1
|
Gu P, Chen J, Xin J, Chen H, Zhang R, Chen D, Zhang Y, Shao S. Network pharmacology-based investigation of the pharmacological mechanisms of diosgenin in nonalcoholic steatohepatitis. Sci Rep 2025; 15:10351. [PMID: 40133701 PMCID: PMC11937522 DOI: 10.1038/s41598-025-95154-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/19/2025] [Indexed: 03/27/2025] Open
Abstract
The prevalence of nonalcoholic steatohepatitis (NASH) is rising annually, posing health and economic challenges, with limited treatments available. Diosgenin, a natural steroidal compound found in various plants, holds potential as a therapeutic candidate. Recent studies have confirmed diosgenin's anti-inflammatory and metabolism-modulating properties. However, its therapeutic effects on NASH and the underlying mechanisms are still unclear. This study aims to explore diosgenin's protective effects and pharmacological mechanisms against NASH using network pharmacology, molecular docking, and experimental validation. We gathered potential targets of diosgenin and NASH from various databases to generate protein-protein interaction (PPI) networks. GO and KEGG pathway enrichment analyses identified key targets and mechanisms. Molecular docking confirmed the binding capacity between diosgenin and core target proteins. Additionally, a NASH cell model was developed to validate the pharmacological effects of diosgenin. Our investigation identified nine key targets (ALB, AKT1, TP53, VEGFA, MAPK3, EGFR, STAT3, CASP3, IGF1) that interact with diosgenin. Molecular docking indicated potential bindings interactions, while enrichment analyses revealed that diosgenin may enhance fatty acid metabolism via the PI3K-Akt pathway. Cellular experiments confirmed that diosgenin activates this pathway, reduces SCD1 expression, and decreases triglyceride and IL-6 levels. Our study elucidates that diosgenin may ameliorate triglyceride deposition and inflammation through the PI3K-Akt pathway.
Collapse
Affiliation(s)
- Peiyuan Gu
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
| | - Juan Chen
- Department of Endocrinology, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jingxin Xin
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Huiqi Chen
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
| | - Ran Zhang
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China
| | - Dan Chen
- Department of Electrocardiographic, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yuhan Zhang
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China.
| | - Shanshan Shao
- Key Laboratory of Endocrine Glucose and Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Shandong Key Laboratory of Endocrine Metabolism and Aging, Jinan, Shandong, China.
| |
Collapse
|
2
|
Huang X, Yu R, Tan X, Guo M, Xia Y, Zou H, Liu X, Qin C. Comparison of NAFLD, MAFLD, and MASLD Prevalence and Clinical Characteristics in Asia Adults. J Clin Exp Hepatol 2025; 15:102420. [PMID: 39564428 PMCID: PMC11570951 DOI: 10.1016/j.jceh.2024.102420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/27/2024] [Indexed: 11/21/2024] Open
Abstract
Background/Aims The principal limitations of the term non-alcoholic fatty liver disease (NAFLD) are the reliance on exclusionary confounder terms and the use of potentially stigmatizing language. Within three years, NAFLD went through two name changes, from NAFLD to metabolic-dysfunction-associated fatty liver disease (MAFLD) to metabolic dysfunction-associated steatotic liver disease (MASLD). However, there is no Asian consensus statement on the renaming of MASLD, and evidence on the epidemiology and characteristics in the Asia population under different diagnostic criteria remain limited. This study aimed to fill these gaps by analyzing the prevalence and characteristics of MASLD, NAFLD, and MAFLD in an Asian population. Methods A retrospective, cross-sectional study was conducted in regional China with participants from the health management database in 2017-2022. Demographic and laboratory metabolic profile and body composition data were obtained. Hepatic steatosis were diagnosed by ultrasound. The likelihood of having fibrosis was assessed using the NAFLD fibrosis score (NFS). Recently proposed criteria for metabolic dysfunction-associated steatotic liver disease (MASLD) were applied. Results A total of 20,226 subjects were included for final analysis. 7465 (36.91%) participants were categorized as MASLD patients, 10,726 (53.03%) participants were MAFLD, and 7333 (36.26%) participants were NAFLD. Compared with MAFLD, body composition of MASLD and NAFLD patients were obviously different. MASLD patients were older, had a higher body mass index and percentage of male gender, and had a higher ALT, diastolic blood pressure, triglyceride, and waist circumference but lower High-Density Lipoprotein Cholesterol (HDL-C) than non-MASLD patients. Using binary regression analysis, we found for the first time that putative bone mass (OR = 4.62, 95CI% 3.12-6.83) is associated with the risk of developing MASLD. The area under the receiver operating curve (AUC) for predicting cardiovascular outcomes (CV) was 0.644 for MAFLD and 0.701 for MASLD. Conclusion MASLD (36.91%) prevalence was closed to NAFLD (36.26%) and lower than MAFLD (53.03%). Presumed bone mass might be the predictor of disease progression in MASLD patients. MASLD better identifies patients likely to have a higher risk of metabolic disorders or CV events.
Collapse
Affiliation(s)
- Xinjuan Huang
- Xiangya School of Nursing, Central South University, Changsha, Hunan Province, China
- Health Management Medicine Centre, The Third Xiangya Hospital, Central South University, China
| | - Ruoling Yu
- Xiangya School of Nursing, Central South University, Changsha, Hunan Province, China
| | - Xinyun Tan
- Xiangya School of Nursing, Central South University, Changsha, Hunan Province, China
| | - Manjie Guo
- Xiangya School of Nursing, Central South University, Changsha, Hunan Province, China
| | - Yuanqin Xia
- School of Medicine, Jishou University, Jishou, Hunan Province, China
| | - Huihui Zou
- School of Medicine, Jishou University, Jishou, Hunan Province, China
| | - Xuelian Liu
- Health Management Medicine Centre, The Third Xiangya Hospital, Central South University, China
| | - Chunxiang Qin
- Health Management Medicine Centre, The Third Xiangya Hospital, Central South University, China
- Department of Nursing, The Third Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| |
Collapse
|
3
|
Saad M, Ibrahim W, Hasanin AH, Elyamany AM, Matboli M. Evaluating the therapeutic potential of genetically engineered probiotic Zbiotics (ZB183) for non-alcoholic steatohepatitis (NASH) management via modulation of the cGAS-STING pathway. RSC Med Chem 2024:d4md00477a. [PMID: 39290381 PMCID: PMC11403872 DOI: 10.1039/d4md00477a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
NAFLD/NASH has emerged as a global health concern with no FDA-approved treatment, necessitating the exploration of novel therapeutic elements for NASH. Probiotics are known as an important adjunct therapy in NASH. Zbiotics (ZB183) is the first commercially available genetically engineered probiotic. Herein, we aimed to evaluate the potential therapeutic effects of Zbiotics administration on NASH management by modulating the cGAS-STING-signaling pathway-related RNA network. In silico data analysis was performed and three DEGs (MAPK3/EDN1/TNF) were selected with their epigenetic modulators (miR-6888-5p miRNA, and lncRNA RABGAP1L-DT-206). The experimental design included NASH induction with an HSHF diet in Wistar rats and Zbiotics administration in NASH rats in comparison to statin treatment. Liver functions and lipid profile were assessed. Additionally, the expression levels of the constructed molecular network were assessed using RT-PCR. Moreover, the Zbiotics effects in NASH were further validated with histopathological examination of liver and colon samples. Also, immunohistochemistry staining of hepatic TNF-α and colonic occludin was assessed. Oral administration of Zbiotics for four weeks downregulated the expression of the cGAS-STING-related network (MAPK3/EDN1/TNF/miR-6888-5p miRNA/lncRNA RABGAP1L-DT-206) in NASH models. Zbiotics also ameliorated hepatic inflammation and steatosis, as evidenced by a notable improvement in NAS score and decreased hepatic TNF-α levels. Furthermore, Zbiotics exhibited favorable effects on colon health, including increased crypt length, reduced inflammatory cell infiltration, and restoration of colonic mucosa occludin expression. In conclusion, our findings suggest that Zbiotics has potential therapeutic effects on NASH via modulating the gut-liver axis and the cGAS-STING signaling pathway.
Collapse
Affiliation(s)
- Maha Saad
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Modern University for Technology and Information Cairo Egypt
- Biomedical Research Department, Faculty of Medicine, Modern University for technology and information Cairo Egypt
- Medical Biochemistry and Molecular Biology, Faculty of Medicine Cairo University Cairo Egypt
| | - Walaa Ibrahim
- Medical Biochemistry and Molecular Biology, Faculty of Medicine Cairo University Cairo Egypt
| | - Amany Helmy Hasanin
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University Cairo 11566 Egypt
| | - Aya Magdy Elyamany
- Anatomic Pathology Department, Faculty of Medicine, Cairo University Cairo Egypt
| | - Marwa Matboli
- Departement of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Ain Shams University Cairo 11566 Egypt
| |
Collapse
|
4
|
Yang Y, Wang X. Nano-drug delivery systems (NDDS) in metabolic dysfunction-associated steatotic liver disease (MASLD): current status, prospects and challenges. Front Pharmacol 2024; 15:1419384. [PMID: 39166109 PMCID: PMC11333238 DOI: 10.3389/fphar.2024.1419384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
About one-third of the global population suffers from metabolic dysfunction-associated steatotic liver disease (MASLD), but specific treatments for MASLD have long been lacking, primarily due to the unclear etiology of the disease. In addition to lifestyle modifications and weight loss surgery, pharmacotherapy is the most common treatment among MASLD patients, and these drugs typically target the pathogenic factors of MASLD. However, bioavailability, efficacy, and side effects all limit the maximum therapeutic potential of the drugs. With the development of nanomedicine, recent years have seen attempts to combine MASLD pharmacotherapy with nanomaterials, such as liposomes, polymer nanoparticles, micelles, and cocrystals, which effectively improves the water solubility and targeting of the drugs, thereby enhancing therapeutic efficacy and reducing toxic side effects, offering new perspectives and futures for the treatment of MASLD.
Collapse
Affiliation(s)
| | - Xiaojing Wang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Wenzhou Medical University and Lishui Municipal Central Hospital, Lishui, China
| |
Collapse
|
5
|
Nixdorf L, Hartl L, Ströhl S, Felsenreich DM, Mairinger M, Jedamzik J, Richwien P, Mozayani B, Semmler G, Balcar L, Schwarz M, Jachs M, Dominik N, Bichler C, Trauner M, Mandorfer M, Reiberger T, Langer FB, Bauer DJM, Prager G. Rapid improvement of hepatic steatosis and liver stiffness after metabolic/bariatric surgery: a prospective study. Sci Rep 2024; 14:17558. [PMID: 39080285 PMCID: PMC11289378 DOI: 10.1038/s41598-024-67415-w] [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: 03/27/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) and related steatohepatitis (MASH) are common among obese patients and may improve after metabolic/bariatric surgery (MBS). 93 Patients undergoing MBS in 2021-2022 were prospectively enrolled. Liver stiffness measurement (LSM; via vibration-controlled transient elastography [VCTE], point [pSWE] and 2D [2DSWE] shear wave elastography) and non-invasive steatosis assessment (via controlled attenuation parameter [CAP]) were performed before (baseline [BL]) and three months (M3) after surgery. 93 patients (median age 40.9 years, 68.8% female, median BL-BMI: 46.0 kg/m2) were included. BL-liver biopsy showed MASLD in 82.8% and MASH in 34.4% of patients. At M3 the median relative total weight loss (%TWL) was 20.1% and the median BMI was 36.1 kg/m2. LSM assessed by VCTE and 2DSWE, as well as median CAP all decreased significantly from BL to M3 both in the overall cohort and among patients with MASH. There was a decrease from BL to M3 in median levels of ALT (34.0 U/L to 31 U/L; p = 0.025), gamma glutamyl transferase (BL: 30.0 to 21.0 U/L; p < 0.001) and MASLD fibrosis score (BL: - 0.97 to - 1.74; p < 0.001). Decreasing LSM and CAP, as well as liver injury markers suggest an improvement of MASLD/MASH as early as 3 months after MBS.
