Only ~20% of heavy drinkers develop alcohol cirrhosis (AC). While differences in metabolism, inflammation, signaling, microbiome signatures and genetic variations have been tied to the pathogenesis of AC, the key underlying mechanisms for this interindividual variability, remain to be fully elucidated. Induced pluripotent stem cell-derived hepatocytes (iHLCs) from patients with AC and healthy controls differ transcriptomically, bioenergetically and histologically. They include a greater number of lipid droplets (LDs) and LD-associated mitochondria compared to control cells. These pre-pathologic indicators are effectively reversed by Aramchol, an inhibitor of stearoyl-CoA desaturase. Bioenergetically, AC iHLCs have lower spare capacity, slower ATP production and their mitochondrial fuel flexibility towards fatty acids and glutamate is weakened. MARC1 and PNPLA3, genes implicated by GWAS in alcohol cirrhosis, show to correlate with lipid droplet-associated and mitochondria-mediated oxidative damage in AC iHLCs. Knockdown of PNPLA3 expression exacerbates mitochondrial deficits and leads to lipid droplets alterations. These findings suggest that differences in mitochondrial bioenergetics and lipid droplet formation are intrinsic to AC hepatocytes and can play a role in its pathogenesis.
Collapse
Affiliation(s)
- Bani Mukhopadhyay
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Cheryl Marietta
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Pei-Hong Shen
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Abdul Oiseni
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Faridoddin Mirshahi
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - Maria Mazzu
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Colin Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Eli Winkler
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Qiaoping Yuan
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Daniel Miranda
- Aivia Machine Learning Team, Leica Microsystems, Inc, Deerfield, IL, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA.
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
16
|
Wu M, Tie M, Hu L, Yang Y, Chen Y, Ferguson D, Chen Y, He A. Fatty liver disease protective MTARC1 p.A165T variant reduces the protein stability of MTARC1. Biochem Biophys Res Commun 2024; 702:149655. [PMID: 38340654 PMCID: PMC10940201 DOI: 10.1016/j.bbrc.2024.149655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common causes of liver disease worldwide. MTARC1, encoded by the MTARC1 gene, is a mitochondrial outer membrane-anchored enzyme. Interestingly, the MTARC1 p.A165T (rs2642438) variant is associated with a decreased risk of NAFLD, indicating that MTARC1 might be an effective target. It has been reported that the rs2642438 variant does not have altered enzymatic activity so we reasoned that this variation may affect MTARC1 stability. In this study, MTARC1 mutants were generated and stability was assessed using a protein stability reporter system both in vitro and in vivo. We found that the MTARC1 p.A165T variant has dramatically reduced the stability of MTARC1, as assessed in several cell lines. In mice, the MTARC1 A168T mutant, the equivalent of human MTARC1 A165T, had diminished stability in mouse liver. Additionally, several MTARC1 A165 mutants, including A165S, A165 N, A165V, A165G, and A165D, had dramatically decreased stability as well, suggesting that the alanine residue of MTARC1 165 site is essential for MTARC1 protein stability. Collectively, our data indicates that the MTARC1 p.A165T variant (rs2642438) leads to reduced stability of MTARC1. Given that carriers of rs2642438 show a decreased risk of NAFLD, the findings herein support the notion that MTARC1 inhibition may be a therapeutic target to combat NAFLD.
Collapse
Affiliation(s)
- Mengyue Wu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Meng Tie
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Liwei Hu
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yunzhi Yang
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yong Chen
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Daniel Ferguson
- Division of Nutritional Science and Obesity Medicine, Washington University in St. Louis, United States
| | - Yali Chen
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Anyuan He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Science, Anhui Medical University, Hefei, 230032, Anhui, China; Department of Biochemistry and Molecular Biology, College of Life Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
17
|
Jokinen MJ, Luukkonen PK. Hepatic mitochondrial reductive stress in the pathogenesis and treatment of steatotic liver disease. Trends Pharmacol Sci 2024; 45:319-334. [PMID: 38471991 DOI: 10.1016/j.tips.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/14/2024]
Abstract
Steatotic liver diseases (SLDs) affect one-third of the population, but the pathogenesis underlying these diseases is not well understood, limiting the available treatments. A common factor in SLDs is increased hepatic mitochondrial reductive stress, which occurs as a result of excessive lipid and alcohol metabolism. Recent research has also shown that genetic risk factors contribute to this stress. This review aims to explore how these risk factors increase hepatic mitochondrial reductive stress and how it disrupts hepatic metabolism, leading to SLDs. Additionally, the review will discuss the latest clinical studies on pharmaceutical treatments for SLDs, specifically peroxisome proliferator-activated receptor gamma (PPAR-γ) agonists, thyroid hormone receptor (THR) agonists, acetyl-CoA carboxylase (ACC) inhibitors, and mitochondrial uncouplers. These treatments have a common effect of decreasing hepatic mitochondrial reductive stress, which has been largely overlooked.
Collapse
Affiliation(s)
- Mari J Jokinen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Internal Medicine, University of Helsinki, Helsinki, Finland; Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Panu K Luukkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland; Department of Internal Medicine, University of Helsinki, Helsinki, Finland; Abdominal Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
18
|
Smagris E, Shihanian LM, Mintah IJ, Bigdelou P, Livson Y, Brown H, Verweij N, Hunt C, Johnson RO, Greer TJ, Hartford SA, Hindy G, Sun L, Nielsen JB, Halasz G, Lotta LA, Murphy AJ, Sleeman MW, Gusarova V. Divergent role of Mitochondrial Amidoxime Reducing Component 1 (MARC1) in human and mouse. PLoS Genet 2024; 20:e1011179. [PMID: 38437227 PMCID: PMC10939284 DOI: 10.1371/journal.pgen.1011179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 03/14/2024] [Accepted: 02/09/2024] [Indexed: 03/06/2024] Open
Abstract
Recent human genome-wide association studies have identified common missense variants in MARC1, p.Ala165Thr and p.Met187Lys, associated with lower hepatic fat, reduction in liver enzymes and protection from most causes of cirrhosis. Using an exome-wide association study we recapitulated earlier MARC1 p.Ala165Thr and p.Met187Lys findings in 540,000 individuals from five ancestry groups. We also discovered novel rare putative loss of function variants in MARC1 with a phenotype similar to MARC1 p.Ala165Thr/p.Met187Lys variants. In vitro studies of recombinant human MARC1 protein revealed Ala165Thr substitution causes protein instability and aberrant localization in hepatic cells, suggesting MARC1 inhibition or deletion may lead to hepatoprotection. Following this hypothesis, we generated Marc1 knockout mice and evaluated the effect of Marc1 deletion on liver phenotype. Unexpectedly, our study found that whole-body Marc1 deficiency in mouse is not protective against hepatic triglyceride accumulation, liver inflammation or fibrosis. In attempts to explain the lack of the observed phenotype, we discovered that Marc1 plays only a minor role in mouse liver while its paralogue Marc2 is the main Marc family enzyme in mice. Our findings highlight the major difference in MARC1 physiological function between human and mouse.
Collapse
Affiliation(s)
- Eriks Smagris
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Lisa M. Shihanian
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Ivory J. Mintah
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Parnian Bigdelou
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Yuliya Livson
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Heather Brown
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Niek Verweij
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Charleen Hunt
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | | | - Tyler J. Greer
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | | | - George Hindy
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Luanluan Sun
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Jonas B. Nielsen
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Gabor Halasz
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Luca A. Lotta
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Andrew J. Murphy
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Mark W. Sleeman
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| | - Viktoria Gusarova
- Regeneron Pharmaceuticals, Tarrytown, New York, Unites States of America
| |
Collapse
|
19
|
Tarantino G, Citro V. What are the common downstream molecular events between alcoholic and nonalcoholic fatty liver? Lipids Health Dis 2024; 23:41. [PMID: 38331795 PMCID: PMC10851522 DOI: 10.1186/s12944-024-02031-1] [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: 11/05/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Liver fat storage, also called hepatic steatosis, is increasingly common and represents a very frequent diagnosis in the medical field. Excess fat is not without consequences. In fact, hepatic steatosis contributes to the progression toward liver fibrosis. There are two main types of fatty liver disease, alcoholic fatty liver disease (AFLD) and nonalcoholic fatty liver disease (NAFLD). Although AFLD and NAFLD are similar in their initial morphological features, both conditions involve the same evolutive forms. Moreover, there are various common mechanisms underlying both diseases, including alcoholic liver disease and NAFLD, which are commonalities. In this Review, the authors explore similar downstream signaling events involved in the onset and progression of the two entities but not completely different entities, predominantly focusing on the gut microbiome. Downstream molecular events, such as the roles of sirtuins, cytokeratins, adipokines and others, should be considered. Finally, to complete the feature, some new tendencies in the therapeutic approach are presented.
Collapse
Affiliation(s)
| | - Vincenzo Citro
- Department of General Medicine, Umberto I Hospital, Nocera Inferiore, SA, 84014, Italy
| |
Collapse
|
20
|
Abstract
Alcohol-related liver disease (ALD) is a major cause of liver-related morbidity and mortality. Epidemiological trends indicate recent and predicted increases in the burden of disease. Disease progression is driven by continued alcohol exposure on a background of genetic predisposition together with environmental cofactors. Most individuals present with advanced disease despite a long history of excessive alcohol consumption and multiple missed opportunities to intervene. Increasing evidence supports the use of non-invasive tests to screen for and identify disease at earlier stages. There is a definite role for public health measures to reduce the overall burden of disease. At an individual level, however, the ability to influence subsequent disease course by modifying alcohol consumption or the underlying pathogenic mechanisms remains limited due to a comparative lack of effective, disease-modifying medical interventions. Abstinence from alcohol is the key determinant of outcome in established ALD and the cornerstone of clinical management. In those with decompensated ALD, liver transplant has a clear role. There is consensus that abstinence from alcohol for an arbitrary period should not be the sole determinant in a decision to transplant. An increasing understanding of the mechanisms by which alcohol causes liver disease in susceptible individuals offers the prospect of new therapeutic targets for disease-modifying drugs. Successful translation will require significant public and private investment in a disease area which has traditionally been underfunded when compared to its overall prevalence.
Collapse
Affiliation(s)
- Mark Thursz
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | | |
Collapse
|
21
|
Miteva D, Peshevska-Sekulovska M, Snegarova V, Peruhova M, Vasilev GH, Vasilev GV, Sekulovski M, Lazova S, Gulinac M, Tomov L, Mihova A, Velikova T. Microbiome and Genetic Factors in the Pathogenesis of Liver Diseases. GASTROENTEROLOGY INSIGHTS 2023; 14:575-597. [DOI: 10.3390/gastroent14040041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
Our genetic background has not changed over the past century, but chronic diseases are on the rise globally. In addition to the genetic component, among the critical factors for many diseases are inhabitants of our intestines (gut microbiota) as a crucial environmental factor. Dysbiosis has been described in liver diseases with different etiologies like non-alcoholic fatty liver disease (NAFLD), alcohol-related liver disease (ALD), viral hepatitis, autoimmune hepatitis (AIH), primary sclerosing cholangitis (PSC), primary biliary cholangitis (PBC), cirrhosis, hepatocellular carcinoma (HCC). On the other hand, new technologies have increased our understanding of liver disease genetics and treatment options. Genome-wide association studies (GWAS) identify unknown genetic risk factors, positional cloning of unknown genes associated with different diseases, gene tests for single nucleotide variations (SNVs), and next-generation sequencing (NGS) of selected genes or the complete genome. NGS also allowed studying the microbiome and its role in various liver diseases has begun. These genes have proven their effect on microbiome composition in host genome–microbiome association studies. We focus on altering the intestinal microbiota, and supplementing some bacterial metabolites could be considered a potential therapeutic strategy. The literature data promote probiotics/synbiotics role in reducing proinflammatory cytokines such as TNF-α and the interleukins (IL-1, IL-6, IL-8), therefore improving transaminase levels, hepatic steatosis, and NAFLD activity score. However, even though microbial therapy appears to be risk-free, evaluating side effects related to probiotics or synbiotics is imperative. In addition, safety profiles for long-term usage should be researched. Thus, this review focuses on the human microbiome and liver diseases, recent GWASs on liver disease, the gut-liver axis, and the associations with the microbiome and microbiome during/after liver disease therapy.