Collapse
Affiliation(s)
- Larissa Nixdorf
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Lukas Hartl
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Stefanie Ströhl
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniel Moritz Felsenreich
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Magdalena Mairinger
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Julia Jedamzik
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Paula Richwien
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Behrang Mozayani
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Georg Semmler
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lorenz Balcar
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Schwarz
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Mathias Jachs
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Nina Dominik
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christoph Bichler
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Mattias Mandorfer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria.
| | - Felix B Langer
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - David Josef Maria Bauer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Gerhard Prager
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Pramanik S, Pal P, Ray S. Non-alcoholic fatty liver disease in type 2 diabetes: Emerging evidence of benefit of peroxisome proliferator-activated receptors agonists and incretin-based therapies. World J Methodol 2024; 14:91319. [PMID: 38983664 PMCID: PMC11229880 DOI: 10.5662/wjm.v14.i2.91319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/20/2024] [Accepted: 02/27/2024] [Indexed: 06/13/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global epidemic, affecting more than half of the people living with type 2 diabetes (T2D). The relationship between NAFLD and T2D is bidirectional and the presence of one perpetuates the other, which significantly increases the hepatic as well as extrahepatic complications. Until recently, there was no approved pharmacological treatment for NAFLD/ nonalcoholic steatohepatitits (NASH). However, there is evidence that drugs used for diabetes may have beneficial effects on NAFLD. Insulin sensitizers acting through peroxisome proliferator-activated receptor (PPAR) modulation act on multiple levels of NAFLD pathogenesis. Pioglitazone (PPARγ agonist) and saroglitazar (PPARα/γ agonist) are particularly beneficial and recommended by several authoritative bodies for treating NAFLD in T2D, although data on biopsy-proven NASH are lacking with the latter. Initial data on elafibanor (PPAR α/δ agonist) and Lanifibranor (pan PPAR agonist) are promising. On the other hand, incretin therapies based on glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RA) and dual- and triple-hormone receptor co-agonists reported impressive weight loss and may have anti-inflammatory and antifibrotic properties. GLP-1 RAs have shown beneficial effects on NAFLD/NASH and more studies on potential direct effects on liver function by dual- and triple-agonists are required. Furthermore, the long-term safety of these therapies in NAFLD needs to be established. Collaborative efforts among healthcare providers such as primary care doctors, hepatologists, and endocrinologists are warranted for selecting patients for the best possible management of NAFLD in T2D.
Collapse
Affiliation(s)
- Subhodip Pramanik
- Department of Endocrinology, Neotia Getwel Multispecialty Hospital, Siliguri 734010, West Bengal, India
| | - Partha Pal
- Department of Medical Gastroenterology, Asian Institute of Gastroenterology, Hyderabad 500082, India
| | - Sayantan Ray
- Department of Endocrinology, All India Institute of Medical Sciences, Bhubaneswar, Bhubaneswar 751019, Odisha, India
| |
Collapse
|
7
|
Linero PL, Castilla-Guerra L. Management of Cardiovascular Risk in the Non-alcoholic Fatty Liver Disease Setting. Eur Cardiol 2024; 19:e02. [PMID: 38807854 PMCID: PMC11131151 DOI: 10.15420/ecr.2023.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/02/2023] [Indexed: 05/30/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an overlooked and undetected pathology, which affects more than 32% of adults worldwide. NAFLD is becoming more common in Western industrialised countries, particularly in patients with central obesity, type 2 diabetes, dyslipidaemia and metabolic syndrome. Although NAFLD has traditionally been interpreted as a liver disease with a high risk of liver-related complications, NAFLD is an underappreciated and independent risk factor for atherosclerotic cardiovascular disease, which is the principal cause of death in patients with NAFLD. Treatment options to counteract both the progression and development of cardiovascular disease and NAFLD include lifestyle interventions, such as weight loss, increased physical activity and dietary modification, and optimal medical therapy of comorbid conditions; nevertheless, further studies are needed to define optimal treatment strategies for the prevention of both hepatic and cardiovascular complications of NAFLD.
Collapse
Affiliation(s)
- Paula Luque Linero
- Vascular Risk Unit, Department of Internal Medicine, Hospital Virgen MacarenaSeville, Spain
| | - Luis Castilla-Guerra
- Vascular Risk Unit, Department of Internal Medicine, Hospital Virgen MacarenaSeville, Spain
- Department of Medicine, University of SevilleSeville, Spain
| |
Collapse
|
8
|
Zhang F, Lo EKK, Chen J, Wang K, Felicianna, Ismaiah MJ, Leung HKM, Zhao D, Lee JCY, El-Nezami H. Probiotic Mixture Ameliorates a Diet-Induced MASLD/MASH Murine Model through the Regulation of Hepatic Lipid Metabolism and the Gut Microbiome. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8536-8549. [PMID: 38575146 PMCID: PMC11037262 DOI: 10.1021/acs.jafc.3c08910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/06/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disease that has no effective treatment. Our proprietary probiotic mixture, Prohep, has been proven in a previous study to be helpful in reducing hepatocellular carcinoma (HCC) in vivo. However, its prospective benefits on the treatment of other liver diseases such as MASLD, which is one of the major risk factors in the development of HCC, are unclear. To investigate the potential of Prohep in modulating the development and progression of MASLD, we first explored the effect of Prohep supplementation via voluntary intake in a high-fat diet (HFD)-induced MASLD/metabolic dysfunction-associated steatohepatitis (MASH) murine model. Our results indicated that Prohep alleviated HFD-induced liver steatosis and reduced excessive hepatic lipid accumulation and improved the plasma lipid profile when compared with HFD-fed control mice through suppressing hepatic de novo lipogenesis and cholesterol biosynthesis gene expressions. In addition, Prohep was able to modulate the gut microbiome, modify the bile acid (BA) profile, and elevate fecal short-chain fatty acid (SCFA) levels. Next, in a prolonged HFD-feeding MASLD/MASH model, we observed the effectiveness of Prohep in preventing the transition from MASLD to MASH via amelioration in hepatic steatosis, inflammation, and fibrosis. Taken together, Prohep could ameliorate HFD-induced MASLD and control the MASLD-to-MASH progression in mice. Our findings provide distinctive insights into the development of novel microbial therapy for the management of MASLD and MASH.
Collapse
Affiliation(s)
- Fangfei Zhang
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Emily Kwun Kwan Lo
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Jiarui Chen
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong
Kong 000, S.A.R., China
- Department
of Medicine, The University of Hong Kong, Hong Kong 000, S.A.R., China
- Leibniz
Institute for Natural Product Research and Infection Biology, Hans
Knöll Institute-Microbiome Dynamics, Jena D-07745, Germany
| | - Ke Wang
- Department
of Food Science and Nutrition, The Hong
Kong Polytechnic University, Hong
Kong 000, S.A.R., China
- Research
Institute for Future Food, The Hong Kong
Polytechnic University, Hong Kong 000, S.A.R., China
| | - Felicianna
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Marsena Jasiel Ismaiah
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Hoi Kit Matthew Leung
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Danyue Zhao
- Department
of Food Science and Nutrition, The Hong
Kong Polytechnic University, Hong
Kong 000, S.A.R., China
- Research
Institute for Future Food, The Hong Kong
Polytechnic University, Hong Kong 000, S.A.R., China
| | - Jetty Chung-Yung Lee
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
| | - Hani El-Nezami
- School
of Biological Sciences, University of Hong
Kong, Pokfulam, Hong Kong 000, S.A.R., China
- Institute
of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio FI-70211, Finland
| |
Collapse
|
9
|
Ran S, Song L, Yang H, Yu J, Zhen Y, Liu Q. Piperine alleviates nonalcoholic steatohepatitis by inhibiting NF-κB-mediated hepatocyte pyroptosis. PLoS One 2024; 19:e0301133. [PMID: 38547097 PMCID: PMC10977780 DOI: 10.1371/journal.pone.0301133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 03/08/2024] [Indexed: 04/02/2024] Open
Abstract
PURPOSE Nonalcoholic steatohepatitis (NASH) is the progressive form of nonalcoholic fatty liver disease (NAFLD), which has a high risk of cirrhosis, liver failure, and hepatocellular carcinoma. Piperine (Pip) is an extract of plants with powerful anti-inflammatory effects, however, the function of Pip in NASH remains elusive. Here, we aim to explore the role of Pip in NASH and to find the possible mechanisms. METHODS Methionine and choline-deficient (MCD) diets were used to induce steatohepatitis, methionine- and choline-sufficient (MCS) diets were used as the control. After Pip treatment, H&E staining, Oil Red O staining, hepatic triglyceride (TG) content and F4/80 expression were performed to analysis liver steatosis and inflammation; Masson's staining, COL1A1 and α-SMA were detected liver fibrosis. Lipopolysaccharide (LPS) -treated AML12 cells were used to as the cell model to induce pyroptosis. Then, pyroptosis-related proteins, IL-1β and LDH release were detected in vivo and in vitro. Finally, NF-κB inhibitor, BAY11-7082, was used to further demonstrate the mechanism of Pip in NASH. RESULTS The study found that Pip alleviated liver steatosis, inflammation, hepatocyte injury, and fibrosis in mice fed with MCD diets. Moreover, the pyroptosis markers (NLRP3, ASC, caspase-1 p20, and GSDMD), IL-1β and LDH release were decreased by Pip treatment. NF-κB activation was suppressed by Pip treatment and pyroptosis-related proteins were down regulated by BAY11-7082. CONCLUSION Pip ameliorates NASH progression, and the therapeutical effect was associated with inhibition of hepatocyte pyroptosis induced by NF-κB.
Collapse
Affiliation(s)
- Suye Ran
- Department of Gastroenterology, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Lingyu Song
- Department of Gastroenterology, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Hong Yang
- Department of Gastroenterology, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Jiangnan Yu
- Department of Gastroenterology, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Yunhuan Zhen
- Department of Colorectal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qi Liu
- Department of Gastroenterology, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| |
Collapse
|
10
|
Silva-Sperb AS, Moraes HA, Barcelos STA, de Moura BC, Longo L, Michalczuk MT, Cerski CTS, Uribe-Cruz C, da Silveira TR, Álvares-da-Silva MR, Dall’Alba V. Probiotic supplementation for 24 weeks in patients with non-alcoholic steatohepatitis: the PROBILIVER randomized clinical trial. Front Nutr 2024; 11:1362694. [PMID: 38600992 PMCID: PMC11004384 DOI: 10.3389/fnut.2024.1362694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/08/2024] [Indexed: 04/12/2024] Open
Abstract
Background and aim Considering the increasing prevalence of non-alcoholic steatohepatitis (NASH) and treatment gaps, this study aimed to evaluate the effect of probiotic supplementation on liver function markers, nutritional status, and clinical parameters. Methods This double-blind, randomized clinical trial (ClinicalTrials.gov ID: NCT0346782) included adult outpatients with biopsy-proven NASH. The intervention consisted of 24 weeks of supplementation with the probiotic mix Lactobacillus acidophilus (1 × 109 CFU) + Lactobacillus rhamnosus (1 × 109 CFU) + Lactobacillus paracasei (1 × 109 CFU) + Bifidobacterium lactis (1 × 109 CFU), or placebo, twice a day. The following parameters were evaluated: demographic and clinical data, transient elastography (FibroScan), liver enzymes, NAFLD fibrosis score, fatty liver index, laboratory assessment, serum concentration of toll-like receptor-4 (sTLR-4) and cytokeratin 18 (CK-18), anthropometric data, dietary intake, and physical activity. Regarding data analysis, the comparison between the groups was based on the delta of the difference of each variable analyzed (value at the end of treatment minus the baseline value) using the t-test for independent samples or the Mann-Whitney U-test. Results Forty-four patients with NASH completed the trial (51.4 ± 11.6 years). At baseline, 87% of participants had a mild liver fibrosis degree on biopsy, normal values of liver enzymes, transient elastography values consistent with grade 1 fibrosis in both groups, increased waist circumference (WC), a BMI of 30.97 kg/m2, and 76% presented with metabolic syndrome (MetS). After the intervention, no differences were observed between the probiotic and placebo groups in terms of MetS, WC, BMI scores, or liver enzyme levels (p > 0.05 for all). The elastography values remained consistent with grade 1 fibrosis in both groups. Although CK-18 was reduced in both groups, a larger effect size was noted in the probiotic group (D = 1.336). sTLR-4 was also reduced in both groups, with no difference between groups (p = 0.885). Conclusion Intervention with probiotics in the early stages of NASH demonstrated no significant change in hepatic and clinical parameters. Clinical trial registration ClinicalTrials.gov, identifier NCT0346782.
Collapse
Affiliation(s)
- Amanda Souza Silva-Sperb
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Helena Abadie Moraes
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Bruna Concheski de Moura
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Larisse Longo
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Matheus Truccolo Michalczuk
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Division of Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carlos Thadeu Schmidt Cerski
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Unit of Surgical Pathology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Carolina Uribe-Cruz
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Themis Reverbel da Silveira
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Mário Reis Álvares-da-Silva
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Division of Gastroenterology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Valesca Dall’Alba
- Graduate Program in Gastroenterology and Hepatology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Experimental Laboratory of Hepatology and Gastroenterology, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
- Nutrition Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
11
|
Wang S, Xia D, Fan H, Liu Z, Chen R, Suo C, Zhang T. Low thyroid function is associated with metabolic dysfunction-associated steatotic liver disease. JGH Open 2024; 8:e13038. [PMID: 38405186 PMCID: PMC10885173 DOI: 10.1002/jgh3.13038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/06/2024] [Accepted: 01/28/2024] [Indexed: 02/27/2024]
Abstract
Background and Aim Metabolic dysfunction-associated steatotic liver disease (MASLD) is recently introduced to better highlight the pathogenic significance of cardiometabolic dysfunction, as compared with non-alcoholic fatty liver disease. This study aimed to investigate the association between low thyroid function and MASLD in the new context. Methods We recruited 2901 participants for our retrospective cohort study from 2016 to 2021. Participants were divided into strict-normal thyroid function and low thyroid function groups (low-normal thyroid function, subclinical hypothyroidism) based on initial thyroid stimulating hormone (TSH) levels, respectively. Cox regression models were used to estimate the hazard ratios (HRs) and 95% CI. Results During a median follow-up of 15.6 months, 165 (8.9%) strict-normal thyroid function subjects and 141 (13.4%) low thyroid function subjects developed MASLD; this result was statistically relevant (P < 0.05). Univariate regression analysis showed that low thyroid function and subclinical hypothyroidism were statistically significantly associated with MASLD (low thyroid function: HR1.53; 95% CI 1.22-1.92; subclinical hypothyroidism: HR1.95; 95% CI 1.47-2.60). Conclusions MASLD is associated with low thyroid function and the relationship between MASLD and low thyroid function is independent.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
| | - Ding Xia
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
- Lifecycle Health Management Center, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hong Fan
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
| | - Zhenqiu Liu
- Fudan University Taizhou Institute of Health SciencesTaizhouChina
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, School of Life SciencesFudan UniversityShanghaiChina
| | - Ruilin Chen
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
| | - Chen Suo
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
- Fudan University Taizhou Institute of Health SciencesTaizhouChina
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public HealthFudan UniversityShanghaiChina
| | - Tiejun Zhang
- Department of Epidemiology, School of Public HealthFudan UniversityShanghaiChina
- Fudan University Taizhou Institute of Health SciencesTaizhouChina
- Shanghai Institute of Infectious Disease and Biosecurity, School of Public HealthFudan UniversityShanghaiChina
| |
Collapse
|
12
|
Zanini B, Benini F, Marullo M, Simonetto A, Rossi A, Cavagnoli P, Bonalumi A, Marconi S, Pigozzi MG, Gilioli G, Valerio A, Donato F, Castellano M, Ricci C. Mediterranean-Oriented Dietary Intervention Is Effective to Reduce Liver Steatosis in Patients with Nonalcoholic Fatty Liver Disease: Results from an Italian Clinical Trial. Int J Clin Pract 2024; 2024:8861126. [PMID: 38303926 PMCID: PMC10834092 DOI: 10.1155/2024/8861126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 12/28/2023] [Accepted: 01/13/2024] [Indexed: 02/03/2024] Open
Abstract
Results One hundred and fifty five subjects aged 20-59 years underwent (i) liver ultrasound (US), (ii) clinical and anthropometric evaluations, (iii) blood tests, and (iv) assessment of dietary habits. According to US evaluation, 73 of them had severe, moderate, or mild liver steatosis (NAFLD patients) and 82 had no liver steatosis (healthy controls). Fifty-eight NAFLD patients and 73 controls completed the study. Among NAFLD patients, 26 (45%) downgraded steatosis severity, 12 of which achieved complete steatosis regression (21%). Three of the healthy controls developed NAFLD. The NAFLD patients improved their dietary habits and reduced BMI and waist circumference, during the study period, more than healthy controls. Liver steatosis remission/regression was independent of changes in BMI or liver enzymes and was more frequent among patients with mild steatosis at baseline. Conclusions Mediterranean dietary advices, without a personalised meal planning, were efficient in reducing/remitting NAFLD, especially among patients with mild disease, which argues in favour of early identification and lifestyle intervention. This trial is registered with NCT03300661.
Collapse
Affiliation(s)
- Barbara Zanini
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Federica Benini
- Department of Medicine, ASST Spedali Civili of Brescia, Piazzale Spedali Civili 1, Brescia, Italy
| | - Monica Marullo
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Anna Simonetto
- Department of Civil Engineering, Architecture, Land and Environment, and Mathematics, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Angelo Rossi
- Health Protection Agency, ATS Brescia, Lombardy, Italy
| | - Paola Cavagnoli
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Alessia Bonalumi
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Silvia Marconi
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Marie Graciella Pigozzi
- Department of Medicine, ASST Spedali Civili of Brescia, Piazzale Spedali Civili 1, Brescia, Italy
| | - Gianni Gilioli
- Department of Civil Engineering, Architecture, Land and Environment, and Mathematics, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Alessandra Valerio
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Francesco Donato
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Viale Europa 11, Brescia, Italy
| | - Maurizio Castellano
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, Brescia, Italy
- Department of Medicine, ASST Spedali Civili of Brescia, Piazzale Spedali Civili 1, Brescia, Italy
| | - Chiara Ricci
- Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, Brescia, Italy
- Department of Medicine, ASST Spedali Civili of Brescia, Piazzale Spedali Civili 1, Brescia, Italy
| |
Collapse
|
13
|
Jin S, Chen P, Yang J, Li D, Liu X, Zhang Y, Xia Q, Li Y, Chen G, Li Y, Tong Y, Yu W, Fan X, Lin H. Phocaeicola vulgatus alleviates diet-induced metabolic dysfunction-associated steatotic liver disease progression by downregulating histone acetylation level via 3-HPAA. Gut Microbes 2024; 16:2309683. [PMID: 38312099 PMCID: PMC10854360 DOI: 10.1080/19490976.2024.2309683] [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: 09/26/2023] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disorder with limited effective interventions available. A novel approach to address this issue is through gut microbiota-based therapy. In our study, we utilized multi-omics analysis to identify Phocaeicola vulgatus (P. vulgatus) as a potential probiotic for the treatment of MASLD. Our findings from murine models clearly illustrate that the supplementation of P. vulgatus mitigates the development of MASLD. This beneficial effect is partly attributed to the metabolite 3-Hydroxyphenylacetic acid (3-HPAA) produced by P. vulgatus, which reduces the acetylation levels of H3K27 and downregulates the transcription of Squalene Epoxidase (SQLE), a rate-limiting enzyme in steroid biosynthesis that promotes lipid accumulation in liver cells. This study underscores the significant role of P. vulgatus in the development of MASLD and the critical importance of its metabolite 3-HPAA in regulating lipid homeostasis. These findings offer a promising avenue for early intervention therapy in the context of MASLD.
Collapse
Affiliation(s)
- Shengxi Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Yang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Duguang Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolong Liu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiyin Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiming Xia
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiling Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guoqiao Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yixuan Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Tong
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weihua Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxiao Fan
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
14
|
Kim Y, Lee SR, Lee SW. First in human randomised trial of J2H-1702: A novel 11β-hydroxysteroid dehydrogenase type 1 inhibitor for non-alcoholic steatohepatitis treatment. Aliment Pharmacol Ther 2023; 58:1132-1142. [PMID: 37743843 DOI: 10.1111/apt.17726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/10/2023] [Accepted: 09/09/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), which is an enzyme that converts cortisone to cortisol, plays a role in the regulation of glucose metabolism and inflammation. J2H-1702 is a novel 11β-HSD1 inhibitor, and the inhibition of 11β-HSD1 has been shown to improve insulin sensitivity, reduce inflammation, and prevent the development of nonalcoholic steatohepatitis (NASH) in preclinical models. AIMS We aimed to assess the pharmacokinetics (PKs), pharmacodynamics (PDs), safety, and tolerability of J2H-1702 after a single-dose oral administration. METHODS A randomised, double-blinded, placebo-controlled, single-dose, dose-escalation study was conducted on 50 healthy volunteers. Blood and urine samples were collected to assess the PK and PD of J2H-1702. RESULTS The peak plasma concentration of J2H-1702 was observed at 2-2.9 h after a single-dose oral administration. J2H-1702 reduced 11β-HSD1 activity compared to the placebo at all dose levels. The drug reached its maximal inhibitory effect within 12-24 h post-dose administration, and the inhibitory effect was maintained till 1 day after administration of the study drug. The drug showed typical first-order elimination kinetics, with a mean elimination half-life of 9.8-14.7 h. Systemic exposure to J2H-1702 increased in a dose-dependent manner. J2H-1702 was well tolerated after a single oral administration of up to 300 mg. A total of 11 treatment-emergent adverse events (TEAEs) occurred in seven (14%) participants, all of which were mild and resolved spontaneously. The most common TEAE was diarrhoea (8%), followed by dizziness (4%). CONCLUSIONS The results of this study suggest that J2H-1702 could be developed as an effective therapeutic option for NASH.
Collapse
Affiliation(s)
- Yun Kim
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Republic of Korea
| | - Shi-Ra Lee
- Department of Clinical Pharmacology and Therapeutics, Hanyang University Seoul Hospital, Seoul, Republic of Korea
| | - Sang Won Lee
- Department of Clinical Pharmacology and Therapeutics, Hanyang University Seoul Hospital, Seoul, Republic of Korea
- Department of Pharmacology, Hanyang University, College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
15
|
Lim WH, Lin SY, Ng CH, Tan DJH, Xiao J, Yong JN, Tay PWL, Syn N, Chin YH, Chan KE, Khoo CM, Chew N, Foo RSY, Shabbir A, Tan EX, Huang DQ, Noureddin M, Sanyal AJ, Siddiqui MS, Muthiah MD. Foregut bypass vs. restrictive bariatric procedures for nonalcoholic fatty liver disease: a meta-analysis of 3,355 individuals. Hepatobiliary Surg Nutr 2023; 12:658-670. [PMID: 37886204 PMCID: PMC10598314 DOI: 10.21037/hbsn-21-520] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 04/12/2022] [Indexed: 10/28/2023]
Abstract
Background Bariatric surgery represents an important treatment option for severely obese patients with nonalcoholic fatty liver disease (NAFLD). However, there remains inadequate data regarding the effects of different bariatric procedures on various NAFLD parameters, especially for histological outcomes. Thus, this meta-analysis aimed to compare the effects of restrictive bariatric procedures and foregut bypass on the metabolic, biochemical, and histological parameters for patients with NAFLD. Methods Medline and Embase were searched for articles relating to bariatric procedures and NAFLD. Pairwise meta-analysis was conducted to compare efficacy of bariatric procedures pre- vs. post-procedure with subgroup analysis to further compare restrictive against foregut bypass procedures. Results Thirty-one articles involving 3,355 patients who underwent restrictive bariatric procedures (n=1,460) and foregut bypass (n=1,895) were included. Both foregut bypass (P<0.01) and restrictive procedures (P=0.03) significantly increased odds of fibrosis resolution. Compared to restrictive procedures, foregut bypass resulted in a borderline non-significant decrease in fibrosis score (P=0.06) and significantly lower steatosis score (P<0.001). For metabolic parameters, foregut bypass significantly lowered body mass index (P=0.003) and low-density lipoprotein (P=0.008) compared to restrictive procedures. No significant differences were observed between both procedures for aspartate aminotransferase (P=0.17) and alkaline phosphatase (P=0.61). However, foregut bypass resulted in significantly lower gamma-glutamyl transferase than restrictive procedures (P=0.01) while restrictive procedures resulted in significantly lower alanine transaminase than foregut bypass (P=0.02). Conclusions The significant histological and metabolic advantages and comparable improvements in biochemical outcomes support the choice of foregut bypass over restrictive bariatric procedures in NAFLD management.