Collapse
Affiliation(s)
- Dimitrina Miteva
- Department of Genetics, Faculty of Biology, Sofia University St. Kliment Ohridski, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Monika Peshevska-Sekulovska
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Gastroenterology, University Hospital Lozenetz, Kozyak 1 Str., 1407 Sofia, Bulgaria
| | - Violeta Snegarova
- Clinic of Internal Diseases, Naval Hospital—Varna, Military Medical Academy, Medical Faculty, Medical University, Blvd. Hristo Smirnenski 3, 9000 Varna, Bulgaria
| | - Milena Peruhova
- Department of Gastroenterology, Heart and Brain Hospital, Zdrave 1 Str., 8000 Burgas, Bulgaria
| | - Georgi H. Vasilev
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Laboratory of Hematopathology and Immunology, National Specialized Hospital for Active Treatment of Hematological Diseases, “Plovdivsko Pole” Str. 6, 1756 Sofia, Bulgaria
| | - Georgi V. Vasilev
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Emergency Medicine and Clinic of Neurology, University Hospital “Sv. Georgi”, Blvd. Peshtersko Shose 66, 4000 Plovdiv, Bulgaria
| | - Metodija Sekulovski
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Anesthesiology and Intensive Care, University Hospital Lozenetz, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Snezhina Lazova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Pediatric Department, University Hospital “N. I. Pirogov”, 21 “General Eduard I. Totleben” Blvd, 1606 Sofia, Bulgaria
- Department of Healthcare, Faculty of Public Health, “Prof. Tsekomir Vodenicharov, MD, DSc”, Medical University of Sofia, Bialo More 8 Str., 1527 Sofia, Bulgaria
| | - Milena Gulinac
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of General and Clinical Pathology, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Latchezar Tomov
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of Informatics, New Bulgarian University, Montevideo 21 Str., 1618 Sofia, Bulgaria
| | - Antoaneta Mihova
- SMDL Ramus, Department of Immunology, Blvd. Kap. Spisarevski 26, 1527 Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str., 1407 Sofia, Bulgaria
| |
Collapse
|
22
|
Struwe MA, Scheidig AJ, Clement B. The mitochondrial amidoxime reducing component-from prodrug-activation mechanism to drug-metabolizing enzyme and onward to drug target. J Biol Chem 2023; 299:105306. [PMID: 37778733 PMCID: PMC10637980 DOI: 10.1016/j.jbc.2023.105306] [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: 06/19/2023] [Revised: 09/17/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023] Open
Abstract
The mitochondrial amidoxime-reducing component (mARC) is one of five known molybdenum enzymes in eukaryotes. mARC belongs to the MOSC domain superfamily, a large group of so far poorly studied molybdoenzymes. mARC was initially discovered as the enzyme activating N-hydroxylated prodrugs of basic amidines but has since been shown to also reduce a variety of other N-oxygenated compounds, for example, toxic nucleobase analogs. Under certain circumstances, mARC might also be involved in reductive nitric oxide synthesis through reduction of nitrite. Recently, mARC enzymes have received a lot of attention due to their apparent involvement in lipid metabolism and, in particular, because many genome-wide association studies have shown a common variant of human mARC1 to have a protective effect against liver disease. The mechanism linking mARC enzymes with lipid metabolism remains unknown. Here, we give a comprehensive overview of what is currently known about mARC enzymes, their substrates, structure, and apparent involvement in human disease.
Collapse
Affiliation(s)
- Michel A Struwe
- Zoologisches Institut - Strukturbiologie, Christian-Albrechts-Universität Kiel, Kiel, Germany; Pharmazeutisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany.
| | - Axel J Scheidig
- Zoologisches Institut - Strukturbiologie, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Bernd Clement
- Pharmazeutisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany
| |
Collapse
|
23
|
Kocas-Kilicarslan ZN, Cetin Z, Faccioli LAP, Motomura T, Amirneni S, Diaz-Aragon R, Florentino RM, Sun Y, Pla-Palacin I, Xia M, Miedel MT, Kurihara T, Hu Z, Ostrowska A, Wang Z, Constantine R, Li A, Taylor DL, Behari J, Soto-Gutierrez A, Tafaleng EN. Polymorphisms Associated With Metabolic Dysfunction-Associated Steatotic Liver Disease Influence the Progression of End-Stage Liver Disease. GASTRO HEP ADVANCES 2023; 3:67-77. [PMID: 38292457 PMCID: PMC10827334 DOI: 10.1016/j.gastha.2023.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
BACKGROUND AND AIMS Chronic liver injury that results in cirrhosis and end-stage liver disease (ESLD) causes more than 1 million deaths annually worldwide. Although the impact of genetic factors on the severity of metabolic dysfunction-associated steatotic liver disease (MASLD) and alcohol-related liver disease (ALD) has been previously studied, their contribution to the development of ESLD remains largely unexplored. METHODS We genotyped 6 MASLD-associated polymorphisms in healthy (n = 123), metabolic dysfunction-associated steatohepatitis (MASH) (n = 145), MASLD-associated ESLD (n = 72), and ALD-associated ESLD (n = 57) cohorts and performed multinomial logistic regression to determine the combined contribution of genetic, demographic, and clinical factors to the progression of ESLD. RESULTS Distinct sets of factors are associated with the progression to ESLD. The PNPLA3 rs738409:G and TM6SF2 rs58542926:T alleles, body mass index (BMI), age, and female sex were positively associated with progression from a healthy state to MASH. The PNPLA3 rs738409:G allele, age, male sex, and having type 2 diabetes mellitus were positively associated, while BMI was negatively associated with progression from MASH to MASLD-associated ESLD. The PNPLA3 rs738409:G and GCKR rs780094:T alleles, age, and male sex were positively associated, while BMI was negatively associated with progression from a healthy state to ALD-associated ESLD. The findings indicate that the PNPLA3 rs738409:G allele increases susceptibility to ESLD regardless of etiology, the TM6SF2 rs58542926:T allele increases susceptibility to MASH, and the GCKR rs780094:T allele increases susceptibility to ALD-associated ESLD. CONCLUSION The PNPLA3, TM6SF2, and GCKR minor alleles influence the progression of MASLD-associated or ALD-associated ESLD. Genotyping for these variants in MASLD and ALD patients can enhance risk assessment, prompting early interventions to prevent ESLD.
Collapse
Affiliation(s)
- Zehra N. Kocas-Kilicarslan
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zeliha Cetin
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lanuza A. P. Faccioli
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Takashi Motomura
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sriram Amirneni
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ricardo Diaz-Aragon
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rodrigo M. Florentino
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yiyue Sun
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- School of Medicine, Tsinghua University, Beijing, China
| | - Iris Pla-Palacin
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mengying Xia
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mark T. Miedel
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Takeshi Kurihara
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Zhiping Hu
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Alina Ostrowska
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zi Wang
- Department of Statistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Albert Li
- Discovery Life Sciences, Huntsville, Alabama
| | - D. Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jaideep Behari
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Alejandro Soto-Gutierrez
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania
| | - Edgar N. Tafaleng
- Department of Pathology, Center for Transcriptional Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
24
|
Stender S, Davey Smith G, Richardson TG. Genetic variation and elevated liver enzymes during childhood, adolescence and early adulthood. Int J Epidemiol 2023; 52:1341-1349. [PMID: 37105232 PMCID: PMC10555681 DOI: 10.1093/ije/dyad048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Genetic factors influence the risk of fatty liver disease (FLD) in adults. The aim of this study was to test if, and when, genetic risk factors known to affect FLD in adults begin to exert their deleterious effects during childhood, adolescence and early adulthood. METHODS We included up to 4018 British children and adolescents from the Avon Longitudinal Study of Parents and Children (ALSPAC) cohort. Three genetic variants known to associate robustly with FLD in adults (PNPLA3 rs738409, TM6SF2 rs58542926 and HSD17B13 rs72613567) were tested for association with plasma levels of alanine transaminase (ALT) and aspartate transaminase (AST) during childhood (mean age: 9.9 years), early adolescence (15.5 years), late adolescence (17.8 years), and early adulthood (24.5 years). We also tested the associations of a 17-variant score and whole-genome polygenic risk scores (PRS) derived from associations in adults with plasma ALT and AST at the same four time points. Associations with elastography-derived liver steatosis and fibrosis were tested in early adulthood. RESULTS Genetic risk factors for FLD (individually, combined into a 3-variant score, a 17-variant score and as a genome-wide PRS), were associated with higher liver enzymes, beginning in childhood and throughout adolescence and early adulthood. The ALT-increasing effects of the genetic risk variants became larger with increasing age. The ALT-PRS was associated with liver steatosis in early adulthood. No genetic associations with fibrosis were observed. CONCLUSIONS Genetic factors that promote FLD in adults associate with elevated liver enzymes already during childhood, and their effects get amplified with increasing age.
Collapse
Affiliation(s)
- Stefan Stender
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, University of Bristol, Bristol, UK
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
25
|
Chen Y, Du X, Kuppa A, Feitosa MF, Bielak LF, O'Connell JR, Musani SK, Guo X, Kahali B, Chen VL, Smith AV, Ryan KA, Eirksdottir G, Allison MA, Bowden DW, Budoff MJ, Carr JJ, Chen YDI, Taylor KD, Oliveri A, Correa A, Crudup BF, Kardia SLR, Mosley TH, Norris JM, Terry JG, Rotter JI, Wagenknecht LE, Halligan BD, Young KA, Hokanson JE, Washko GR, Gudnason V, Province MA, Peyser PA, Palmer ND, Speliotes EK. Genome-wide association meta-analysis identifies 17 loci associated with nonalcoholic fatty liver disease. Nat Genet 2023; 55:1640-1650. [PMID: 37709864 PMCID: PMC10918428 DOI: 10.1038/s41588-023-01497-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 08/07/2023] [Indexed: 09/16/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is common and partially heritable and has no effective treatments. We carried out a genome-wide association study (GWAS) meta-analysis of imaging (n = 66,814) and diagnostic code (3,584 cases versus 621,081 controls) measured NAFLD across diverse ancestries. We identified NAFLD-associated variants at torsin family 1 member B (TOR1B), fat mass and obesity associated (FTO), cordon-bleu WH2 repeat protein like 1 (COBLL1)/growth factor receptor-bound protein 14 (GRB14), insulin receptor (INSR), sterol regulatory element-binding transcription factor 1 (SREBF1) and patatin-like phospholipase domain-containing protein 2 (PNPLA2), as well as validated NAFLD-associated variants at patatin-like phospholipase domain-containing protein 3 (PNPLA3), transmembrane 6 superfamily 2 (TM6SF2), apolipoprotein E (APOE), glucokinase regulator (GCKR), tribbles homolog 1 (TRIB1), glycerol-3-phosphate acyltransferase (GPAM), mitochondrial amidoxime-reducing component 1 (MARC1), microsomal triglyceride transfer protein large subunit (MTTP), alcohol dehydrogenase 1B (ADH1B), transmembrane channel like 4 (TMC4)/membrane-bound O-acyltransferase domain containing 7 (MBOAT7) and receptor-type tyrosine-protein phosphatase δ (PTPRD). Implicated genes highlight mitochondrial, cholesterol and de novo lipogenesis as causally contributing to NAFLD predisposition. Phenome-wide association study (PheWAS) analyses suggest at least seven subtypes of NAFLD. Individuals in the top 10% and 1% of genetic risk have a 2.5-fold to 6-fold increased risk of NAFLD, cirrhosis and hepatocellular carcinoma. These genetic variants identify subtypes of NAFLD, improve estimates of disease risk and can guide the development of targeted therapeutics.
Collapse
Affiliation(s)
- Yanhua Chen
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Xiaomeng Du
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Annapurna Kuppa
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey R O'Connell
- Department of Endocrinology, Diabetes and Nutrition, University of Maryland - Baltimore, Baltimore, MD, USA
| | - Solomon K Musani
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Bratati Kahali
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Vincent L Chen
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Albert V Smith
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Kathleen A Ryan
- Department of Endocrinology, Diabetes and Nutrition, University of Maryland - Baltimore, Baltimore, MD, USA
| | | | - Matthew A Allison
- Department of Family Medicine, University of California San Diego, San Diego, CA, USA
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Matthew J Budoff
- Department of Internal Medicine, Lundquist Institute at Harbor-UCLA, Torrance, CA, USA
| | - John Jeffrey Carr
- Department of Radiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yii-Der I Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Antonino Oliveri
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Breland F Crudup
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Thomas H Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jill M Norris
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - James G Terry
- Department of Radiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Brian D Halligan
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Kendra A Young
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - John E Hokanson
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
| | - George R Washko
- Department of Medicine, Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Elizabeth K Speliotes
- Department of Internal Medicine, Division of Gastroenterology and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
26
|
Clement B, Struwe MA. The History of mARC. Molecules 2023; 28:4713. [PMID: 37375270 DOI: 10.3390/molecules28124713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
The mitochondrial amidoxime-reducing component (mARC) is the most recently discovered molybdoenzyme in humans after sulfite oxidase, xanthine oxidase and aldehyde oxidase. Here, the timeline of mARC's discovery is briefly described. The story begins with investigations into N-oxidation of pharmaceutical drugs and model compounds. Many compounds are N-oxidized extensively in vitro, but it turned out that a previously unknown enzyme catalyzes the retroreduction of the N-oxygenated products in vivo. After many years, the molybdoenzyme mARC could finally be isolated and identified in 2006. mARC is an important drug-metabolizing enzyme and N-reduction by mARC has been exploited very successfully for prodrug strategies, that allow oral administration of otherwise poorly bioavailable therapeutic drugs. Recently, it was demonstrated that mARC is a key factor in lipid metabolism and likely involved in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). The exact link between mARC and lipid metabolism is not yet fully understood. Regardless, many now consider mARC a potential drug target for the prevention or treatment of liver diseases. This article focusses on discoveries related to mammalian mARC enzymes. mARC homologues have been studied in algae, plants and bacteria. These will not be discussed extensively here.