Collapse
Affiliation(s)
- Wen Hui Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Snow Yunni Lin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jieling Xiao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jie Ning Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Phoebe Wen Lin Tay
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nicholas Syn
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yip Han Chin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai En Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chin Meng Khoo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Endocrinology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Nicholas Chew
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore
| | - Roger S. Y. Foo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore
| | - Asim Shabbir
- Division of General Surgery (Upper Gastrointestinal Surgery), Department of Surgery, National University Hospital, Singapore, Singapore
| | - Eunice X. Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
- National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| | - Daniel Q. Huang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
- National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| | - Mazen Noureddin
- Cedars-Sinai Fatty Liver Program, Division of Digestive and Liver Diseases, Department of Medicine, Comprehensive Transplant Center, Cedars-Sinai Medical Centre, Los Angeles, CA, USA
| | - Arun J. Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Mohammad Shadab Siddiqui
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Mark D. Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
- National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| |
Collapse
|
16
|
Choudhuri G, Shah S, Kulkarni A, Jagtap N, Gaonkar P, Desai A, Adhav C. Non-alcoholic Steatohepatitis in Asians: Current Perspectives and Future Directions. Cureus 2023; 15:e42852. [PMID: 37664266 PMCID: PMC10473263 DOI: 10.7759/cureus.42852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2023] [Indexed: 09/05/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a subset of non-alcoholic fatty liver disease (NAFLD), which, apart from excess fat in the liver, may be characterised by some level of inflammatory infiltration and fibrogenesis, occasionally progressing to liver cirrhosis or hepatocellular carcinoma (HCC). The objective of the current review is to elucidate the rising prevalence, the role of microbiome and genetics in pathogenesis, diagnostic challenges, and novel treatment alternatives for NASH. Newer diagnostic techniques are being developed since using liver biopsy in a larger population is not a reasonable option and is primarily restricted to clinical research, at least in developing countries. Besides these technical challenges, another important factor leading to deviation from guideline practice is the lack of health insurance coverage in countries like India. It leads to reluctance on the part of physicians and patients to delay required tests to curb out-of-pocket expenditure. There is no cure for NASH, with liver transplantation remaining the last option for those who progress to end-stage liver disease (ESLD) or are detected with early-stage HCC. Thus, lifestyle modification remains the only viable option for many, but compliance and long-term adherence remain major challenges. In obese individuals, bariatric surgery and weight reduction have shown favourable results. In patients with less severe obesity, endoscopic bariatric metabolic therapies (EBMT) are rapidly emerging as less invasive therapies. However, access and acceptability remain poor for these weight reduction methods. Therefore, intense research is being conducted for potential newer drug classes with several agents currently in phase II or III of clinical development. Some of these have demonstrated promising results, such as a reduction in hepatic fat content, and attenuation of fibrosis with an acceptable tolerability profile in phase II studies. The developments in the management of NASH have been fairly encouraging. Further well-designed long-term prospective studies should be undertaken to generate evidence with definitive results.
Collapse
Affiliation(s)
| | - Saumin Shah
- Gastroenterology, Gujarat Gastro and Vascular Hospital, Surat, IND
| | - Anand Kulkarni
- Gastroenterology and Hepatology, Asian Institute of Gastroenterology, Hyderabad, IND
| | - Nitin Jagtap
- Gastroenterology, Asian Institute of Gastroenterology, Hyderabad, IND
| | | | | | | |
Collapse
|
17
|
Yenilmez B, Harney S, DiMarzio C, Kelly M, Min K, Echeverria D, Bramato BM, Jackson SO, Reddig K, Kim JK, Khvorova A, Czech MP. Dual targeting of hepatocyte DGAT2 and stellate cell FASN alleviates nonalcoholic steatohepatitis in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547848. [PMID: 37461560 PMCID: PMC10350091 DOI: 10.1101/2023.07.05.547848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a malady of multiple cell types associated with hepatocyte triglyceride (TG) accumulation, macrophage inflammation, and stellate cell-induced fibrosis, with no approved therapeutics yet available. Here, we report that stellate cell fatty acid synthase (FASN) in de novo lipogenesis drives the autophagic flux that is required for stellate cell activation and fibrotic collagen production. Further, we employ a dual targeting approach to NASH that selectively depletes collagen through selective stellate cell knockout of FASN (using AAV9-LRAT Cre in FASNfl/fl mice), while lowering hepatocyte triglyceride by depleting DGAT2 with a GalNac-conjugated, fully chemically modified siRNA. DGAT2 silencing in hepatocytes alone or in combination with stellate cell FASNKO reduced liver TG accumulation in a choline-deficient NASH mouse model, while FASNKO in hepatocytes alone (using AAV8-TBG Cre in FASNfl/fl mice) did not. Neither hepatocyte DGAT2 silencing alone nor FASNKO in stellate cells alone decreased fibrosis (total collagen), while loss of both DGAT2 plus FASN caused a highly significant attenuation of NASH. These data establish proof of concept that dual targeting of DGAT2 plus FASN alleviates NASH progression in mice far greater than targeting either gene product alone.
Collapse
Affiliation(s)
- Batuhan Yenilmez
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shauna Harney
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Kyounghee Min
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dimas Echeverria
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Brianna M. Bramato
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Samuel O. Jackson
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Keith Reddig
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Jason K. Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Anastasia Khvorova
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Michael P. Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
18
|
Zeng W, Xu X, Xu F, Zhu F, Li Y, Ma J. Exploring Key Genes with Diagnostic Value for Nonalcoholic Steatohepatitis Based on Bioinformatics Analysis. ACS OMEGA 2023; 8:20959-20967. [PMID: 37323410 PMCID: PMC10268261 DOI: 10.1021/acsomega.3c01709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/27/2023] [Indexed: 06/17/2023]
Abstract
We aimed to screen specific genes in liver tissue samples of patients with nonalcoholic steatohepatitis (NASH) with clinical diagnostic value based on bioinformatics analysis. The datasets of liver tissue samples from healthy individuals and NASH patients were retrieved for consistency cluster analysis to obtain the NASH sample typing, followed by verification of the diagnostic value of sample genotyping-specific genes. All samples were subjected to logistic regression analysis, followed by the establishment of the risk model, and then, the diagnostic value was determined by receiver operating characteristic curve analysis. NASH samples could be divided into cluster 1, cluster 2, and cluster 3, which could predict the nonalcoholic fatty liver disease activity score of patients. A total of 162 sample genotyping-specific genes were extracted from patient clinical parameters, and the top 20 core genes in the protein interaction network were obtained for logistic regression analysis. Five sample genotyping-specific genes (WD repeat and HMG-box DNA-binding protein 1 [WDHD1], GINS complex subunit 2 [GINS2], replication factor C subunit 3 (RFC3), secreted phosphoprotein 1 [SPP1], and spleen tyrosine kinase [SYK]) were extracted to construct the risk models with high diagnostic value in NASH. Compared with the low-risk group, the high-risk group of the model showed increased lipoproduction and decreased lipolysis and lipid β oxidation. The risk models based on WDHD1, GINS2, RFC3, SPP1, and SYK have high diagnostic value in NASH, and this risk model is closely related to lipid metabolism pathways.
Collapse
Affiliation(s)
- Wenchun Zeng
- Department
of Gastroenterology, The First People’s
Hospital of Yongkang, Affiliated to Hangzhou Medical College, Jinhua 321300, P. R. China
| | - Xiangwei Xu
- Department
of Pharmacy, The First People’s Hospital
of Yongkang, Affiliated to Hangzhou Medical College, Jinhua 321300, P. R. China
| | - Fang Xu
- Department
of Gastroenterology, The First People’s
Hospital of Yongkang, Affiliated to Hangzhou Medical College, Jinhua 321300, P. R. China
| | - Fang Zhu
- Department
of Gastroenterology, The First People’s
Hospital of Yongkang, Affiliated to Hangzhou Medical College, Jinhua 321300, P. R. China
| | - Yuecui Li
- Department
of Infectious Liver Disease, The First People’s
Hospital of Yongkang, Affiliated to Hangzhou Medical College, Jinhua 321300, P. R. China
| | - Ji Ma
- Department
of Gastroenterology, The First People’s
Hospital of Yongkang, Affiliated to Hangzhou Medical College, Jinhua 321300, P. R. China
| |
Collapse
|
19
|
Bariatric Surgery Is Associated with Alcohol-Related Liver Disease and Psychiatric Disorders Associated with AUD. Obes Surg 2023; 33:1494-1505. [PMID: 36881347 PMCID: PMC10156826 DOI: 10.1007/s11695-023-06490-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND/AIMS Bariatric surgery can increase the risk of addictive disorders and nutritional deficiencies. The aim of this study was to evaluate the association between bariatric surgery and alcohol use disorder (AUD), alcohol-related liver disease (ALD), and psychiatric disorders associated with AUD. The impact of vitamin D deficiency in these associations was also investigated. METHODS A cross-sectional study was performed using the National Inpatient Sample database and its ICD-9 codes information. Diagnostic and comorbidity data from hospital discharges were obtained from patients with bariatric surgery and other abdominal surgeries between 2005 and 2015. The two groups were then compared for alcohol-related outcomes after propensity-score matching. RESULTS The final study cohort included 537,757 patients with bariatric surgery and 537,757 with other abdominal surgeries. The bariatric surgery group had an increased risk of AUD [odds ratio (OR): 1.90; 95% CI: 1.85-1.95], ALD [OR: 1.29; 95% CI: 1.22-1.37], cirrhosis [OR, 1.39; 95% CI: 1.37-1.42], and psychiatric disorders associated with AUD [OR, 3.59; 95% CI: 3.37-3.84]. Vitamin D deficiency did not impact in the association between bariatric surgery and AUD, ALD, or psychiatric disorders associated with AUD. CONCLUSIONS Bariatric surgery is associated with an increased prevalence of AUD, ALD, and psychiatric disorders associated with AUD. These associations appear to be independent from vitamin D deficiency.
Collapse
|
20
|
Examining the interim proposal for name change to steatotic liver disease in the US population. Hepatology 2023; 77:1712-1721. [PMID: 36645228 DOI: 10.1097/hep.0000000000000043] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/13/2022] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND AIMS Fatty liver is the commonest liver condition globally and traditionally associated with NAFLD. A consensus meeting was held in Chicago to explore various terminologies. Herein, we explore the proposed changes in nomenclature in a population data set from the US. APPROACH AND RESULTS Statistical analysis was conducted using survey-weighted analysis. Assessment of fatty liver was conducted with vibration-controlled transient elastography. A controlled attenuation parameter of 288 dB/m was used to identify hepatic steatosis. Patients were classified into nonalcoholic steatotic liver disease, alcohol-associated steatotic liver disease, and viral hepatitis steatotic liver disease. Liver stiffness measures at ≥8.8, ≥11.7, and ≥14 kPa were used to identify clinically significant fibrosis, advanced fibrosis, and cirrhosis, respectively. A total of 5102 individuals were included in the analysis. Using a survey-weighted analysis, a total of 25.43%, 6.95%, and 0.73% of the population were classified as nonalcoholic steatotic liver disease, alcohol-associated steatotic liver disease, and viral hepatitis steatotic liver disease, respectively. A sensitivity analysis at controlled attenuation parameter of 248 dB/m and fatty liver index found similar distribution. In a comparison between nonalcoholic steatotic liver disease, alcohol-associated steatotic liver disease, and viral hepatitis steatotic liver disease, there was no significant difference between the odds of advanced fibrosis and cirrhosis between groups. However, viral hepatitis steatotic liver disease individuals were found to have a significantly higher odds of clinically significant fibrosis (OR: 3.76, 95% CI, 1.27-11.14, p =0.02) compared with nonalcoholic steatotic liver disease. CONCLUSIONS The current analysis assessed the proposed changes based on discussions from the consensus meeting. Although the definitions are an interim analysis of discussions, steatotic liver disease respects the underlying liver etiology and reduces stigma while increasing awareness of FL among viral and alcohol-associated steatosis/steatohepatitis.