Collapse
Affiliation(s)
- Bernd Clement
- Pharmazeutisches Institut, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118 Kiel, Germany
| | - Michel A Struwe
- Pharmazeutisches Institut, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118 Kiel, Germany
- Zoologisches Institut-Strukturbiologie, Zentrum für Biochemie und Molekularbiologie, Christian-Albrechts-Universität zu Kiel, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| |
Collapse
|
27
|
Lewis LC, Chen L, Hameed LS, Kitchen RR, Maroteau C, Nagarajan SR, Norlin J, Daly CE, Szczerbinska I, Hjuler ST, Patel R, Livingstone EJ, Durrant TN, Wondimu E, BasuRay S, Chandran A, Lee WH, Hu S, Gilboa B, Grandi ME, Toledo EM, Erikat AH, Hodson L, Haynes WG, Pursell NW, Coppieters K, Fleckner J, Howson JM, Andersen B, Ruby MA. Hepatocyte mARC1 promotes fatty liver disease. JHEP Rep 2023; 5:100693. [PMID: 37122688 PMCID: PMC10133763 DOI: 10.1016/j.jhepr.2023.100693] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 05/02/2023] Open
Abstract
Background & Aims Non-alcoholic fatty liver disease (NAFLD) has a prevalence of ∼25% worldwide, with significant public health consequences yet few effective treatments. Human genetics can help elucidate novel biology and identify targets for new therapeutics. Genetic variants in mitochondrial amidoxime-reducing component 1 (MTARC1) have been associated with NAFLD and liver-related mortality; however, its pathophysiological role and the cell type(s) mediating these effects remain unclear. We aimed to investigate how MTARC1 exerts its effects on NAFLD by integrating human genetics with in vitro and in vivo studies of mARC1 knockdown. Methods Analyses including multi-trait colocalisation and Mendelian randomisation were used to assess the genetic associations of MTARC1. In addition, we established an in vitro long-term primary human hepatocyte model with metabolic readouts and used the Gubra Amylin NASH (GAN)-diet non-alcoholic steatohepatitis mouse model treated with hepatocyte-specific N-acetylgalactosamine (GalNAc)-siRNA to understand the in vivo impacts of MTARC1. Results We showed that genetic variants within the MTARC1 locus are associated with liver enzymes, liver fat, plasma lipids, and body composition, and these associations are attributable to the same causal variant (p.A165T, rs2642438 G>A), suggesting a shared mechanism. We demonstrated that increased MTARC1 mRNA had an adverse effect on these traits using Mendelian randomisation, implying therapeutic inhibition of mARC1 could be beneficial. In vitro mARC1 knockdown decreased lipid accumulation and increased triglyceride secretion, and in vivo GalNAc-siRNA-mediated knockdown of mARC1 lowered hepatic but increased plasma triglycerides. We found alterations in pathways regulating lipid metabolism and decreased secretion of 3-hydroxybutyrate upon mARC1 knockdown in vitro and in vivo. Conclusions Collectively, our findings from human genetics, and in vitro and in vivo hepatocyte-specific mARC1 knockdown support the potential efficacy of hepatocyte-specific targeting of mARC1 for treatment of NAFLD. Impact and implications We report that genetically predicted increases in MTARC1 mRNA associate with poor liver health. Furthermore, knockdown of mARC1 reduces hepatic steatosis in primary human hepatocytes and a murine NASH model. Together, these findings further underscore the therapeutic potential of targeting hepatocyte MTARC1 for NAFLD.
Collapse
Affiliation(s)
| | | | | | | | | | - Shilpa R. Nagarajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
| | | | | | | | | | - Rahul Patel
- Novo Nordisk Research Centre Oxford, Oxford, UK
| | | | | | | | | | | | - Wan-Hung Lee
- Dicerna Pharmaceuticals Inc., Lexington, MA, USA
| | - Sile Hu
- Novo Nordisk Research Centre Oxford, Oxford, UK
| | | | | | | | | | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford, UK
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital Trusts, Oxford, UK
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Zawistowski M, Fritsche LG, Pandit A, Vanderwerff B, Patil S, Schmidt EM, VandeHaar P, Willer CJ, Brummett CM, Kheterpal S, Zhou X, Boehnke M, Abecasis GR, Zöllner S. The Michigan Genomics Initiative: A biobank linking genotypes and electronic clinical records in Michigan Medicine patients. CELL GENOMICS 2023; 3:100257. [PMID: 36819667 PMCID: PMC9932985 DOI: 10.1016/j.xgen.2023.100257] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/07/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023]
Abstract
Biobanks of linked clinical patient histories and biological samples are an efficient strategy to generate large cohorts for modern genetics research. Biobank recruitment varies by factors such as geographic catchment and sampling strategy, which affect biobank demographics and research utility. Here, we describe the Michigan Genomics Initiative (MGI), a single-health-system biobank currently consisting of >91,000 participants recruited primarily during surgical encounters at Michigan Medicine. The surgical enrollment results in a biobank enriched for many diseases and ideally suited for a disease genetics cohort. Compared with the much larger population-based UK Biobank, MGI has higher prevalence for nearly all diagnosis-code-based phenotypes and larger absolute case counts for many phenotypes. Genome-wide association study (GWAS) results replicate known findings, thereby validating the genetic and clinical data. Our results illustrate that opportunistic biobank sampling within single health systems provides a unique and complementary resource for exploring the genetics of complex diseases.
Collapse
Affiliation(s)
- Matthew Zawistowski
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Lars G. Fritsche
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Anita Pandit
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Brett Vanderwerff
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Snehal Patil
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Ellen M. Schmidt
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Peter VandeHaar
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Cristen J. Willer
- Department of Internal Medicine, Division of Cardiovascular Medicine, Department of Human Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Chad M. Brummett
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Sachin Kheterpal
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Xiang Zhou
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Gonçalo R. Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
- Regeneron Genetics Center, Tarrytown, NY 10591, USA
| | - Sebastian Zöllner
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48103, USA
| |
Collapse
|
29
|
Beyoğlu D, Huang P, Skelton-Badlani D, Zong C, Popov YV, Idle JR. Metabolic Hijacking of Hexose Metabolism to Ascorbate Synthesis Is the Unifying Biochemical Basis of Murine Liver Fibrosis. Cells 2023; 12:cells12030485. [PMID: 36766828 PMCID: PMC9914390 DOI: 10.3390/cells12030485] [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: 01/12/2023] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
We wished to understand the metabolic reprogramming underlying liver fibrosis progression in mice. Administration to male C57BL/6J mice of the hepatotoxins carbon tetrachloride (CCl4), thioacetamide (TAA), or a 60% high-fat diet, choline-deficient, amino-acid-defined diet (HF-CDAA) was conducted using standard protocols. Livers collected at different times were analyzed by gas chromatography-mass spectrometry-based metabolomics. RNA was extracted from liver and assayed by qRT-PCR for mRNA expression of 11 genes potentially involved in the synthesis of ascorbic acid from hexoses, Gck, Adpgk, Hk1, Hk2, Ugp2, Ugdh, Ugt1a1, Akr1a4, Akr1b3, Rgn and Gulo. All hepatotoxins resulted in similar metabolic changes during active fibrogenesis, despite different etiology and resultant scarring pattern. Diminished hepatic glucose, galactose, fructose, pentose phosphate pathway intermediates, glucuronic acid and long-chain fatty acids were compensated by elevated ascorbate and the product of collagen prolyl 4-hydroxylase, succinate and its downstream metabolites fumarate and malate. Recovery from the HF-CDAA diet challenge (F2 stage fibrosis) after switching to normal chow was accompanied by increased glucose, galactose, fructose, ribulose 5-phosphate, glucuronic acid, the ascorbate metabolite threonate and diminished ascorbate. During the administration of CCl4, TAA and HF-CDAA, aldose reductase Akr1b3 transcription was induced six- to eightfold, indicating increased conversion of glucuronic acid to gulonic acid, a precursor of ascorbate synthesis. Triggering hepatic fibrosis by three independent mechanisms led to the hijacking of glucose and galactose metabolism towards ascorbate synthesis, to satisfy the increased demand for ascorbate as a cofactor for prolyl 4-hydroxylase for mature collagen production. This metabolic reprogramming and causal gene expression changes were reversible. The increased flux in this pathway was mediated predominantly by increased transcription of aldose reductase Akr1b3.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
- Arthur G. Zupko Institute for Systems Pharmacology and Pharmacogenomics, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA
| | - Pinzhu Huang
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Disha Skelton-Badlani
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Christine Zong
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Yury V. Popov
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Jeffrey R. Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
- Arthur G. Zupko Institute for Systems Pharmacology and Pharmacogenomics, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Correspondence: ; Tel.: +1-929-888-6534
| |
Collapse
|
30
|
Smyk W, Papapostoli I, Żorniak M, Sklavounos P, Blukacz Ł, Madej P, Koutsou A, Weber SN, Friesenhahn-Ochs B, Cebula M, Bosowska J, Solomayer EF, Hartleb M, Milkiewicz P, Lammert F, Stokes CS, Krawczyk M. Liver phenotypes in PCOS: Analysis of exogenous and inherited risk factors for liver injury in two European cohorts. Liver Int 2023; 43:1080-1088. [PMID: 36683562 DOI: 10.1111/liv.15527] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
BACKGROUND & AIMS Fatty liver disease (FLD) is common in women with polycystic ovary syndrome (PCOS). Here, we use non-invasive tests to quantify liver injury in women with PCOS and analyse whether FLD-associated genetic variants contribute to liver phenotypes in PCOS. METHODS Prospectively, we recruited women with PCOS and controls at two university centres in Germany and Poland. Alcohol abuse was regarded as an exclusion criterion. Genotyping of variants associated with FLD was performed using TaqMan assays. Liver stiffness measurements (LSM), controlled attenuation parameters (CAP) and non-invasive HSI, FLI, FIB-4 scores were determined to assess hepatic steatosis and fibrosis. RESULTS A total of 42 German (age range 18-53 years) and 143 Polish (age range 18-40 years) women with PCOS, as well as 245 German and 289 Polish controls were recruited. In contrast to Polish patients, Germans were older, presented with more severe metabolic profiles and had significantly higher LSM (median 5.9 kPa vs. 3.8 kPa). In the German cohort, carriers of the PNPLA3 p.I148M risk variant had an increased LSM (p = .01). In the Polish cohort, the minor MTARC1 allele was linked with significantly lower serum aminotransferases activities, whereas the HSD17B13 polymorphism was associated with lower concentrations of 17-OH progesterone, total testosterone, and androstenedione (all p < .05). CONCLUSIONS FLD is common in women with PCOS. Its extent is modulated by both genetic and metabolic risk factors. Genotyping of variants associated with FLD might help to stratify the risk of liver disease progression in women suffering from PCOS.