Collapse
|
21
|
Fu CE, Ng CH, Yong JN, Chan KE, Xiao J, Nah B, Bong SHS, Win KM, Bwa AH, Lim WH, Tan DJH, Zeng RW, Chew N, Teng MLP, Siddiqui MS, Oben JA, Sanyal AJ, Wong VWS, Noureddin M, Muthiah M. A Meta-analysis on Associated Risk of Mortality in Nonalcoholic Fatty Liver Disease. Endocr Pract 2023; 29:33-39. [PMID: 36273685 DOI: 10.1016/j.eprac.2022.10.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Nonalcoholic fatty liver disease (NAFLD) affects much of the worldwide population and poses a significant burden to the global healthcare. The rising numbers of individuals with NAFLD and instances of mortality point toward the importance of understanding the association causes of mortality in NAFLD. This meta-analysis aimed to seek the associations of NAFLD with all-cause, cardiovascular disease (CVD)-related, liver-related, and cancer-related mortality. METHODS MEDLINE and Embase were searched for articles relating to causes of mortality between NAFLD and non-NAFLD. The DerSimonian and Laird random-effects model was used to analyze adjusted hazard ratios (HR), and a sensitivity analysis was conducted to reduce heterogeneity through a graphical display of study heterogeneity. RESULTS Fifteen studies involving 10 286 490 patients were included. Individuals with NAFLD exhibited an increased risk of all-cause mortality (HR, 1.32; 95% CI, 1.09-1.59; P < .01; I2 = 96.00%), CVD-related mortality (HR, 1.22; 95% CI, 1.06-1.41; P < .01; I2 = 81.00%), and cancer-related mortality (HR, 1.67; 95% CI, 1.15-2.41; P < .01; I2 = 95.00%). However, no significant association was found between liver-related mortality and NAFLD (HR, 3.58; 95% CI, 0.69-18.46; P =.13; I2 = 96.00%). The sensitivity analysis conducted with graphic display of heterogeneity and only population-based studies found similar results. CONCLUSION NAFLD was associated with an increased risk of all-cause, CVD-related, and cancer-related mortality but not liver-related mortality. The finding is likely because of low fibrosis prevalence in the community. However, the significant burden in other causes of mortality beyond the liver points to a need for multidisciplinary efforts to reduce the mortality risks.
Collapse
Affiliation(s)
- Clarissa Elysia Fu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Jie Ning Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kai En Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jieling Xiao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Benjamin Nah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Shirley Huey Shin Bong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
| | | | | | - Wen Hui Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Nicholas Chew
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Cardiology, National University Heart Centre, National University Hospital, Singapore
| | - Margaret L P Teng
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Mohammad Shadab Siddiqui
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Virginia
| | - Jude A Oben
- Institute for Liver and Digestive Health, University College London, London, United Kingdom; Department of Gastroenterology and Hepatology, Guy's and St Thomas' Hospital, NHS Foundation Trust, London, United Kingdom
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Virginia
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Mazen Noureddin
- Houston Research Institute, Houston Liver Institute, Houston, Texas
| | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore.
| |
Collapse
|
22
|
Chew NW, Muthiah MD, Sanyal AJ. Nonalcoholic fatty liver disease and nonalcoholic steatohepatitis: pathophysiology and implications for cardiovascular disease. CARDIOVASCULAR ENDOCRINOLOGY AND METABOLISM 2023:137-173. [DOI: 10.1016/b978-0-323-99991-5.00003-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
23
|
Xiao J, Ng CH, Chin YH, Tan DJH, Lim WH, Lim G, Quek J, Tang ASP, Chan KE, Soong RY, Chew N, Tay B, Huang DQ, Tamaki N, Foo R, Chan MY, Noureddin M, Siddiqui MS, Sanyal AJ, Muthiah MD. A Class Effect Network Meta-analysis of Lipid Modulation in Non-alcoholic Steatohepatitis for Dyslipidemia. J Clin Transl Hepatol 2022; 10:1042-1049. [PMID: 36381095 PMCID: PMC9634784 DOI: 10.14218/jcth.2022.00095] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/13/2022] [Accepted: 05/04/2022] [Indexed: 12/04/2022] Open
Abstract
Background and Aims Pharmaceutical therapy for NASH is associated with lipid modulation, but the consensus on drug treatment is limited and lacks comparative analysis of effectiveness. A network meta-analysis was conducted to compare NASH drug classes in lipid modulation. Methods Online databases were searched for randomized controlled trails (RCTs) evaluating NASH treatments in biopsy-proven NASH patients. Treatments were classified into four groups: (1) inflammation, (2) energy, (3) bile acids, and (4) fibrosis based on the mechanism of action. A Bayesian network analysis was conducted with outcome measured by mean difference (MD) with credible intervals (Crl) and surface under the cumulative ranking curve (SUCRA). Results Forty-four RCTs were included in the analysis. Bile acid modulating treatments (MD: 0.05, Crl: 0.03-0.07) were the best treatment for improvement in high-density lipid (HDL) cholesterol, followed by treatments modulating energy (MD: 0.03, Crl: 0.02-0.04) and fibrosis (MD: 0.01, Crl: -0.12 to 0.14) compared with placebo. The top three treatments for reduction in triglycerides were treatments modulating energy (MD: -0.46, Crl: -0.49 to -0.43), bile acids (MD: -0.22, Crl: -0.35 to -0.09), and inflammation (MD: -0.08, Crl: -0.13 to -0.03) compared with placebo. SUCRA found treatment modulating fibrosis (MD: -1.27, Crl: -1.76 to -0.79) was the best treatment for reduction in low-density lipid (LDL) cholesterol followed by treatment modulating inflammation (MD: -1.03, Crl: -1.09 to -0.97) and energy (MD: -0.37, Crl: -0.39 to -0.34) compared with placebo, but LDL cholesterol was worsened by treatments modulating bile acids. Conclusions Network analysis comparing the class effects of dyslipidemia modulation in NASH found that treatment targets can include optimization of atherogenic dyslipidemia. Future studies are required to evaluate the cardiovascular outcomes.
Collapse
Affiliation(s)
- Jieling Xiao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Cheng-Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yip-Han Chin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wen-Hui Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Grace Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jingxuan Quek
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ansel Shao Pin Tang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kai-En Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Rou-Yi Soong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nicholas Chew
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Center, National University Hospital, Singapore
| | - Benjamin Tay
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Daniel Q. Huang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- National University Center for Organ Transplantation, National University Health System, Singapore
| | - Nobuharu Tamaki
- NAFLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, La Jolla, CA, USA
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Roger Foo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Center, National University Hospital, Singapore
| | - Mark Y. Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Cardiology, National University Heart Center, National University Hospital, Singapore
| | - Mazen Noureddin
- Cedars-Sinai Fatty Liver Program, Division of Digestive and Liver Diseases, Department of Medicine, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mohammad Shadab Siddiqui
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Arun J. Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Mark D. Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
- National University Center for Organ Transplantation, National University Health System, Singapore
| |
Collapse
|
24
|
Ng CH, Xiao J, Chew NWS, Chin YH, Chan KE, Quek J, Lim WH, Tan DJH, Loke RWK, Tan C, Tang ASP, Goh XL, Nah B, Syn N, Young DY, Tamaki N, Huang DQ, Siddiqui MS, Noureddin M, Sanyal A, Muthiah M. Depression in non-alcoholic fatty liver disease is associated with an increased risk of complications and mortality. Front Med (Lausanne) 2022; 9:985803. [PMID: 36275825 PMCID: PMC9582593 DOI: 10.3389/fmed.2022.985803] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Background and aims The global prevalence of non-alcoholic fatty liver disease (NAFLD) is expected to rise continuously. Furthermore, emerging evidence has also shown the potential for concomitant depression in NAFLD. This study aims to examine the prevalence, risk factors, and adverse events of depression in NAFLD and evaluate whether treated depression can reverse the increased risks of adverse outcomes. Materials and methods This study analyses the 2000–2018 cycles of NHANES that examined liver steatosis with fatty liver index (FLI). The relationship between NAFLD and depression was assessed with a generalized linear mix model and a sensitivity analysis was conducted in the no depression, treated depression, and untreated depression groups. Survival analysis was conducted with cox regression and fine gray sub-distribution model. Results A total of 21,414 patients were included and 6,726 were diagnosed with NAFLD. The risk of depression in NAFLD was 12% higher compared to non-NAFLD individuals (RR: 1.12, CI: 1.00–1.26, p = 0.04). NAFLD individuals with depression were more likely to be older, females, Hispanics or Caucasians, diabetic, and have higher BMI. Individuals with depression have high risk for cardiovascular diseases (CVD) (RR: 1.40, CI: 1.25–1.58, p < 0.01), stroke (RR: 1.71, CI: 1.27–2.23, p < 0.01), all-cause mortality (HR: 1.50, CI: 1.25–1.81, p < 0.01), and cancer-related mortality (SHR: 1.43, CI: 1.14–1.80, p = 0.002) compared to NAFLD individuals without depression. The risk of CVD, stroke, all-cause mortality, and cancer-related mortality in NAFLD individuals with treated depression and depression with untreated treatment was higher compared to individuals without depression. Conclusion This study shows that concomitant depression in NAFLD patients can increase the risk of adverse outcomes. Early screening of depression in high-risk individuals should be encouraged to improve the wellbeing of NAFLD patients.
Collapse
Affiliation(s)
- Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore,*Correspondence: Cheng Han Ng, ; orcid.org/0000-0002-8297-1569
| | - Jieling Xiao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nicholas W. S. Chew
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore,Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore
| | - Yip Han Chin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai En Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jingxuan Quek
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wen Hui Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ryan Wai Keong Loke
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Caitlyn Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ansel Shao Pin Tang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xin Lei Goh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Benjamin Nah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Nicholas Syn
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dan Yock Young
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore,Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore,National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| | - Nobuharu Tamaki
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Daniel Q. Huang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore,Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore,National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| | - Mohammad Shadab Siddiqui
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | | | - Arun Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore,Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore,National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore,Mark Muthiah, ; orcid.org/0000-0002-9724-4743
| |
Collapse
|
25
|
Ng CH, Huang DQ, Nguyen MH. Nonalcoholic fatty liver disease versus metabolic-associated fatty liver disease: Prevalence, outcomes and implications of a change in name. Clin Mol Hepatol 2022; 28:790-801. [PMID: 35545437 PMCID: PMC9597238 DOI: 10.3350/cmh.2022.0070] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 05/10/2022] [Indexed: 01/05/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects about a third of the world's adult population and is a major public health concern. NAFLD is defined by the presence of hepatic steatosis and the absence of other causes of liver disease. As NAFLD is closely associated with the presence of the metabolic syndrome, several experts have called for a change in nomenclature from NAFLD to metabolic-associated fatty liver disease (MAFLD) to better reflect the underlying pathophysiology of NAFLD as a metabolically driven disease and shift to a "positive" diagnostic criteria rather than one of exclusion. Recent studies have suggested that the global prevalence of MAFLD is higher than that of NAFLD, and patients with MAFLD have more metabolic comorbidities compared to those with NAFLD. Emerging data also suggest that all-cause and cardiovascular mortality may be higher in MAFLD compared with NAFLD. In this synopsis, we discuss differences in clinical features, prevalence and clinical outcomes between NAFLD and MAFLD. In addition, we highlight the advantages and disadvantages of a name change from NAFLD to MAFLD from the perspective of the scientific community, care providers and patients.
Collapse
Affiliation(s)
- Cheng Han Ng
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Daniel Q. Huang
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Division of Gastroenterology and Hepatology, National University Health System, Singapore
| | - Mindie H. Nguyen
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, Palo Alto, CA, USA,Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA,Corresponding author : Mindie H. Nguyen Division of Gastroenterology and Hepatology, Stanford University Medical Center, 780 Welch Road, Palo Alto, CA 94304, USA Tel: +1-650-498-6081, Fax: +1-650-721-8710, E-mail:
| |
Collapse
|
26
|
Yenilmez B, Kelly M, Zhang GF, Wetoska N, Ilkayeva OR, Min K, Rowland L, DiMarzio C, He W, Raymond N, Lifshitz L, Pan M, Han X, Xie J, Friedline RH, Kim JK, Gao G, Herman MA, Newgard CB, Czech MP. Paradoxical activation of transcription factor SREBP1c and de novo lipogenesis by hepatocyte-selective ATP-citrate lyase depletion in obese mice. J Biol Chem 2022; 298:102401. [PMID: 35988648 PMCID: PMC9490592 DOI: 10.1016/j.jbc.2022.102401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 01/26/2023] Open
Abstract
Hepatic steatosis associated with high-fat diet, obesity, and type 2 diabetes is thought to be the major driver of severe liver inflammation, fibrosis, and cirrhosis. Cytosolic acetyl CoA (AcCoA), a central metabolite and substrate for de novo lipogenesis (DNL), is produced from citrate by ATP-citrate lyase (ACLY) and from acetate through AcCoA synthase short chain family member 2 (ACSS2). However, the relative contributions of these two enzymes to hepatic AcCoA pools and DNL rates in response to high-fat feeding are unknown. We report here that hepatocyte-selective depletion of either ACSS2 or ACLY caused similar 50% decreases in liver AcCoA levels in obese mice, showing that both pathways contribute to the generation of this DNL substrate. Unexpectedly however, the hepatocyte ACLY depletion in obese mice paradoxically increased total DNL flux measured by D2O incorporation into palmitate, whereas in contrast, ACSS2 depletion had no effect. The increase in liver DNL upon ACLY depletion was associated with increased expression of nuclear sterol regulatory element-binding protein 1c and of its target DNL enzymes. This upregulated DNL enzyme expression explains the increased rate of palmitate synthesis in ACLY-depleted livers. Furthermore, this increased flux through DNL may also contribute to the observed depletion of AcCoA levels because of its increased conversion to malonyl CoA and palmitate. Together, these data indicate that in fat diet-fed obese mice, hepatic DNL is not limited by its immediate substrates AcCoA or malonyl CoA but rather by activities of DNL enzymes.