Collapse
Affiliation(s)
- Wiktor Smyk
- Department of Gastroenterology and Hepatology, Medical University of Gdansk, Gdansk, Poland
| | - Ifigeneia Papapostoli
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Michał Żorniak
- Department of Gastroenterology, Hepatology and Oncology, Center for Postgraduate Medical Education, Warsaw, Poland.,Department of Oncological Gastroenterology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Panagiotis Sklavounos
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Łukasz Blukacz
- Department of Gynecological Endocrinology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Paweł Madej
- Department of Gynecological Endocrinology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Andreani Koutsou
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Susanne N Weber
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Bettina Friesenhahn-Ochs
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Maciej Cebula
- Department of Radiology and Nuclear Medicine, Faculty of Medicine in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Joanna Bosowska
- Department of Radiology and Nuclear Medicine, Faculty of Medicine in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Erich-Franz Solomayer
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Marek Hartleb
- Department of Gastroenterology, Faculty of Medicine in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Medical University of Warsaw, Warsaw, Poland.,Translational Medicine Group, Pomeranian Medical University, Szczecin, Poland
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.,Hannover Medical School, Hannover, Germany
| | - Caroline S Stokes
- Food and Health Research Group, Faculty of Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Molecular Toxicology, German Institute of Human Nutrition, Potsdam-Rehbrücke, Germany
| | - Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.,Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
31
|
Kalinowski P, Smyk W, Nowosad M, Paluszkiewicz R, Michałowski Ł, Ziarkiewicz-Wróblewska B, Weber SN, Milkiewicz P, Lammert F, Zieniewicz K, Krawczyk M. MTARC1 and HSD17B13 Variants Have Protective Effects on Non-Alcoholic Fatty Liver Disease in Patients Undergoing Bariatric Surgery. Int J Mol Sci 2022; 23:ijms232415825. [PMID: 36555467 PMCID: PMC9781679 DOI: 10.3390/ijms232415825] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
The severity of hepatic steatosis is modulated by genetic variants, such as patatin-like phospholipase domain containing 3 (PNPLA3) rs738409, transmembrane 6 superfamily member 2 (TM6SF2) rs58542926, and membrane-bound O-acyltransferase domain containing 7 (MBOAT7) rs641738. Recently, mitochondrial amidoxime reducing component 1 (MTARC1) rs2642438 and hydroxysteroid 17-beta dehydrogenase 13 (HSD17B13) rs72613567 polymorphisms were shown to have protective effects on liver diseases. Here, we evaluate these variants in patients undergoing bariatric surgery. A total of 165 patients who underwent laparoscopic sleeve gastrectomy and intraoperative liver biopsies and 314 controls were prospectively recruited. Genotyping was performed using TaqMan assays. Overall, 70.3% of operated patients presented with hepatic steatosis. NASH (non-alcoholic steatohepatitis) was detected in 28.5% of patients; none had cirrhosis. The increment of liver fibrosis stage was associated with decreasing frequency of the MTARC1 minor allele (p = 0.03). In multivariate analysis MTARC1 was an independent protective factor against fibrosis ≥ 1b (OR = 0.52, p = 0.03) and ≥ 1c (OR = 0.51, p = 0.04). The PNPLA3 risk allele was associated with increased hepatic steatosis, fibrosis, and NASH (OR = 2.22, p = 0.04). The HSD17B13 polymorphism was protective against liver injury as reflected by lower AST (p = 0.04) and ALT (p = 0.03) activities. The TM6SF2 polymorphism was associated with increased ALT (p = 0.04). In conclusion, hepatic steatosis is common among patients scheduled for bariatric surgery, but the MTARC1 and HSD17B13 polymorphisms lower liver injury in these individuals.
Collapse
Affiliation(s)
- Piotr Kalinowski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Wiktor Smyk
- Department of Gastroenterology and Hepatology, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Małgorzata Nowosad
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Rafał Paluszkiewicz
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Łukasz Michałowski
- Department of Pathology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | | | - Susanne N. Weber
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany
- Hannover Health Science Campus, Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Krzysztof Zieniewicz
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Saarland University, 66421 Homburg, Germany
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
- Correspondence: ; Tel.: +49-684-1116-15000
| |
Collapse
|
32
|
Association of MARC1, ADCY5, and BCO1 Variants with the Lipid Profile, Suggests an Additive Effect for Hypertriglyceridemia in Mexican Adult Men. Int J Mol Sci 2022; 23:ijms231911815. [PMID: 36233117 PMCID: PMC9569691 DOI: 10.3390/ijms231911815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/22/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Epidemiological studies have reported that the Mexican population is highly susceptible to dyslipidemia. The MARC1, ADCY5, and BCO1 genes have recently been involved in lipidic abnormalities. This study aimed to analyze the association of single nucleotide polymorphisms (SNPs) rs2642438, rs56371916, and rs6564851 on MARC1, ADCY5, and BCO1 genes, respectively, with the lipid profile in a cohort of Mexican adults. We included 1900 Mexican adults from the Health Workers Cohort Study. Demographic and clinical data were collected through a structured questionnaire and standardized procedures. Genotyping was performed using a predesigned TaqMan assay. A genetic risk score (GRS) was created on the basis of the three genetic variants. Associations analysis was estimated using linear and logistic regression. Our results showed that rs2642438-A and rs6564851-A alleles had a risk association for hypertriglyceridemia (OR = 1.57, p = 0.013; and OR = 1.33, p = 0.031, respectively), and rs56371916-C allele a trend for low HDL-c (OR = 1.27, p = 0.060) only in men. The GRS revealed a significant association for hypertriglyceridemia (OR = 2.23, p = 0.022). These findings provide evidence of an aggregate effect of the MARC1, ADCY5, and BCO1 variants on the risk of hypertriglyceridemia in Mexican men. This knowledge could represent a tool for identifying at-risk males who might benefit from early interventions and avoid secondary metabolic traits.
Collapse
|
33
|
Sasaki-Tanaka R, Ray R, Moriyama M, Ray RB, Kanda T. Molecular Changes in Relation to Alcohol Consumption and Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms23179679. [PMID: 36077080 PMCID: PMC9456124 DOI: 10.3390/ijms23179679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 12/12/2022] Open
Abstract
Alcohol is the one of the major causes of liver diseases and promotes liver cirrhosis and hepatocellular carcinoma (HCC). In hepatocytes, alcohol is converted to acetaldehyde, which causes hepatic steatosis, cellular apoptosis, endoplasmic reticulum stress, peroxidation, production of cytokines and reduces immune surveillance. Endotoxin and lipopolysaccharide produced from intestinal bacteria also enhance the production of cytokines. The development of hepatic fibrosis and the occurrence of HCC are induced by these alcohol metabolites. Several host genetic factors have recently been identified in this process. Here, we reviewed the molecular mechanism associated with HCC in alcoholic liver disease.
Collapse
Affiliation(s)
- Reina Sasaki-Tanaka
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
- Correspondence: (R.S.-T.); (T.K.); Tel.: +81-3-3972-8111 (R.S.-T. & T.K.)
| | - Ranjit Ray
- Departments of Internal Medicine, and Molecular Microbiology and Immunology, Saint Louis University, Saint Louis, MO 63104, USA
| | - Mitsuhiko Moriyama
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Ratna B. Ray
- Department of Pathology, Saint Louis University, Saint Louis, MO 63104, USA
| | - Tatsuo Kanda
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
- Correspondence: (R.S.-T.); (T.K.); Tel.: +81-3-3972-8111 (R.S.-T. & T.K.)
| |
Collapse
|
34
|
Hudert CA, Adams LA, Alisi A, Anstee QM, Crudele A, Draijer LG, Furse S, Hengstler JG, Jenkins B, Karnebeek K, Kelly DA, Koot BG, Koulman A, Meierhofer D, Melton PE, Mori TA, Snowden SG, van Mourik I, Vreugdenhil A, Wiegand S, Mann JP. Variants in mitochondrial amidoxime reducing component 1 and hydroxysteroid 17-beta dehydrogenase 13 reduce severity of nonalcoholic fatty liver disease in children and suppress fibrotic pathways through distinct mechanisms. Hepatol Commun 2022; 6:1934-1948. [PMID: 35411667 PMCID: PMC9315139 DOI: 10.1002/hep4.1955] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/19/2022] [Indexed: 12/14/2022] Open
Abstract
Genome-wide association studies in adults have identified variants in hydroxysteroid 17-beta dehydrogenase 13 (HSD17B13) and mitochondrial amidoxime reducing component 1 (MTARC1) as protective against nonalcoholic fatty liver disease (NAFLD). We aimed to test their association with pediatric NAFLD liver histology and investigate their function using metabolomics. A total of 1450 children (729 with NAFLD, 399 with liver histology) were genotyped for rs72613567T>TA in HSD17B13, rs2642438G>A in MTARC1, and rs738409C>G in patatin-like phospholipase domain-containing protein 3 (PNPLA3). Genotype-histology associations were tested using ordinal regression. Untargeted hepatic proteomics and plasma lipidomics were performed in a subset of children. We found rs72613567T>TA in HSD17B13 to be associated with lower odds of NAFLD diagnosis (odds ratio, 0.7; 95% confidence interval, 0.6-0.9) and a lower grade of portal inflammation (p < 0.001). rs2642438G>A in MTARC1 was associated with a lower grade of hepatic steatosis (p = 0.02). Proteomics found reduced expression of HSD17B13 in carriers of the protective -TA allele. MTARC1 levels were unaffected by genotype. Both variants were associated with down-regulation of fibrogenic pathways. HSD17B13 perturbs plasma phosphatidylcholines and triglycerides. In silico modeling suggested p.Ala165Thr disrupts the stability and metal binding of MTARC1. Conclusion: Both HSD17B13 and MTARC1 variants are associated with less severe pediatric NAFLD. These results provide further evidence for shared genetic mechanisms between pediatric and adult NAFLD.
Collapse
Affiliation(s)
- Christian A. Hudert
- Department of Pediatric Gastroenterology, Nephrology and Metabolic DiseasesCharité Universitätsmedizin BerlinBerlinGermany
| | - Leon A. Adams
- Medical SchoolUniversity of Western AustraliaPerthAustralia
- Department of HepatologySir Charles Gairdner HospitalPerthAustralia
| | - Anna Alisi
- Research Unit of Molecular Genetics of Complex PhenotypesBambino Gesù Children's Hospital‐Istituto di Ricovero e Cura a Carattere ScientificoRomeItaly
| | - Quentin M. Anstee
- Translational and Clinical Research InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle upon TyneUK
- Newcastle National Institute for Health Research Biomedical Research CentreNewcastle upon Tyne Hospitals National Health Service Foundation TrustNewcastle upon TyneUK
| | - Annalisa Crudele
- Research Unit of Molecular Genetics of Complex PhenotypesBambino Gesù Children's Hospital‐Istituto di Ricovero e Cura a Carattere ScientificoRomeItaly
| | - Laura G. Draijer
- Department of Pediatric Gastroenterology and NutritionAmsterdam University Medical CenterEmma Children’s HospitalUniversity of AmsterdamAmsterdamthe Netherlands
| | | | - Samuel Furse
- Core Metabolomics and Lipidomics LaboratoryWellcome Trust–Medical Research Council Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - Jan G. Hengstler
- Systems ToxicologyLeibniz Research Center for Working Environment and Human Factors at the Technical University DortmundDortmundGermany
| | - Benjamin Jenkins
- Core Metabolomics and Lipidomics LaboratoryWellcome Trust–Medical Research Council Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - Kylie Karnebeek
- Center for Overweight Adolescent and Children's Health CareDepartment of PediatricsMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Deirdre A. Kelly
- Liver UnitBirmingham Womens and Children’s Hospital TrustBirminghamUK
| | - Bart G. Koot
- Department of Pediatric Gastroenterology and NutritionAmsterdam University Medical CenterEmma Children’s HospitalUniversity of AmsterdamAmsterdamthe Netherlands
| | - Albert Koulman
- Core Metabolomics and Lipidomics LaboratoryWellcome Trust–Medical Research Council Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - David Meierhofer
- Max Planck Institute for Molecular GeneticsMass Spectrometry FacilityBerlinGermany
| | - Phillip E. Melton
- School of Global Population HealthFaculty of Health and Medical SciencesUniversity of Western AustraliaPerthAustralia
- School of Pharmacy and Biomedical SciencesFaculty of Health SciencesCurtin UniversityPerthAustralia
- Menzies Institute for Medical ResearchCollege of Health and MedicineUniversity of TasmaniaHobartAustralia
| | - Trevor A. Mori
- Medical SchoolUniversity of Western AustraliaPerthAustralia
| | - Stuart G. Snowden
- Core Metabolomics and Lipidomics LaboratoryWellcome Trust–Medical Research Council Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| | - Indra van Mourik
- Liver UnitBirmingham Womens and Children’s Hospital TrustBirminghamUK
| | - Anita Vreugdenhil
- Center for Overweight Adolescent and Children's Health CareDepartment of PediatricsMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Susanna Wiegand
- Center for Chronically Sick ChildrenCharité Universitätsmedizin BerlinBerlinGermany
| | - Jake P. Mann
- Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
| |
Collapse
|
35
|
Mann JP, Hoare M. A minority of somatically mutated genes in pre-existing fatty liver disease have prognostic importance in the development of NAFLD. Liver Int 2022; 42:1823-1835. [PMID: 35474605 PMCID: PMC9544140 DOI: 10.1111/liv.15283] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Understanding the genetics of liver disease has the potential to facilitate clinical risk stratification. We recently identified acquired somatic mutations in six genes and one lncRNA in pre-existing fatty liver disease. We hypothesised that germline variation in these genes might be associated with the risk of developing steatosis and contribute to the prediction of disease severity. METHODS Genome-wide association study (GWAS) summary statistics were extracted from seven studies (>1.7 million participants) for variants near ACVR2A, ALB, CIDEB, FOXO1, GPAM, NEAT1 and TNRC6B for: aminotransferases, liver fat, HbA1c, diagnosis of NAFLD, ARLD and cirrhosis. Findings were replicated using GWAS data from multiple independent cohorts. A phenome-wide association study was performed to examine for related metabolic traits, using both common and rare variants, including gene-burden testing. RESULTS There was no evidence of association between rare germline variants or SNPs near five genes (ACVR2A, ALB, CIDEB, FOXO1 and TNRC6B) and risk or severity of liver disease. Variants in GPAM (proxies for p.Ile43Val) were associated with liver fat (p = 3.6 × 10-13 ), ALT (p = 2.8 × 10-39 ) and serum lipid concentrations. Variants in NEAT1 demonstrated borderline significant associations with ALT (p = 1.9 × 10-11 ) and HbA1c, but not with liver fat, as well as influencing waist-to-hip ratio, adjusted for BMI. CONCLUSIONS Despite the acquisition of somatic mutations at these loci during progressive fatty liver disease, we did not find associations between germline variation and markers of liver disease, except in GPAM. In the future, larger sample sizes may identify associations. Currently, germline polygenic risk scores will not capture data from genes affected by somatic mutations.
Collapse
Affiliation(s)
- Jake P. Mann
- Institute of Metabolic ScienceUniversity of CambridgeCambridgeUK
- School of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Matthew Hoare
- School of Clinical MedicineUniversity of CambridgeCambridgeUK
- CRUK Cambridge InstituteUniversity of CambridgeCambridgeUK
| |
Collapse
|
36
|
Hardesty J, Day L, Warner J, Warner D, Gritsenko M, Asghar A, Stolz A, Morgan T, McClain C, Jacobs J, Kirpich I. Hepatic Protein and Phosphoprotein Signatures of Alcohol-Associated Cirrhosis and Hepatitis. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1066-1082. [PMID: 35490715 PMCID: PMC9253914 DOI: 10.1016/j.ajpath.2022.04.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022]
Abstract
Alcohol-associated liver disease is a global health care burden, with alcohol-associated cirrhosis (AC) and alcohol-associated hepatitis (AH) being two clinical manifestations with poor prognosis. The limited efficacy of standard of care for AC and AH highlights a need for therapeutic targets and strategies. The current study aimed to address this need through the identification of hepatic proteome and phosphoproteome signatures of AC and AH. Proteomic and phosphoproteomic analyses were conducted on explant liver tissue (test cohort) and liver biopsies (validation cohort) from patients with AH. Changes in protein expression across AH severity and similarities and differences in AH and AC hepatic proteome were analyzed. Significant alterations in multiple proteins involved in various biological processes were observed in both AC and AH, including elevated expression of transcription factors involved in fibrogenesis (eg, Yes1-associated transcriptional regulator). Another finding was elevated levels of hepatic albumin (ALBU) concomitant with diminished ALBU phosphorylation, which may prevent ALBU release, leading to hypoalbuminemia. Furthermore, altered expression of proteins related to neutrophil function and chemotaxis, including elevated myeloperoxidase, cathelicidin antimicrobial peptide, complement C3, and complement C5 were observed in early AH, which declined at later stages. Finally, a loss in expression of mitochondria proteins, including enzymes responsible for the synthesis of cardiolipin was observed. The current study identified hepatic protein signatures of AC and AH as well as AH severity, which may facilitate the development of therapeutic strategies.
Collapse
Affiliation(s)
- Josiah Hardesty
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky; Department of Pharmacology and Toxicology, University of Louisville Alcohol Center, Louisville, Kentucky
| | - Le Day
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington
| | - Jeffrey Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky; Department of Pharmacology and Toxicology, University of Louisville Alcohol Center, Louisville, Kentucky
| | - Dennis Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Marina Gritsenko
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington
| | - Aliya Asghar
- Gastroenterology, VA Long Beach Healthcare, VA Long Beach Healthcare System, Long Beach, California
| | - Andrew Stolz
- Division of Gastrointestinal and Liver Disease, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Timothy Morgan
- Gastroenterology, VA Long Beach Healthcare, VA Long Beach Healthcare System, Long Beach, California
| | - Craig McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky; Department of Pharmacology and Toxicology, University of Louisville Alcohol Center, Louisville, Kentucky; Robley Rex Veterans Medical Center, Louisville, Kentucky; University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, Kentucky; University of Louisville Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, Kentucky
| | - Jon Jacobs
- Biological Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington
| | - Irina Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky; Department of Pharmacology and Toxicology, University of Louisville Alcohol Center, Louisville, Kentucky; University of Louisville Alcohol Center, University of Louisville School of Medicine, Louisville, Kentucky; University of Louisville Hepatobiology and Toxicology Center, University of Louisville School of Medicine, Louisville, Kentucky.
| |
Collapse
|
37
|
Liu K, Yip TCF, Masson S, Fateen W, Schwantes‐An T, McCaughan GW, Morgan TR, Aithal GP, Seth D. Validation of the
aMAP
score to predict hepatocellular carcinoma development in a cohort of alcohol‐related cirrhosis patients. LIVER CANCER INTERNATIONAL 2022; 3:99-104. [DOI: 10.1002/lci2.47] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Affiliation(s)
- Ken Liu
- AW Morrow Gastroenterology and Liver Unit Royal Prince Alfred Hospital Sydney New South Wales Australia
- Sydney Medical School University of Sydney Sydney New South Wales Australia
- Centenary Institute of Cancer Medicine and Cell Biology The University of Sydney Sydney New South Wales Australia
| | - Terry C. F. Yip
- Department of Medicine and Therapeutics, Medical Data Analytics Centre, and Institute of Digestive Disease The Chinese University of Hong Kong Hong Kong SAR China
| | - Steven Masson
- Faculty of Medical Sciences Newcastle University Medical School, Framlington Place and Liver Transplant Unit, Freeman Hospital Newcastle upon Tyne UK
| | - Waleed Fateen
- NIHR Nottingham Biomedical Research Centre Nottingham University Hospitals and the University of Nottingham Nottingham UK
| | - Tae‐Hwi Schwantes‐An
- Department of Medical and Molecular Genetics Indiana University Indianapolis Indiana USA
| | - Geoffrey W. McCaughan
- AW Morrow Gastroenterology and Liver Unit Royal Prince Alfred Hospital Sydney New South Wales Australia
- Sydney Medical School University of Sydney Sydney New South Wales Australia
- Centenary Institute of Cancer Medicine and Cell Biology The University of Sydney Sydney New South Wales Australia
| | - Timothy R. Morgan
- Medical and Research Services VA Long Beach Healthcare System Long Beach California USA
| | - Guruprasad P. Aithal
- NIHR Nottingham Biomedical Research Centre Nottingham University Hospitals and the University of Nottingham Nottingham UK
| | - Devanshi Seth
- Sydney Medical School University of Sydney Sydney New South Wales Australia
- Centenary Institute of Cancer Medicine and Cell Biology The University of Sydney Sydney New South Wales Australia
- Edith Collins Centre Translational Research in Alcohol Drugs & Toxicology Sydney Local Health District Camperdown New South Wales Australia
| |
Collapse
|
38
|
Zhu XB, Hou YQ, Ye XY, Zou YX, Xia XS, Yang S, Huang P, Yu RB. Identifying and Exploring the Candidate Susceptibility Genes of Cirrhosis Using the Multi-Tissue Transcriptome-Wide Association Study. Front Genet 2022; 13:878607. [PMID: 35646080 PMCID: PMC9136150 DOI: 10.3389/fgene.2022.878607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/30/2022] [Indexed: 12/03/2022] Open
Abstract
Objective: We identify and explore the candidate susceptibility genes for cirrhosis and their underlying biological mechanism. Methods: We downloaded the genome-wide association studies summary data of 901 cirrhosis cases and 451,363 controls and integrated them with reference models of five potential tissues from the Genotype-Tissue Expression (GTEx) Project, including whole blood, liver, pancreas, spleen, and thyroid, to identify genes whose expression is predicted to be associated with cirrhosis. Then, we downloaded gene expression data of individuals with hepatocellular carcinoma from TCGA database to conduct differential expression analysis to validate these identified genes and explored their possible role in driving cirrhosis via functional enrichment and gene set enrichment analysis (GSEA). Results: We identified 10 significant genes (SKIV2L, JPH4, UQCC2, RP11-91I8.3, MAU2, ERAP1, PUS3, ZNF677, ARHGAP40, and SHANK3) associated with cirrhosis at a Bonferroni-corrected threshold of p < 0.01, among which two (SKIV2L and JPH4) were identified in the liver and five (SKIV2L, JPH4, MAU2, SHANK3, and UQCC2) were validated by differential expression analysis at an FDR-corrected threshold of p < 0.01. The enrichment analysis showed that the degradation process of RNA, which is enriched by 58 genes, is significantly under-enriched in liver cancer tissues (p = 0.0268). Conclusion: We have identified several candidate genes for cirrhosis in multiple tissues and performed differential genetic analysis using the liver cancer database to verify the significant genes. We found that the genes SKIV2L and JPH4 identified in the liver are of particular concern. Finally, through enrichment analysis, we speculate that the process of mRNA transcription and RNA degradation may play a role in cirrhosis.
Collapse
Affiliation(s)
- Xiao-Bo Zhu
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, China
| | - Yu-Qing Hou
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiang-Yu Ye
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yi-Xin Zou
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xue-Shan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Sheng Yang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Peng Huang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- *Correspondence: Peng Huang, ; Rong-Bin Yu,
| | - Rong-Bin Yu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- *Correspondence: Peng Huang, ; Rong-Bin Yu,
| |
Collapse
|
39
|
Innes H, Nischalke HD, Guha IN, Weiss KH, Irving W, Gotthardt D, Barnes E, Fischer J, Ansari MA, Rosendahl J, Lin S, Marot A, Pedergnana V, Casper M, Benselin J, Lammert F, McLauchlan J, Lutz PL, Hamill V, Mueller S, Morling JR, Semmler G, Eyer F, von Felden J, Link A, Vogel A, Marquardt JU, Sulk S, Trebicka J, Valenti L, Datz C, Reiberger T, Schafmayer C, Berg T, Deltenre P, Hampe J, Stickel F, Buch S. The rs429358 Locus in Apolipoprotein E Is Associated With Hepatocellular Carcinoma in Patients With Cirrhosis. Hepatol Commun 2022; 6:1213-1226. [PMID: 34958182 PMCID: PMC9035556 DOI: 10.1002/hep4.1886] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/02/2021] [Accepted: 11/24/2021] [Indexed: 12/17/2022] Open
Abstract
The host genetic background for hepatocellular carcinoma (HCC) is incompletely understood. We aimed to determine if four germline genetic polymorphisms, rs429358 in apolipoprotein E (APOE), rs2642438 in mitochondrial amidoxime reducing component 1 (MARC1), rs2792751 in glycerol-3-phosphate acyltransferase (GPAM), and rs187429064 in transmembrane 6 superfamily member 2 (TM6SF2), previously associated with progressive alcohol-related and nonalcoholic fatty liver disease, are also associated with HCC. Four HCC case-control data sets were constructed, including two mixed etiology data sets (UK Biobank and FinnGen); one hepatitis C virus (HCV) cohort (STOP-HCV), and one alcohol-related HCC cohort (Dresden HCC). The frequency of each variant was compared between HCC cases and cirrhosis controls (i.e., patients with cirrhosis without HCC). Population controls were also considered. Odds ratios (ORs) associations were calculated using logistic regression, adjusting for age, sex, and principal components of genetic ancestry. Fixed-effect meta-analysis was used to determine the pooled effect size across all data sets. Across four case-control data sets, 2,070 HCC cases, 4,121 cirrhosis controls, and 525,779 population controls were included. The rs429358:C allele (APOE) was significantly less frequent in HCC cases versus cirrhosis controls (OR, 0.71; 95% confidence interval [CI], 0.61-0.84; P = 2.9 × 10-5 ). Rs187429064:G (TM6SF2) was significantly more common in HCC cases versus cirrhosis controls and exhibited the strongest effect size (OR, 2.03; 95% CI, 1.45-2.86; P = 3.1 × 10-6 ). In contrast, rs2792751:T (GPAM) was not associated with HCC (OR, 1.01; 95% CI, 0.90-1.13; P = 0.89), whereas rs2642438:A (MARC1) narrowly missed statistical significance (OR, 0.91; 95% CI, 0.84-1.00; P = 0.043). Conclusion: This study associates carriage of rs429358:C (APOE) with a reduced risk of HCC in patients with cirrhosis. Conversely, carriage of rs187429064:G in TM6SF2 is associated with an increased risk of HCC in patients with cirrhosis.