Collapse
Affiliation(s)
- Batuhan Yenilmez
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Guo-Fang Zhang
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Nicole Wetoska
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Olga R Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Kyounghee Min
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Leslie Rowland
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Wentao He
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Naideline Raymond
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Lawrence Lifshitz
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Meixia Pan
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Xianlin Han
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jun Xie
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Randall H Friedline
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Jason K Kim
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Guangping Gao
- Viral Vector Core, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Mark A Herman
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA
| | - Christopher B Newgard
- Sarah W. Stedman Nutrition and Metabolism Center and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA; Department of Pharmacology and Cancer Biology, and Department of Medicine, Endocrinology and Metabolism Division, Duke University Medical Center, Durham, North Carolina, USA.
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA.
| |
Collapse
|
27
|
Ng CH, Lim WH, Chin YH, Yong JN, Zeng RW, Chan KE, Tan DJH, Fu CE, Tang ASP, Goh LH, Devi K, Chew NWS, Mak LYL, Tamaki N, Huang DQ, Noureddin M, Siddiqui MS, Loomba R, Sanyal AJ, Muthiah M. Living in the non-alcoholic fatty liver disease silent epidemic: a qualitative systematic review of patients' perspectives. Aliment Pharmacol Ther 2022; 56:570-579. [PMID: 35791632 DOI: 10.1111/apt.17121] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/04/2022] [Accepted: 06/22/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) affects one-fourth of the global population. Yet, the care of these patients is limited and awareness of NAFLD remains low in the general public. Investigations into the lives of these patients are often forgotten and traditional quantitative studies only paint part of the picture. AIM To assess the first-hand accounts of these individuals and their perspective on living with NAFLD. METHODS A systematic search was conducted on Medline, Embase, CINAHL, PsycINFO and Web of Science database for qualitative literature regarding patients' perspectives on NAFLD. An inductive thematic analysis was conducted to generate themes and supportive subthemes. RESULTS We incuded eight articles in the review. There were three major themes including the impact on the quality of life, knowledge and information, and attitudes and perceptions on care. The impact of the quality of life details the emotional and physical distress of NAFLD. Knowledge and information include the lack of sufficient communication between healthcare providers and patients with a distinct knowledge gap. Attitudes and perceptions on care extrapolate the current active participation of patients and needs of the patients and the future care that they desire. CONCLUSION This review synthesises first-hand accounts of individuals with NAFLD. With the growing burden of NAFLD, future public interventions must consider individual views for success to be found. The identified themes serve as a forefront for consideration for public policies. Ultimately, NAFLD is a multisystem disease, which must be managed by a multidisciplinary team.
Collapse
Affiliation(s)
- Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wen Hui Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yip Han Chin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jie Ning Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Kai En Chan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Clarissa Elysia Fu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ansel Shao Pin Tang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lay Hoon Goh
- Division of Family Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kamala Devi
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nicholas W S Chew
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Cardiology, National University Heart Centre, National University Hospital, Singapore
| | - Lung-Yi Loey Mak
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong
| | - Nobuharu Tamaki
- NAFLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, San Diego, California, USA.,Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Daniel Q Huang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore.,National University Centre for Organ Transplantation, National University Health System, Singapore
| | - Mazen Noureddin
- Cedars-Sinai Fatty Liver Program, Division of Digestive and Liver Diseases, Department of Medicine, Comprehensive Transplant Center, Cedars-Sinai Medical Centre, Los Angeles, California, USA
| | - Mohammad Shadab Siddiqui
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, San Diego, California, USA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore.,National University Centre for Organ Transplantation, National University Health System, Singapore
| |
Collapse
|
28
|
Yu XD, Wang JW. Ceramide de novo synthesis in non-alcoholic fatty liver disease: Pathogenic mechanisms and therapeutic perspectives. Biochem Pharmacol 2022; 202:115157. [PMID: 35777449 DOI: 10.1016/j.bcp.2022.115157] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, and its advanced form non-alcoholic steatohepatitis (NASH) may progress to cirrhosis and hepatocellular carcinoma. Ceramides have been shown to exacerbate NAFLD development through enhancing insulin resistance, reactive oxygen species production, liver steatosis, lipotoxicity and hepatocyte apoptosis, and eventually causing hepatic inflammation and fibrosis. Emerging evidence indicates that ceramide production in NAFLD is predominantly attributed to activation of the de novo synthesis pathway of ceramides in hepatocytes. More importantly, pharmacological modulation of ceramide de novo synthesis in preclinical studies seems efficacious for the treatment of NAFLD. In this review, we provide an overview of the pathogenic mechanisms of ceramides in NAFLD, discuss recent advances and challenges in pharmacological interventions targeting ceramide de novo synthesis, and propose some research directions in the field.
Collapse
Affiliation(s)
- Xiao-Dong Yu
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore (NUHCS), Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
29
|
Nakatsuka T, Tateishi R, Koike K. Changing clinical management of NAFLD in Asia. Liver Int 2022; 42:1955-1968. [PMID: 34459096 DOI: 10.1111/liv.15046] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/30/2021] [Accepted: 08/21/2021] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the leading cause of chronic liver disease, affecting approximately 25% of the world's population. Recently, because of the sedentary lifestyle and overnutrition resulting from urbanisation, the burden of NAFLD has rapidly increased in many Asian countries. Currently, the prevalence of NAFLD in Asia is approximately 30%, as is the case in many Western countries. In Asia, the prevalence and presentation of NAFLD vary widely across regions because of the substantial diversity in race, socioeconomic status and living environment. Furthermore, the dual aetiology of fatty liver, particularly with viral hepatitis in Asia, makes it complex and challenging to manage. Because Asians are likely to have central adiposity and insulin resistance, approximately 7%-20% of non-obese Asians with body mass indexes of less than 25 kg/m2 are estimated to have NAFLD. Accumulating evidence indicates that NAFLD is associated with various extrahepatic comorbidities such as cardiovascular disease, chronic kidney disease, malignancy, in addition to liver-specific complications. Therefore, NAFLD should be managed as a multisystem disease in conjunction with metabolic syndrome. Lifestyle modification remains the basis of NAFLD management, but few patients can achieve adequate weight loss and maintain it long term. While various pharmacological agents are in phase 3 trials for steatohepatitis, Asian patients are underrepresented in most trials. This article reviews the epidemiological trends, clinical features, optimal assessment and current management practices for NAFLD in Asia.
Collapse
Affiliation(s)
- Takuma Nakatsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Ryosuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Japan
| |
Collapse
|
30
|
Chew NWS, Ng CH, Truong E, Noureddin M, Kowdley KV. Nonalcoholic Steatohepatitis Drug Development Pipeline: An Update. Semin Liver Dis 2022; 42:379-400. [PMID: 35709720 DOI: 10.1055/a-1877-9656] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a burgeoning global health crisis that mirrors the obesity pandemic. This global health crisis has stimulated active research to develop novel NASH pharmacotherapies targeting dysregulated inflammatory, cellular stress, and fibrogenetic processes that include (1) metabolic pathways to improve insulin sensitivity, de novo lipogenesis, and mitochondrial utilization of fatty acids; (2) cellular injury or inflammatory targets that reduce inflammatory cell recruitment and signaling; (3) liver-gut axis targets that influence bile acid enterohepatic circulation and signaling; and (4) antifibrotic targets. In this review, we summarize several of the therapeutic agents that have been studied in phase 2 and 3 randomized trials. In addition to reviewing novel therapeutic drugs targeting nuclear receptor pathways, liver chemokine receptors, liver lipid metabolism, lipotoxicity or cell death, and glucagon-like peptide-1 receptors, we also discuss the rationale behind the use of combination therapy and the lessons learned from unsuccessful or negative clinical trials.
Collapse
Affiliation(s)
- Nicholas W S Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Emily Truong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Mazen Noureddin
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Fatty Liver Program, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kris V Kowdley
- Liver Institute Northwest and Elson S. Floyd College of Medicine, Washington State University, Seattle, Washington
| |
Collapse
|
31
|
Comprehensive Review and Updates on Holistic Approach Towards Non-Alcoholic Fatty Liver Disease Management with Cardiovascular Disease. Curr Atheroscler Rep 2022; 24:515-532. [PMID: 35507280 DOI: 10.1007/s11883-022-01027-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2022] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The global prevalence of non-alcoholic fatty liver disease (NAFLD) presents an unmet need in treating these, often asymptomatic, individuals. In this review, we summarised NAFLD management and described recent developments in non-alcoholic steatohepatitis (NASH) therapeutics that can shape the future of NAFLD. RECENT FINDINGS A multi-disciplinary effort in promoting sustainable lifestyle measures is paramount, with the goal of either limiting energy surplus alone or in combination with targeting downstream pathways of inflammation and fibrosis. Several antidiabetic medications like PPAR-γ agonist and glucagon-like peptide receptor agonists have beneficial effects on the metabolic profile as well as NASH histology. Vitamin E has shown promise in specific groups of patients with the haptoglobin2 allele protein. Newer drugs have demonstrated promising results in NASH resolution and fibrosis improvement such as obeticholic acid, resmetirom, aramchol, efruxifermin, aldafermin and lanifibranor. Apart from discussing the results of late stage clinical trials and the possible challenges in managing these patients with limited approved therapies, we also discussed the specific management of comorbidities (diabetes, hypertension, hyperlipidaemia, cardiovascular diseases) in NAFLD patients. Treatment strategy needs to target improvements in liver-related outcomes and cardiometabolic profile.
Collapse
|
32
|
Ng CH, Muthiah MD, Xiao J, Chin YH, Lim G, Lim WH, Tay P, Tan DJH, Yong JN, Pan XH, Koh JWH, Chew N, Syn N, Tan E, Huang DQ, Siddiqui MS, Loomba R, Sanyal AJ, Noureddin M. Meta-analysis: analysis of mechanistic pathways in the treatment of non-alcoholic steatohepatitis. Evidence from a Bayesian network meta-analysis. Aliment Pharmacol Ther 2022; 55:1076-1087. [PMID: 35285529 DOI: 10.1111/apt.16808] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/29/2021] [Accepted: 01/26/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Non-alcoholic steatohepatitis (NASH) is the most common cause of liver disease. However, there is lack of comparison of efficacy between different NASH drug classes. We conducted a network meta-analysis evaluating drug classes through comparing histological outcomes and targets of drugs. APPROACH AND RESULTS Medline, EMBASE and CENTRAL were searched for randomised controlled trials evaluating NASH drugs in biopsy-proven NASH patients. Primary outcomes included NASH resolution without worsening of fibrosis, at least 2-point reduction in Non-alcoholic fatty liver disease Activity Score (NAS) without worsening of fibrosis and at least 1-point reduction in fibrosis. Treatments were classified into inflammation, energy, bile acid and fibrosis modulators. The analysis was conducted with Bayesian network model and surface under the cumulative ranking curve (SUCRA) analysis. Among 49 included trials, treatments modulating energy (Risk ratio (RR): 1.92, Credible intervals (Crl): 1.59-2.34) were most likely to achieve NASH resolution followed by treatments modulating fibrosis (RR 1.66, Crl: 0.65-4.50), bile acids (RR: 1.37, Crl: 0.99-1.92) and inflammation (RR: 1.00, Crl: 0.75-1.33). Energy and bile acids modulation were effective in at least 2-point NAS reduction without worsening of fibrosis (RR: 1.52, Crl 1.30-1.77; RR: 1.69, Crl 1.41-2.03) and at least 1-point reduction in fibrosis (RR: 1.26, Crl:1.05-1.49; RR: 1.54, Crl: 1.20-1.97). CONCLUSIONS This network analysis demonstrates the relative superiority of drugs modulating energy pathways and bile acids in NASH treatment. This guides the development and selection of drugs for combination therapies.
Collapse
Affiliation(s)
- Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mark D Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore.,National University Centre for Organ Transplantation, National University Health System, Singapore
| | - Jieling Xiao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yip Han Chin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Grace Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Wen Hui Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Phoebe Tay
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jie Ning Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xin-Hui Pan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Nicholas Chew
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nicholas Syn
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Eunice Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore.,National University Centre for Organ Transplantation, National University Health System, Singapore
| | - Daniel Q Huang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore.,National University Centre for Organ Transplantation, National University Health System, Singapore
| | - Mohammad Shadab Siddiqui
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Hepatology, Department of Medicine, University of California at San Diego, San Diego, California, USA
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Mazen Noureddin
- Cedars-Sinai Fatty Liver Program, Division of Digestive and Liver Diseases, Department of Medicine, Comprehensive Transplant Center, Cedars-Sinai Medical Centre, Los Angeles, California, USA
| |
Collapse
|
33
|
Song Q, Liu H, Zhang Y, Qiao C, Ge S. Lipidomics Revealed Alteration of the Sphingolipid Metabolism in the Liver of Nonalcoholic Steatohepatitis Mice Treated with Scoparone. ACS OMEGA 2022; 7:14121-14127. [PMID: 35559132 PMCID: PMC9089391 DOI: 10.1021/acsomega.2c00693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/24/2022] [Indexed: 05/07/2023]
Abstract
Perturbation in sphingolipid metabolism has been regarded as a risk factor for nonalcoholic steatohepatitis (NASH) development, predisposing to inflammation, insulin resistance, and weight gain. Scoparone can regulate the level of ceramide in primary hepatocytes and effectively ameliorate hepatic inflammation, apoptosis, steatosis, and fibrogenesis in a mice model of NASH. Nevertheless, the potential effects of scoparone in sphingolipid metabolism, which is dysregulated in NASH, have not been explored so far. To uncover the impact of scoparone on sphingolipid metabolism in NASH and potential therapeutic targets for treating NASH, the liver tissue samples were collected and lipidomics analysis based on UPLC-QTRAP-MRM/MS was carried out. The collected raw data was handled with multivariate data treatment to discover the potential biomarkers in sphingolipid metabolism. Compared to the control group, 22 potential sphingolipid biomarkers were discovered in the NASH group, of which 10 were downregulated and 12 were upregulated. Orally administrated scoparone contributed to the reversal of the levels of these potential biomarkers. Ten differential metabolites showed a tendency of recovery compared to the control group and may be potential targets for scoparone to treat NASH. This study indicated that lipidomics can detect the perturbed sphingolipids to unravel the therapeutic effects of scoparone on NASH.
Collapse
Affiliation(s)
- Qi Song
- College
of Traditional Chinese Medicine, Hebei University, Baoding 071000, P.R. China
| | - Hu Liu
- College
of Traditional Chinese Medicine, Hebei University, Baoding 071000, P.R. China
| | - Yunqi Zhang
- College
of Traditional Chinese Medicine, Hebei University, Baoding 071000, P.R. China
| | - Chuanqi Qiao
- College
of Traditional Chinese Medicine, Hebei University, Baoding 071000, P.R. China
| | - Shaoqin Ge
- College
of Traditional Chinese Medicine, Hebei University, Baoding 071000, P.R. China
- College
of Basic Medical Science, Hebei University, Baoding 071000, P.R. China
- (S.G.). Phone: +86-312-5075644. Fax: +86-312-5075644
| |
Collapse
|
34
|
Yenilmez B, Wetoska N, Kelly M, Echeverria D, Min K, Lifshitz L, Alterman JF, Hassler MR, Hildebrand S, DiMarzio C, McHugh N, Vangjeli L, Sousa J, Pan M, Han X, Brehm MA, Khvorova A, Czech MP. An RNAi therapeutic targeting hepatic DGAT2 in a genetically obese mouse model of nonalcoholic steatohepatitis. Mol Ther 2022; 30:1329-1342. [PMID: 34774753 PMCID: PMC8899521 DOI: 10.1016/j.ymthe.2021.11.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/31/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a severe liver disorder characterized by triglyceride accumulation, severe inflammation, and fibrosis. With the recent increase in prevalence, NASH is now the leading cause of liver transplant, with no approved therapeutics available. Although the exact molecular mechanism of NASH progression is not well understood, a widely held hypothesis is that fat accumulation is the primary driver of the disease. Therefore, diacylglycerol O-acyltransferase 2 (DGAT2), a key enzyme in triglyceride synthesis, has been explored as a NASH target. RNAi-based therapeutics is revolutionizing the treatment of liver diseases, with recent chemical advances supporting long-term gene silencing with single subcutaneous administration. Here, we identified a hyper-functional, fully chemically stabilized GalNAc-conjugated small interfering RNA (siRNA) targeting DGAT2 (Dgat2-1473) that, upon injection, elicits up to 3 months of DGAT2 silencing (>80%-90%, p < 0.0001) in wild-type and NSG-PiZ "humanized" mice. Using an obesity-driven mouse model of NASH (ob/ob-GAN), Dgat2-1473 administration prevents and reverses triglyceride accumulation (>85%, p < 0.0001) without increased accumulation of diglycerides, resulting in significant improvement of the fatty liver phenotype. However, surprisingly, the reduction in liver fat did not translate into a similar impact on inflammation and fibrosis. Thus, while Dgat2-1473 is a practical, long-lasting silencing agent for potential therapeutic attenuation of liver steatosis, combinatorial targeting of a second pathway may be necessary for therapeutic efficacy against NASH.
Collapse
Affiliation(s)
- Batuhan Yenilmez
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Nicole Wetoska
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Dimas Echeverria
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Kyounghee Min
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Lawrence Lifshitz
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Julia F Alterman
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Matthew R Hassler
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Samuel Hildebrand
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Nicholas McHugh
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Lorenc Vangjeli
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Jacquelyn Sousa
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Meixia Pan
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xianlin Han
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA
| | - Anastasia Khvorova
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA; RNA Therapeutics Institute, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA.
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street Biotech Two, Suite 100, Worcester, MA 01605, USA.
| |
Collapse
|
35
|
Borodavkin P, Sheridan W, Coelho C, Oštarijaš E, Zaïr ZM, Miras AD, McGowan B, le Roux CW, Vincent RP, Dimitriadis GK. Effects of glucagon-like peptide-1 receptor agonists on histopathological and secondary biomarkers of non-alcoholic steatohepatitis: A systematic review and meta-analysis. Diabetes Obes Metab 2022; 24:337-342. [PMID: 34605124 DOI: 10.1111/dom.14565] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/19/2021] [Accepted: 09/28/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Petr Borodavkin
- Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London, UK
| | - William Sheridan
- Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London, UK
| | - Claudia Coelho
- Department of Diabetes and Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Eduard Oštarijaš
- Institute for Translational Medicine, University of Pecs Medical School, Pecs, Hungary
| | - Zoulikha M Zaïr
- Princess Royal University Hospital, Farnborough Common, London, UK
| | - Alexander D Miras
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Barbara McGowan
- Department of Diabetes and Endocrinology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Carel W le Roux
- Diabetes Complication Research Centre, School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Ireland
| | - Royce P Vincent
- Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London, UK
- Department of Clinical Biochemistry, King's College Hospital NHS Foundation Trust, London, UK
| | - Georgios K Dimitriadis
- Department of Endocrinology ASO/EASO COM, King's College Hospital NHS Foundation Trust, London, UK
- Obesity, Type 2 Diabetes and Immunometabolism Research Group, Department of Diabetes, Faculty of Life Sciences, School of Life Course Sciences, King's College London, London, UK
- Division of Reproductive Health, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
36
|
Muthiah MD, Cheng Han N, Sanyal AJ. A clinical overview of non-alcoholic fatty liver disease: A guide to diagnosis, the clinical features, and complications-What the non-specialist needs to know. Diabetes Obes Metab 2022; 24 Suppl 2:3-14. [PMID: 34387409 DOI: 10.1111/dom.14521] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 11/29/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has a rapidly rising prevalence worldwide and is the most common cause of liver disease in developed countries. In this article, we discuss the spectrum of disease of NAFLD with a focus on the earlier spectrum of the disease that is commonly encountered by non-specialists, as well as the hepatic and extra-hepatic associations of the disease. We discuss in detail the two common presentations of NAFLD, incidentally detected hepatic steatosis and asymptomatic raised liver enzymes, and provide an algorithm for management and continued to follow up for these patients. Considerations for the management of cardiovascular comorbidities in these patients are also discussed. Finally, we cover the topic of screening for NAFLD in high-risk populations.
Collapse
Affiliation(s)
- Mark D Muthiah
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, National University Hospital, National University Health System, Singapore, Singapore
| | - Ng Cheng Han
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
37
|
Song L, Feng D, Tan J, Zhang H. Effects of TMEM206 on the malignant behavior of HepG2 human hepatocellular carcinoma cells. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221122724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common cancer of the digestive system. Recently, transmembrane proteins (TMEMs) have been extensively studied in different types of malignant tumors. However, the influence of TMEM206 in hepatocellular carcinoma is unclear. The UALCAN database was used to investigate the expression of TMEM206 mRNA in liver cancer tissues and used the Human Protein Atlas (THPA) to study the expression of TMEM206 in HCC tissues. The expression of TMEM206 was measured in normal liver HL-7702 cells and HepG2, SMMC-7721, and Bel-7402 liver cancer cells. Next, a lentivirus was used to knockdown TMEM206 in HepG2 cells. Furthermore, after verifying knockdown, we studied the effect of TMEM206 downregulation on the malignant behavior of HepG2 and on the PI3K/AKT pathway. TMEM206 was highly expressed in liver cancer cells ( p < 0.001). Downregulation of TMEM206 significantly inhibited the proliferation, migration and invasion of HepG2, significantly promoted the apoptosis of HepG2, and inhibited the expression of P-PI3K and P-AKT. TMEM206 can affect the malignant behavior of HCC. And the PI3K/AKT pathway was affected. This study provides new ideas for the treatment of HCC.
Collapse
Affiliation(s)
- Ling Song
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dou Feng
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiajie Tan
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hong Zhang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
38
|
Lazarus JV, Mark HE, Anstee QM, Arab JP, Batterham RL, Castera L, Cortez-Pinto H, Crespo J, Cusi K, Dirac MA, Francque S, George J, Hagström H, Huang TTK, Ismail MH, Kautz A, Sarin SK, Loomba R, Miller V, Newsome PN, Ninburg M, Ocama P, Ratziu V, Rinella M, Romero D, Romero-Gómez M, Schattenberg JM, Tsochatzis EA, Valenti L, Wong VWS, Yilmaz Y, Younossi ZM, Zelber-Sagi S. Advancing the global public health agenda for NAFLD: a consensus statement. Nat Rev Gastroenterol Hepatol 2022; 19:60-78. [PMID: 34707258 DOI: 10.1038/s41575-021-00523-4] [Citation(s) in RCA: 427] [Impact Index Per Article: 142.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2021] [Indexed: 12/11/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a potentially serious liver disease that affects approximately one-quarter of the global adult population, causing a substantial burden of ill health with wide-ranging social and economic implications. It is a multisystem disease and is considered the hepatic component of metabolic syndrome. Unlike other highly prevalent conditions, NAFLD has received little attention from the global public health community. Health system and public health responses to NAFLD have been weak and fragmented, and, despite its pervasiveness, NAFLD is largely unknown outside hepatology and gastroenterology. There is only a nascent global public health movement addressing NAFLD, and the disease is absent from nearly all national and international strategies and policies for non-communicable diseases, including obesity. In this global Delphi study, a multidisciplinary group of experts developed consensus statements and recommendations, which a larger group of collaborators reviewed over three rounds until consensus was achieved. The resulting consensus statements and recommendations address a broad range of topics - from epidemiology, awareness, care and treatment to public health policies and leadership - that have general relevance for policy-makers, health-care practitioners, civil society groups, research institutions and affected populations. These recommendations should provide a strong foundation for a comprehensive public health response to NAFLD.