Collapse
Affiliation(s)
- Hamish Innes
- School of Health and Life SciencesGlasgow Caledonian UniversityGlasgowUnited Kingdom
- Population and Lifespan SciencesSchool of MedicineUniversity of NottinghamNottinghamUnited Kingdom
- Public Health ScotlandGlasgowUnited Kingdom
| | | | - Indra Neil Guha
- National Institute for Health Research (NIHR), Nottingham Biomedical Research CentreNottingham University Hospitals National Health Service Trust and the University of NottinghamNottinghamUnited Kingdom
| | - Karl Heinz Weiss
- Department of Gastroenterology and HepatologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Will Irving
- National Institute for Health Research (NIHR), Nottingham Biomedical Research CentreNottingham University Hospitals National Health Service Trust and the University of NottinghamNottinghamUnited Kingdom
| | - Daniel Gotthardt
- Department of Internal Medicine IVMedical University of HeidelbergHeidelbergGermany
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen ResearchNuffield Department of Medicine and the Oxford NIHR Biomedical Research CentreOxford UniversityOxfordUnited Kingdom
| | - Janett Fischer
- Division of HepatologyDepartment of Medicine IILaboratory for Clinical and Experimental HepatologyLeipzig University Medical CenterLeipzigGermany
| | - M. Azim Ansari
- Peter Medawar Building for Pathogen ResearchNuffield Department of Medicine and the Oxford NIHR Biomedical Research CentreOxford UniversityOxfordUnited Kingdom
| | - Jonas Rosendahl
- Medical Department 1University Hospital HalleMartin‐Luther Universität Halle‐WittenbergHalleGermany
| | - Shang‐Kuan Lin
- Peter Medawar Building for Pathogen ResearchNuffield Department of Medicine and the Oxford NIHR Biomedical Research CentreOxford UniversityOxfordUnited Kingdom
| | - Astrid Marot
- Division of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversité de LausanneLausanneSwitzerland
- Department of Gastroenterology and HepatologyCentre Hospitalier UniversitaireUCLouvain NamurUniversité Catholique de LouvainYvoirBelgium
| | | | - Markus Casper
- Department of Medicine IISaarland University Medical CenterSaarland UniversityHomburgGermany
| | - Jennifer Benselin
- National Institute for Health Research (NIHR), Nottingham Biomedical Research CentreNottingham University Hospitals National Health Service Trust and the University of NottinghamNottinghamUnited Kingdom
| | - Frank Lammert
- Department of Medicine IISaarland University Medical CenterSaarland UniversityHomburgGermany
| | - John McLauchlan
- Medical Research Council‐University of Glasgow Centre for Virus ResearchGlasgowUnited Kingdom
| | - Philip L. Lutz
- Department of Internal Medicine IUniversity HospitalUniversity of BonnBonnGermany
| | - Victoria Hamill
- School of Health and Life SciencesGlasgow Caledonian UniversityGlasgowUnited Kingdom
- Public Health ScotlandGlasgowUnited Kingdom
| | - Sebastian Mueller
- Center for Alcohol ResearchUniversity of HeidelbergHeidelbergGermany
- Medical DepartmentSalem Medical CenterHeidelbergGermany
| | - Joanne R. Morling
- Population and Lifespan SciencesSchool of MedicineUniversity of NottinghamNottinghamUnited Kingdom
- National Institute for Health Research (NIHR), Nottingham Biomedical Research CentreNottingham University Hospitals National Health Service Trust and the University of NottinghamNottinghamUnited Kingdom
| | - Georg Semmler
- Department of Internal Medicine IIIDivision of Gastroenterology and HepatologyMedical University of ViennaViennaAustria
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University SalzburgOberndorfAustria
| | - Florian Eyer
- Department of Clinical ToxicologyKlinikum Rechts der IsarTechnical University of MunichMunichGermany
| | - Johann von Felden
- Department of MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Alexander Link
- Department of Gastroenterology, Hepatology, and Infectious DiseasesOtto‐von‐Guericke University HospitalMagdeburgGermany
| | - Arndt Vogel
- Department of Gastroenterology, Hepatology, and EndocrinologyHannover Medical SchoolHannoverGermany
| | - Jens U. Marquardt
- Department of Medicine IUniversity Hospital Schleswig Holstein–Campus LübeckLübeckGermany
| | - Stefan Sulk
- Medical Department 1University Hospital DresdenTechnische Universität DresdenDresdenGermany
| | - Jonel Trebicka
- Department of Internal Medicine IGoethe UniversityFrankfurtGermany
- European Foundation for Study of Chronic Liver FailureBarcelonaSpain
| | - Luca Valenti
- Precision Medicine–Department of Transfusion Medicine and HematologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Christian Datz
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University SalzburgOberndorfAustria
| | - Thomas Reiberger
- Department of Internal Medicine IIIDivision of Gastroenterology and HepatologyMedical University of ViennaViennaAustria
| | - Clemens Schafmayer
- Department of General, Visceral, Vascular, and Transplant SurgeryRostock University Medical CenterRostockGermany
| | - Thomas Berg
- Division of HepatologyDepartment of Medicine IILaboratory for Clinical and Experimental HepatologyLeipzig University Medical CenterLeipzigGermany
| | - Pierre Deltenre
- Division of Gastroenterology and HepatologyCentre Hospitalier Universitaire VaudoisUniversité de LausanneLausanneSwitzerland
- Department of GastroenterologyHepatopancreatology, and Digestive OncologyUniversity Clinics of Brussels Hospital ErasmeBrusselsBelgium
- Department of Gastroenterology and HepatologyClinique St LucBougeBelgium
| | - Jochen Hampe
- Medical Department 1University Hospital DresdenTechnische Universität DresdenDresdenGermany
- Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
| | - Felix Stickel
- Department of Gastroenterology and HepatologyUniversity Hospital of ZurichZurichSwitzerland
| | - Stephan Buch
- Medical Department 1University Hospital DresdenTechnische Universität DresdenDresdenGermany
| |
Collapse
|
40
|
Di Ciaula A, Bonfrate L, Krawczyk M, Frühbeck G, Portincasa P. Synergistic and Detrimental Effects of Alcohol Intake on Progression of Liver Steatosis. Int J Mol Sci 2022; 23:ijms23052636. [PMID: 35269779 PMCID: PMC8910376 DOI: 10.3390/ijms23052636] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD) are the most common liver disorders worldwide and the major causes of non-viral liver cirrhosis in the general population. In NAFLD, metabolic abnormalities, obesity, and metabolic syndrome are the driving factors for liver damage with no or minimal alcohol consumption. ALD refers to liver damage caused by excess alcohol intake in individuals drinking more than 5 to 10 daily units for years. Although NAFLD and ALD are nosologically considered two distinct entities, they show a continuum and exert synergistic effects on the progression toward liver cirrhosis. The current view is that low alcohol use might also increase the risk of advanced clinical liver disease in NAFLD, whereas metabolic factors increase the risk of cirrhosis among alcohol risk drinkers. Therefore, special interest is now addressed to individuals with metabolic abnormalities who consume small amounts of alcohol or who binge drink, for the role of light-to-moderate alcohol use in fibrosis progression and clinical severity of the liver disease. Evidence shows that in the presence of NAFLD, there is no liver-safe limit of alcohol intake. We discuss the epidemiological and clinical features of NAFLD/ALD, aspects of alcohol metabolism, and mechanisms of damage concerning steatosis, fibrosis, cumulative effects, and deleterious consequences which include hepatocellular carcinoma.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “Augusto Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School—Piazza Giulio Cesare 11, 70124 Bari, Italy; (A.D.C.); (L.B.)
| | - Leonilde Bonfrate
- Clinica Medica “Augusto Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School—Piazza Giulio Cesare 11, 70124 Bari, Italy; (A.D.C.); (L.B.)
| | - Marcin Krawczyk
- Department of Medicine II Saarland University Medical Center, Saarland University, 66424 Homburg, Germany;
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Gema Frühbeck
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, 31008 Pamplona, Spain;
- Metabolic Research Laboratory, Clínica Universidad de Navarra, 31008 Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), ISCIII, 31009 Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31009 Pamplona, Spain
| | - Piero Portincasa
- Clinica Medica “Augusto Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School—Piazza Giulio Cesare 11, 70124 Bari, Italy; (A.D.C.); (L.B.)
- Correspondence:
| |
Collapse
|
41
|
Innes H, Walker AJ, Benselin J, Grove JI, Pedergnana V, Azim Ansari M, Lin SK, McLauchlan J, Hutchinson SJ, Barnes E, Irving WL, Guha IN. Comprehensive Comparative Analysis of Standard Validated, Genetic, and Novel Biomarkers to Enhance Prognostic Risk-Stratification in Patients With Hepatitis C Virus Cirrhosis. Clin Transl Gastroenterol 2022; 13:e00462. [PMID: 35142723 PMCID: PMC8963831 DOI: 10.14309/ctg.0000000000000462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 12/06/2021] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Risk-stratifying patients with hepatitis C virus (HCV) cirrhosis according to medium-term prognosis will inform clinical decision-making. It is unclear which biomarkers/models are optimal for this purpose. We quantified the discriminative ability of 14 diverse biomarkers for prognosis prediction over a 4-year time. METHODS We recruited 1196 patients with HCV cirrhosis from the United Kingdom for a prospective study. Genetic risk score, collagen (e.g., PROC3), comorbidity (e.g., CirCom), and validated biomarkers from routine data were measured at enrollment. Participants were linked to UK hospital admission, cancer, and mortality registries. Primary endpoints were (i) liver-related outcomes for patients with compensated cirrhosis and (ii) all-cause mortality for decompensated cirrhosis. The discriminative ability of all biomarkers was quantified individually and also by the fraction of new prognostic information provided. RESULTS At enrollment, 289 (24%) and 907 (76%) had decompensated and compensated cirrhosis, respectively. Participants were followed for 3-4 years on average, with >70% of the follow-up time occurring post-HCV cure. Seventy-five deaths in the decompensated subgroup and 98 liver-related outcomes in the compensated subgroup were reported. The discriminative ability of the albumin-bilirubin-fibrosis-4 index (C-index: 0.71-0.72) was superior to collagen biomarkers (C-index = 0.58-0.67), genetic risk scores (C-index = 0.50-0.57), and comorbidity markers (0.53-0.60). Validated biomarkers showed the greatest prognostic improvement when combined with a comorbidity or a collagen biomarker (generally >30% of new prognostic information added). DISCUSSION Inexpensive biomarkers such as the albumin-bilirubin-fibrosis-4 index predict medium-term cirrhosis prognosis moderately well and outperform collagen, genetic, and comorbidity biomarkers. Improvement of performance was greatest when a validated test was combined with comorbidity or collagen biomarker.
Collapse
Affiliation(s)
- Hamish Innes
- School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
- Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK
- Public Health Scotland, Glasgow, UK
| | - Alex J. Walker
- The DataLab, Centre for Evidence-Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, UK
| | - Jennifer Benselin
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Jane I. Grove
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Vincent Pedergnana
- Laboratoire MIVEGEC (UMR CNRS 5290, UR IRD 224, UM), Montpellier, France
| | - M. Azim Ansari
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine and the Oxford NIHR Biomedical Research Centre, Oxford University, UK
| | - Shang-Kuan Lin
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine and the Oxford NIHR Biomedical Research Centre, Oxford University, UK
| | - John McLauchlan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Sharon J. Hutchinson
- School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
- Public Health Scotland, Glasgow, UK
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine and the Oxford NIHR Biomedical Research Centre, Oxford University, UK
| | - William L. Irving
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Indra Neil Guha
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| |
Collapse
|
42
|
Whitfield JB, Schwantes-An TH, Darlay R, Aithal GP, Atkinson SR, Bataller R, Botwin G, Chalasani NP, Cordell HJ, Daly AK, Day CP, Eyer F, Foroud T, Gleeson D, Goldman D, Haber PS, Jacquet JM, Liang T, Liangpunsakul S, Masson S, Mathurin P, Moirand R, McQuillin A, Moreno C, Morgan MY, Mueller S, Müllhaupt B, Nagy LE, Nahon P, Nalpas B, Naveau S, Perney P, Pirmohamed M, Seitz HK, Soyka M, Stickel F, Thompson A, Thursz MR, Trépo E, Morgan TR, Seth D. A genetic risk score and diabetes predict development of alcohol-related cirrhosis in drinkers. J Hepatol 2022; 76:275-282. [PMID: 34656649 PMCID: PMC8803006 DOI: 10.1016/j.jhep.2021.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/06/2021] [Accepted: 10/05/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Only a minority of excess alcohol drinkers develop cirrhosis. We developed and evaluated risk stratification scores to identify those at highest risk. METHODS Three cohorts (GenomALC-1: n = 1,690, GenomALC-2: n = 3,037, UK Biobank: relevant n = 6,898) with a history of heavy alcohol consumption (≥80 g/day (men), ≥50 g/day (women), for ≥10 years) were included. Cases were participants with alcohol-related cirrhosis. Controls had a history of similar alcohol consumption but no evidence of liver disease. Risk scores were computed from up to 8 genetic loci identified previously as associated with alcohol-related cirrhosis and 3 clinical risk factors. Score performance for the stratification of alcohol-related cirrhosis risk was assessed and compared across the alcohol-related liver disease spectrum, including hepatocellular carcinoma (HCC). RESULTS A combination of 3 single nucleotide polymorphisms (SNPs) (PNPLA3:rs738409, SUGP1-TM6SF2:rs10401969, HSD17B13:rs6834314) and diabetes status best discriminated cirrhosis risk. The odds ratios (ORs) and (95% CIs) between the lowest (Q1) and highest (Q5) score quintiles of the 3-SNP score, based on independent allelic effect size estimates, were 5.99 (4.18-8.60) (GenomALC-1), 2.81 (2.03-3.89) (GenomALC-2), and 3.10 (2.32-4.14) (UK Biobank). Patients with diabetes and high risk scores had ORs of 14.7 (7.69-28.1) (GenomALC-1) and 17.1 (11.3-25.7) (UK Biobank) compared to those without diabetes and with low risk scores. Patients with cirrhosis and HCC had significantly higher mean risk scores than patients with cirrhosis alone (0.76 ± 0.06 vs. 0.61 ± 0.02, p = 0.007). Score performance was not significantly enhanced by information on additional genetic risk variants, body mass index or coffee consumption. CONCLUSIONS A risk score based on 3 genetic risk variants and diabetes status enables the stratification of heavy drinkers based on their risk of cirrhosis, allowing for the provision of earlier preventative interventions. LAY SUMMARY Excessive chronic drinking leads to cirrhosis in some people, but so far there is no way to identify those at high risk of developing this debilitating disease. We developed a genetic risk score that can identify patients at high risk. The risk of cirrhosis is increased >10-fold with just two risk factors - diabetes and a high genetic risk score. Risk assessment using this test could enable the early and personalised management of this disease in high-risk patients.
Collapse
Affiliation(s)
- John B Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Queensland 4029, Australia.
| | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis IN, USA
| | - Rebecca Darlay
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Guruprasad P Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals and the University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Stephen R Atkinson
- Department of Metabolism, Digestion & Reproduction, Imperial College London, UK
| | - Ramon Bataller
- Center for Liver Diseases, University of Pittsburgh Medical Center, 3471 Fifth Avenue, Pittsburgh, PA 15213, USA
| | - Greg Botwin
- Department of Veterans Affairs, VA Long Beach Healthcare System, 5901 East Seventh Street, Long Beach, CA 90822, USA; F. Widjaja Family Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California CA 90048, USA
| | - Naga P Chalasani
- Department of Medicine, Indiana University, Indianapolis, Indiana, IN 46202-5175, USA
| | - Heather J Cordell
- Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Ann K Daly
- Faculty of Medical Sciences, Newcastle University Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Christopher P Day
- Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Florian Eyer
- Division of Clinical Toxicology, Department of Internal Medicine 2, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis IN, USA
| | - Dermot Gleeson
- Liver Unit, Sheffield Teaching Hospitals, AO Floor Robert Hadfield Building, Northern General Hospital, Sheffied S5 7AU, UK
| | - David Goldman
- Laboratory of Neurogenetics, NIAAA, Rockville, MD 20852, USA
| | - Paul S Haber
- Drug Health Services, Royal Prince Alfred Hospital, Missenden Road, Camperdown, NSW 2050, Australia; Faculty of Medicine and Health, the University of Sydney, Sydney, NSW 2006, Australia
| | | | - Tiebing Liang
- Department of Medicine, Indiana University, Indianapolis, Indiana, IN 46202-5175, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University and Roudebush Veterans Administration Medical Center, Indianapolis, USA
| | - Steven Masson
- Faculty of Medical Sciences, Newcastle University Medical School, Framlington Place, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Philippe Mathurin
- CHRU de Lille, Hôpital Claude Huriez, Rue M. Polonovski CS 70001, 59 037 Lille Cedex, France
| | - Romain Moirand
- Univ Rennes, INRA, INSERM, CHU Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer), F-35000 Rennes, France
| | - Andrew McQuillin
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College London, London WC1E 6DE, UK
| | - Christophe Moreno
- CUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium; Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Marsha Y Morgan
- UCL Institute for Liver & Digestive Health, Division of Medicine, Royal Free Campus, University College London, London NW3 2PF, UK
| | - Sebastian Mueller
- Department of Internal Medicine, Salem Medical Center and Center for Alcohol Research, University of Heidelberg, Zeppelinstraße 11-33, 69121 Heidelberg, Germany
| | - Beat Müllhaupt
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, CH-8901 Zurich, Switzerland
| | - Laura E Nagy
- Lerner Research Institute, 9500 Euclid Avenue, Cleveland, Ohio, OH 44195, USA
| | - Pierre Nahon
- Service d'Hépatologie, APHP Hôpital Avicenne et Université Paris 13, Bobigny, France; University Paris 13, Bobigny, France; Inserm U1162 Génomique fonctionnelle des tumeurs solides, Paris, France
| | - Bertrand Nalpas
- Service Addictologie, CHRU Caremeau, 30029 Nîmes, France; DISC, Inserm, 75013 Paris, France
| | - Sylvie Naveau
- Hôpital Antoine-Béclère, 157 Rue de la Porte de Trivaux, 92140 Clamart, France
| | - Pascal Perney
- Hôpital Universitaire Caremeau, Place du Pr. Robert Debre, 30029 Nîmes, France
| | - Munir Pirmohamed
- MRC Centre for Drug Safety Science and Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GL, UK
| | - Helmut K Seitz
- Department of Internal Medicine, Salem Medical Center and Center for Alcohol Research, University of Heidelberg, Zeppelinstraße 11-33, 69121 Heidelberg, Germany
| | - Michael Soyka
- Psychiatric Hospital University of Munich, Nussbaumsstr.7, 80336 Munich, Germany; Privatklinik Meiringen, Willigen, CH 3860 Meiringen, Switzerland
| | - Felix Stickel
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Rämistrasse 100, CH-8901 Zurich, Switzerland
| | - Andrew Thompson
- MRC Centre for Drug Safety Science and Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GL, UK; Health Analytics, Lane Clark & Peacock LLP, London, UK
| | - Mark R Thursz
- Department of Metabolism, Digestion & Reproduction, Imperial College London, UK
| | - Eric Trépo
- CUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium; Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Timothy R Morgan
- Department of Veterans Affairs, VA Long Beach Healthcare System, 5901 East Seventh Street, Long Beach, CA 90822, USA; Department of Medicine, University of California, Irvine, USA
| | - Devanshi Seth
- Drug Health Services, Royal Prince Alfred Hospital, Missenden Road, Camperdown, NSW 2050, Australia; Faculty of Medicine and Health, the University of Sydney, Sydney, NSW 2006, Australia; Centenary Institute of Cancer Medicine and Cell Biology, the University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
43
|
Wei YG, Yang CK, Wei ZL, Liao XW, He YF, Zhou X, Huang HS, Lan CL, Han CY, Peng T. High-Mobility Group AT-Hook 1 Served as a Prognosis Biomarker and Associated with Immune Infiltrate in Hepatocellular Carcinoma. Int J Gen Med 2022; 15:609-621. [PMID: 35058711 PMCID: PMC8765458 DOI: 10.2147/ijgm.s344858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/23/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The protein high-mobility group AT-hook 1 (HMGA1) has been demonstrated that modulated cellular proliferation, invasion, and apoptosis with a poor prognosis in miscellaneous carcinomas. However, the mechanism of circumstantial carcinogenesis and association with the immune microenvironment of HMGA1 in hepatocellular carcinoma (HCC) had not been extensively explored. METHODS The gene expression, clinicopathological correlation, and prognosis analysis were performed in the data obtained from TCGA. The results were further validated by ICGC and GEO database and external validation cohort from Guangxi. The HMGA1 protein expression was further examined in the HPA database. Biological function analyses were conducted by GSEA, STRING database, and Coexpedia online tool. Using TIMER and CIBERSORT method, the relationship between immune infiltrate and HMGA1 was investigated. RESULTS In HCC, HMGA1 had much higher transcriptional and proteomic expression than in corresponding paraneoplastic tissue. Patients with high HMGA1 expression had a poor prognosis and unpromising clinicopathological features. High HMGA1 expression was closely related to the cell cycle, tumorigenesis, substance metabolism, and immune processes by regulating complex signaling pathways. Notably, HMGA1 may be associated with TP53 mutational carcinogenesis. Moreover, increased HMGA1 expression may lead to an increase in immune infiltration and a decrease in tumor purity in HCC. CIBERSORT analysis elucidated that the amount of B cell naive, B cell memory, T cells gamma delta, macrophages M2, and mast cell resting decreased when HMGA1 expression was high, whereas T cells follicular helper, macrophages M0, and Dendritic cells resting increased. CONCLUSION In conclusions, HMGA1 is a potent prognostic biomarker and a sign of immune infiltration in HCC, which may be a potential immunotherapy target for HCC.
Collapse
Affiliation(s)
- Yong-Guang Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Cheng-Kun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Zhong-Liu Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xi-Wen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Yong-Fei He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Hua-Sheng Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Chen-Lu Lan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Chuang-Ye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| |
Collapse
|
44
|
Zhu X, Xia M, Gao X. Update on genetics and epigenetics in metabolic associated fatty liver disease. Ther Adv Endocrinol Metab 2022; 13:20420188221132138. [PMID: 36325500 PMCID: PMC9619279 DOI: 10.1177/20420188221132138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/25/2022] [Indexed: 11/06/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is becoming the most frequent chronic liver disease worldwide. Metabolic (dysfunction) associated fatty liver disease (MAFLD) is suggested to replace the nomenclature of NAFLD. For individuals with metabolic dysfunction, multiple NAFLD-related factors also contribute to the development and progression of MAFLD including genetics and epigenetics. The application of genome-wide association study (GWAS) and exome-wide association study (EWAS) uncovers single-nucleotide polymorphisms (SNPs) in MAFLD. In addition to the classic SNPs in PNPLA3, TM6SF2, and GCKR, some new SNPs have been found recently to contribute to the pathogenesis of liver steatosis. Epigenetic factors involving DNA methylation, histone modifications, non-coding RNAs regulations, and RNA methylation also play a critical role in MAFLD. DNA methylation is the most reported epigenetic modification. Developing a non-invasion biomarker to distinguish metabolic steatohepatitis (MASH) or liver fibrosis is ongoing. In this review, we summarized and discussed the latest progress in genetic and epigenetic factors of NAFLD/MAFLD, in order to provide potential clues for MAFLD treatment.
Collapse
Affiliation(s)
- Xiaopeng Zhu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| | | | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Schnabl B, Arteel GE, Stickel F, Hengstler J, Vartak N, Ghallab A, Dooley S, Li Y, Schwabe RF. Liver specific, systemic and genetic contributors to alcohol-related liver disease progression. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:36-44. [PMID: 35042252 PMCID: PMC8941985 DOI: 10.1055/a-1714-9330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Alcohol-related liver disease (ALD) impacts millions of patients worldwide each year and the numbers are increasing. Disease stages range from steatosis via steatohepatitis and fibrosis to cirrhosis, severe alcohol-associated hepatitis and liver cancer. ALD is usually diagnosed at an advanced stage of progression with no effective therapies. A major research goal is to improve diagnosis, prognosis and also treatments for early ALD. This however needs prioritization of this disease for financial investment in basic and clinical research to more deeply investigate mechanisms and identify biomarkers and therapeutic targets for early detection and intervention. Topics of interest are communication of the liver with other organs of the body, especially the gut microbiome, the individual genetic constitution, systemic and liver innate inflammation, including bacterial infections, as well as fate and number of hepatic stellate cells and the composition of the extracellular matrix in the liver. Additionally, mechanical forces and damaging stresses towards the sophisticated vessel system of the liver, including the especially equipped sinusoidal endothelium and the biliary tract, work together to mediate hepatocytic import and export of nutritional and toxic substances, adapting to chronic liver disease by morphological and functional changes. All the aforementioned parameters contribute to the outcome of alcohol use disorder and the risk to develop advanced disease stages including cirrhosis, severe alcoholic hepatitis and liver cancer. In the present collection, we summarize current knowledge on these alcohol-related liver disease parameters, excluding the aspect of inflammation, which is presented in the accompanying review article by Lotersztajn and colleagues.
Collapse
Affiliation(s)
- Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, United States
- Department of Medicine, VA San Diego Healthcare System, San Diego, United States
| | - Gavin E Arteel
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, United States
- Pittsburgh Liver Research Center, Pittsburgh, United States
| | - Felix Stickel
- Department of Gastroenterology and Hepatology, University Hospital Zürich, Zürich, Switzerland
| | - Jan Hengstler
- Systems Toxicology, Leibniz Research Centre for Working Environment and Human Factors, TU Dortmund University, Dortmund, Germany
| | - Nachiket Vartak
- Systems Toxicology, Leibniz Research Centre for Working Environment and Human Factors, TU Dortmund University, Dortmund, Germany
| | - Ahmed Ghallab
- Systems Toxicology, Leibniz Research Centre for Working Environment and Human Factors, TU Dortmund University, Dortmund, Germany
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yujia Li
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, United States
| |
Collapse
|
46
|
Janik MK, Smyk W, Kruk B, Szczepankiewicz B, Górnicka B, Lebiedzińska-Arciszewska M, Potes Y, Simões ICM, Weber SN, Lammert F, Więckowski MR, Milkiewicz P, Krawczyk M. MARC1 p.A165T variant is associated with decreased markers of liver injury and enhanced antioxidant capacity in autoimmune hepatitis. Sci Rep 2021; 11:24407. [PMID: 34949757 PMCID: PMC8702547 DOI: 10.1038/s41598-021-03521-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
The clinical picture of autoimmune hepatitis (AIH) varies markedly between patients, potentially due to genetic modifiers. The aim of this study was to evaluate genetic variants previously associated with fatty liver as potential modulators of the AIH phenotype. The study cohort comprised 313 non-transplanted adults with AIH. In all patients, the MARC1 (rs2642438), HSD17B13 (rs72613567), PNPLA3 (rs738409), TM6SF2 (rs58542926), and MBOAT7 (rs641738) variants were genotyped using TaqMan assays. Mitochondrial damage markers in serum were analyzed in relation to the MARC1 variant. Carriers of the protective MARC1 allele had lower ALT and AST (both P < 0.05). In patients treated for AIH for ≥ 6 months, MARC1 correlated with reduced AST, ALP, GGT (all P ≤ 0.01), and lower APRI (P = 0.02). Patients carrying the protective MARC1 genotype had higher total antioxidant activity (P < 0.01) and catalase levels (P = 0.02) in serum. The PNPLA3 risk variant was associated with higher MELD (P = 0.02) in treated patients, whereas MBOAT7 increased the odds for liver cancer (OR = 3.71). None of the variants modulated the risk of death or transplantation. In conclusion, the MARC1 polymorphism has protective effects in AIH. Genotyping of MARC1, PNPLA3, and MBOAT7 polymorphisms might help to stratify patients with AIH.
Collapse
|
47
|
Haas ME, Pirruccello JP, Friedman SN, Wang M, Emdin CA, Ajmera VH, Simon TG, Homburger JR, Guo X, Budoff M, Corey KE, Zhou AY, Philippakis A, Ellinor PT, Loomba R, Batra P, Khera AV. Machine learning enables new insights into genetic contributions to liver fat accumulation. CELL GENOMICS 2021; 1:100066. [PMID: 34957434 PMCID: PMC8699145 DOI: 10.1016/j.xgen.2021.100066] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/13/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022]
Abstract
Excess liver fat, called hepatic steatosis, is a leading risk factor for end-stage liver disease and cardiometabolic diseases but often remains undiagnosed in clinical practice because of the need for direct imaging assessments. We developed an abdominal MRI-based machine-learning algorithm to accurately estimate liver fat (correlation coefficients, 0.97-0.99) from a truth dataset of 4,511 middle-aged UK Biobank participants, enabling quantification in 32,192 additional individuals. 17% of participants had predicted liver fat levels indicative of steatosis, and liver fat could not have been reliably estimated based on clinical factors such as BMI. A genome-wide association study of common genetic variants and liver fat replicated three known associations and identified five newly associated variants in or near the MTARC1, ADH1B, TRIB1, GPAM, and MAST3 genes (p < 3 × 10-8). A polygenic score integrating these eight genetic variants was strongly associated with future risk of chronic liver disease (hazard ratio > 1.32 per SD score, p < 9 × 10-17). Rare inactivating variants in the APOB or MTTP genes were identified in 0.8% of individuals with steatosis and conferred more than 6-fold risk (p < 2 × 10-5), highlighting a molecular subtype of hepatic steatosis characterized by defective secretion of apolipoprotein B-containing lipoproteins. We demonstrate that our imaging-based machine-learning model accurately estimates liver fat and may be useful in epidemiological and genetic studies of hepatic steatosis.
Collapse
Affiliation(s)
- Mary E. Haas
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Molecular Biology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - James P. Pirruccello
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Samuel N. Friedman
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Minxian Wang
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Connor A. Emdin
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Veeral H. Ajmera
- NAFLD Research Center, Department of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Tracey G. Simon
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Liver Center, Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Xiuqing Guo
- The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Matthew Budoff
- The Lundquist Institute for Biomedical Innovation and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Kathleen E. Corey
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Liver Center, Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Anthony Philippakis
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Patrick T. Ellinor
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, University of California San Diego, La Jolla, CA 92103, USA
| | - Puneet Batra
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Amit V. Khera
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Machine Learning for Health, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
48
|
Edwards AC, Sundquist K, Sundquist J, Kendler KS, Lönn SL. Genetic and environmental influences on the progression from alcohol use disorder to alcohol-related medical conditions. Alcohol Clin Exp Res 2021; 45:2528-2535. [PMID: 34923650 PMCID: PMC8712390 DOI: 10.1111/acer.14731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/27/2021] [Accepted: 10/21/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Medical conditions related to alcohol use disorders (AUD) represent a substantial public health concern. However, only a subset of individuals with AUD develop these conditions and the extent to which genetic and environmental factors that are shared with AUD, versus those distinct from it, contribute to this progression has not yet been determined. METHODS Using data from Swedish national registries for a cohort born from 1932 to 1970 (N = 1,319,214, 48.9% women), we conducted twin-sibling biometric model fitting to examine the genetic and environmental sources of variance that contribute to the liability to alcohol-related medical conditions (AMC). Progression to AMC, determined using medical registry data, was contingent on an AUD registration, which was determined using medical and criminal registry data. RESULTS We identified AUD registrations in 3.2% of women and 9.2% of men. Among individuals with an AUD registration, 14.4% of women and 15.4% of men had an AMC registration. In the final models, we constrained the beta pathway from AUD to AMC and the genetic and unique environmental paths to be equal across sexes. The beta path was estimated at 0.59. AMC was modestly heritable in women (A = 0.32) and men (A = 0.30). The proportion of total heritability unique to AMC was 39.6% among women and 41.3% among men. A higher proportion of total environmental variance was unique to AMC: 76.7% for women and 77.2% for men. In a sensitivity analysis limited to liver-related AMC, we observed similar results, with a slightly lower beta path from AUD to AMC (0.46) and higher proportions of AMC-specific genetic (70.0% in women; 71.7% in men) and environmental (84.5% in both sexes) variance. CONCLUSIONS A moderate-to-substantial proportion of genetic and environmental variance that contributes to AMC risk is not shared with AUD, underscoring the need for additional gene identification efforts for AMC. Furthermore, the prominent influence of environmental factors specific to AMC provides a promising area for the identification of prevention targets. We did not observe significant sex differences in the etiology of AMC, although follow-up is warranted in other well-powered studies.
Collapse
Affiliation(s)
- Alexis C. Edwards
- Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA, US
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, US
| | | | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Kenneth S. Kendler
- Department of Psychiatry, Virginia Commonwealth University School of Medicine, Richmond, VA, US
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, US
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA, US
| | - Sara Larsson Lönn
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| |
Collapse
|
49
|
Meroni M, Longo M, Tria G, Dongiovanni P. Genetics Is of the Essence to Face NAFLD. Biomedicines 2021; 9:1359. [PMID: 34680476 PMCID: PMC8533437 DOI: 10.3390/biomedicines9101359] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the commonest cause of chronic liver disease worldwide. It is closely related to obesity, insulin resistance (IR) and dyslipidemia so much so it is considered the hepatic manifestation of the Metabolic Syndrome. The NAFLD spectrum extends from simple steatosis to nonalcoholic steatohepatitis (NASH), a clinical condition which may progress up to fibrosis, cirrhosis and hepatocellular carcinoma (HCC). NAFLD is a complex disease whose pathogenesis is shaped by both environmental and genetic factors. In the last two decades, several heritable modifications in genes influencing hepatic lipid remodeling, and mitochondrial oxidative status have been emerged as predictors of progressive hepatic damage. Among them, the patatin-like phospholipase domain-containing 3 (PNPLA3) p.I148M, the Transmembrane 6 superfamily member 2 (TM6SF2) p.E167K and the rs641738 membrane bound-o-acyltransferase domain-containing 7 (MBOAT7) polymorphisms are considered the most robust modifiers of NAFLD. However, a forefront frontier in the study of NAFLD heritability is to postulate score-based strategy, building polygenic risk scores (PRS), which aggregate the most relevant genetic determinants of NAFLD and biochemical parameters, with the purpose to foresee patients with greater risk of severe NAFLD, guaranteeing the most highly predictive value, the best diagnostic accuracy and the more precise individualized therapy.
Collapse
Affiliation(s)
- Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (G.T.)
| | - Miriam Longo
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (G.T.)
- Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, 20122 Milano, Italy
| | - Giada Tria
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (G.T.)
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, Via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (G.T.)
| |
Collapse
|
50
|
Nicoll R, Gerasimidis K, Forrest E. The Role of Micronutrients in the Pathogenesis of Alcohol-Related Liver Disease. Alcohol Alcohol 2021; 57:275-282. [PMID: 34491307 DOI: 10.1093/alcalc/agab060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
AIMS Chronic alcohol consumption may result in liver injury and chronic liver disease, but other factors are likely to influence disease progression. Malnutrition, specifically micronutrient deficiency, is frequently associated with both alcohol use disorder and chronic liver disease. We hypothesize that micronutrient deficiencies may affect the progression of liver disease in this population. METHODS Systematic integrative review of the medical literature; electronic search of MEDLINE 1950-2021; studies investigating role of any micronutrient in the acceleration of alcohol-related liver injury in humans or animals. Studies which specifically related to alcoholic hepatitis were excluded. Outcomes were extracted and recorded in tabulated form and discussed narratively. RESULTS We identified 46 studies investigating the role of micronutrient deficiencies in the pathogenesis of alcohol-related liver disease. Specific micronutrients which were identified included folic acid or related B vitamins (n = 9 studies), Vitamin D (n = 9 studies), magnesium (n = 8 studies), zinc (n = 8 studies) and selenium (n = 12 including one systematic review). Observational evidence suggests a potential role of magnesium deficiency in accelerating alcohol-related liver injury with weak or negative evidence for other micronutrients. CONCLUSIONS Magnesium deficiency may increase the risk of alcohol-related liver injury and adverse liver outcomes. However, currently, there is insufficient evidence to support magnesium supplementation except for clinically relevant magnesium deficiency. Long-term prospective cohort studies assessing the impact of micronutrients on liver disease progression in patients with alcohol use disorder are lacking and may help determine whether there is a causal role for micronutrient deficiencies in alcohol-related liver injury.
Collapse
Affiliation(s)
- Ruairidh Nicoll
- Department of Gastroenterology, Glasgow Royal Infirmary, 84 Castle Street, Glasgow G4 0SF, UK
| | - Konstantinos Gerasimidis
- Department of Human Nutrition, School of Medicine, College of Medicine, Veterinary and Life Sciences, University of Glasgow, New Lister Building, Glasgow Royal Infirmary, Glasgow G31 2ER, UK
| | - Ewan Forrest
- Department of Gastroenterology, Glasgow Royal Infirmary, 84 Castle Street, Glasgow G4 0SF, UK
| |
Collapse
|