Collapse
Affiliation(s)
- Jeffrey V Lazarus
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain.
- EASL International Liver Foundation, Geneva, Switzerland.
| | - Henry E Mark
- EASL International Liver Foundation, Geneva, Switzerland
| | - Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne, UK
| | - Juan Pablo Arab
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Rachel L Batterham
- Centre for Obesity Research, University College London and National Institute of Health Research, UCLH Biomedical Research Centre, London, UK
| | - Laurent Castera
- Department of Hepatology, Hôpital Beaujon, Université de Paris, Paris, France
| | - Helena Cortez-Pinto
- Clínica Universitária de Gastrenterologia, Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Javier Crespo
- Gastroenterology and Heptology Unit, Hospital Universitario Marqués de Valdecilla, Santander, IDIVAL, Santander, Spain
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Veterans Health Administration and University of Florida, Gainesville, FL, USA
| | - M Ashworth Dirac
- Department of Health Metrics Sciences, Department of Family Medicine, University of Washington, Seattle, WA, USA
| | - Sven Francque
- Department of Gastroenterology Hepatology, University Hospital Antwerp, Antwerp, Belgium
- Translational Sciences in Inflammation and Immunology TWI2N, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Hannes Hagström
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Terry T-K Huang
- Center for Systems and Community Design and NYU-CUNY Prevention Research Center, Graduate School of Public Health and Health Policy, City University of New York, New York, NY, USA
| | - Mona H Ismail
- Division of Gastroenterology, Department of Internal Medicine, King Fahd Hospital of the University, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | | | - Shiv Kumar Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Rohit Loomba
- Department of Medicine, NAFLD Research Center, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Veronica Miller
- University of California Berkeley, School of Public Health, Forum for Collaborative Research, Washington, DC, USA
| | - Philip N Newsome
- National Institute for Health Research Birmingham Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, UK
| | | | - Ponsiano Ocama
- Department of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Vlad Ratziu
- Assistance Publique-Hôpitaux de Paris, Hôpital Pitie-Salpetriere, University of Paris, Paris, France
| | - Mary Rinella
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Diana Romero
- Department of Community Health and Social Sciences, CUNY Graduate School of Public Health and Health Policy, New York, NY, USA
| | - Manuel Romero-Gómez
- UCM Digestive Diseases, CIBEREHD and IBIS, Virgen del Rocío University Hospital, University of Seville, Seville, Spain
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Centre Mainz, Mainz, Germany
| | - Emmanuel A Tsochatzis
- University College London Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
- Sheila Sherlock Liver Centre, Royal Free Hospital, London, UK
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Precision Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Yusuf Yilmaz
- Department of Gastroenterology, Marmara University School of Medicine, Istanbul, Turkey
- Liver Research Unit, Institute of Gastroenterology, Marmara University, Istanbul, Turkey
| | | | - Shira Zelber-Sagi
- University of Haifa, Faculty of Social Welfare and Health Sciences, School of Public Health, Mount Carmel, Haifa, Israel
- Department of Gastroenterology, Tel-Aviv Medical Centre, Tel-Aviv, Israel
| |
Collapse
|
39
|
Ng CH, Lin SY, Chin YH, Lee MH, Syn N, Goh XL, Koh JH, Quek J, Hao Tan DJ, Mok SF, Tan E, Dan YY, Chew N, Khoo CM, Siddiqui MS, Muthiah M. Antidiabetic Medications for Type 2 Diabetics with Nonalcoholic Fatty Liver Disease: Evidence From a Network Meta-Analysis of Randomized Controlled Trials. Endocr Pract 2021; 28:223-230. [PMID: 34606980 DOI: 10.1016/j.eprac.2021.09.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/17/2021] [Accepted: 09/25/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Type 2 diabetes mellitus and nonalcoholic fatty liver disease (NAFLD) are closely related, and antidiabetic medications have been shown to be potential therapeutics in NAFLD. Using a network meta-analysis, we sought to examine the effectiveness of antidiabetic agents for the treatment of NAFLD in patients with type 2 diabetes mellitus. METHODS Medline and Embase were searched for randomized controlled trials relating to the use of antidiabetic agents, including sodium-glucose transport protein 2 (SGLT2) inhibitors, glucagon-like peptide-1 receptor agonists, and peroxisome proliferator-activated receptor gamma (PPARγ) agonists, biguanides, sulfonylureas and insulin, on NAFLD in patients with diabetes. The p-score was used as a surrogate marker of effectiveness. RESULTS A total of 14 articles were included in the analysis. PPARγ agonists were ranked as the best treatment in steatosis reduction, resulting in the greatest reduction of steatosis. There was statistical significance between PPARγ agonists [mean difference (MD): -6.02%, confidence interval (CI): -10.37% to -1.67%] and SGLT2 inhibitors (MD: -2.60%, CI: -4.87% to -0.33%) compared with standard of care for steatosis reduction. Compared with PPARγ agonists, SGLT2 inhibitors resulted in a statistical significant reduction in fibrosis (MD: -0.06, CI: -0.10 to -0.02). Body mass index reduction was highest in SGLT2 inhibitors and glucagon-like peptide-1 receptor agonists. Additionally, SGLT2 inhibitors were ranked as the best treatment for increasing high-density lipoprotein and reducing low-density lipoprotein. CONCLUSION Glucagon-like peptide-1 receptor agonists and SGLT2 inhibitors were suitable alternatives for the treatment of NAFLD in those with type 2 diabetes mellitus with a reduction in body mass index, fibrosis, and steatosis. SGLT2 inhibitors also have the added benefit of lipid modulation.
Collapse
Affiliation(s)
- Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Snow Yunni Lin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yip Han Chin
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ming Hui Lee
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nicholas Syn
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Biostatistics & Modelling Domain, Saw Swee Hock School of Public Health, Singapore
| | - Xin Lei Goh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jin Hean Koh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jingxuan Quek
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shao Feng Mok
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Division of Endocrinology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Eunice Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Yock Young Dan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Nicholas Chew
- Division of Cardiology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Chin Meng Khoo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Division of Endocrinology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Mohammad Shadab Siddiqui
- Department of Internal Medicine, Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Virginia
| | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore.
| |
Collapse
|
40
|
Lazarus JV, Anstee QM, Hagström H, Cusi K, Cortez-Pinto H, Mark HE, Roden M, Tsochatzis EA, Wong VWS, Younossi ZM, Zelber-Sagi S, Romero-Gómez M, Schattenberg JM. Defining comprehensive models of care for NAFLD. Nat Rev Gastroenterol Hepatol 2021; 18:717-729. [PMID: 34172937 DOI: 10.1038/s41575-021-00477-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is now the leading cause of chronic liver disease globally. Despite the increased demand placed on health-care systems, little attention has been given to the design and implementation of efficient and effective models of care for patients with NAFLD. In many health-care settings, no formal pathways exist and, where pathways are in place, they are often not standardized according to good practices. We systematically searched the peer-reviewed literature with the aim of identifying published examples of comprehensive models of care that answered four key questions: what services are provided? Where are they provided? Who is offering them? How are they coordinated and integrated within health-care systems? We identified seven models of care and synthesized the findings into eight recommendations nested within the 'what, where, who and how' of care models. These recommendations, aimed at policy-makers and practitioners designing and implementing models of care, can help to address the increasing need for the provision of good practice care for patients with NAFLD.
Collapse
Affiliation(s)
- Jeffrey V Lazarus
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain. .,EASL International Liver Foundation, Geneva, Switzerland.
| | - Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.,The Liver Unit & NIHR Biomedical Research Centre, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | | | - Kenneth Cusi
- Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Veterans Health Administration and University of Florida, Gainesville, FL, United States
| | - Helena Cortez-Pinto
- Clínica Universitária de Gastrenterologia, Laboratório de Nutrição, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Henry E Mark
- EASL International Liver Foundation, Geneva, Switzerland
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital, Heinrich Heine University, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at Heinrich-Heine University, Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Emmanuel A Tsochatzis
- University College London Institute for Liver and Digestive Health, Royal Free Hospital, London, United Kingdom.,Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom
| | - Vincent Wai-Sun Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Zobair M Younossi
- Center for Liver Diseases, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, United States
| | - Shira Zelber-Sagi
- University of Haifa, Faculty of Social Welfare and Health Sciences, School of Public Health, Mount Carmel, Haifa, Israel.,Department of Gastroenterology, Tel-Aviv Medical Centre, Tel-Aviv, Israel
| | - Manuel Romero-Gómez
- UCM Digestive Diseases, CIBEREHD and IBIS, Virgen del Rocío University Hospital, University of Seville, Seville, Spain.
| | - Jörn M Schattenberg
- Metabolic Liver Research Program, I. Department of Medicine, University Medical Centre Mainz, Mainz, Germany.
| |
Collapse
|
41
|
Pereira ENGDS, Paula DP, Araujo BPD, Fonseca MDJMD, Diniz MDFHS, Daliry A, Griep RH. Advanced glycation end product: A potential biomarker for risk stratification of non-alcoholic fatty liver disease in ELSA-Brasil study. World J Gastroenterol 2021; 27:4913-4928. [PMID: 34447235 PMCID: PMC8371502 DOI: 10.3748/wjg.v27.i29.4913] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/18/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Liver diseases are associated with the excess formation of advanced glycation end products (AGEs), which induce tissue inflammation and oxidative damage. However, the trend of oxidative marker levels according to the steatosis grade in non-alcoholic fatty liver disease (NAFLD) is unclear.
AIM To compare serum AGE levels between participants with NAFLD accordingly to steatosis severity in the baseline ELSA-Brasil population.
METHODS In 305 individuals at baseline ELSA-Brasil, NAFLD-associated steatosis was classified by ultrasound hepatic attenuation. The participants were grouped according to the severity of steatosis: mild and moderate/severe pooled. The measurement of serum fluorescent AGE concentrations was based on spectrofluorimetric detection. Serum AGE content and clinical and laboratory characteristics of the participants were compared between groups. The correlation between serum AGE levels and the grade of steatosis was analyzed. Logistic regression analysis was used to investigate the relationship between serum AGE levels and steatosis severity. A P value < 0.05 was considered statistically significant.
RESULTS According to the steatosis severity spectrum in NAFLD, from mild to moderate/severe, individuals with the most severe steatosis grade had a higher incidence of metabolic syndrome (63% vs 34%, P ≤ 0.001), diabetes mellitus (37% vs 14%, P ≤ 0.001), and high cholesterol levels (51% vs 33%, P < 0.001). Moreover, individuals with increasing severity of steatosis presented increasing waist circumference, body mass index, systolic and diastolic blood pressure, fasting blood glucose, glycated hemoglobin, insulin, triglycerides, alanine aminotransferase, gamma-glutamyl transferase, C-reactive protein, and uric acid levels and lower high-density lipoprotein. Higher serum AGE content was present in the moderate/severe group of individuals than in the mild group (P = 0.008). In addition, the serum AGE levels were correlated with the steatosis grade in the overall sample (rho = 0.146, P = 0.010). Logistic regression analysis, after adjusting for confounding variables, showed that subjects with higher serum AGE content had a 4.6-fold increased chance of having moderate or severe steatosis when compared to low levels of serum AGEs. According to the results of the receiver operator characteristic curves analyses (areas under the curve, AUC = 0.83), AGEs could be a good marker of steatosis severity in patients with NAFLD and might be a potential biomarker in predicting NAFLD progression, strengthening the involvement of AGE in NAFLD pathogenesis.
CONCLUSION NAFLD-associated steatosis was associated with serum AGE levels; therefore, plasmatic fluorescent AGE quantification by spectroscopy could be a promising alternative method to monitor progression from mild to severe NAFLD accordingly to steatosis grade.
Collapse
Affiliation(s)
| | - Daniela Polessa Paula
- National School of Statistical Sciences, Brazilian Institute of Geography and Statistics, Rio de Janeiro 20231-050, Brazil
| | - Beatriz Peres de Araujo
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | | | | | - Anissa Daliry
- Laboratory of Cardiovascular Investigation, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Rosane Harter Griep
- Laboratory of Health and Environment Education, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| |
Collapse
|
42
|
Faris M, Jahrami H, Abdelrahim D, Bragazzi N, BaHammam A. The effects of Ramadan intermittent fasting on liver function in healthy adults: A systematic review, meta-analysis, and meta-regression. Diabetes Res Clin Pract 2021; 178:108951. [PMID: 34273453 DOI: 10.1016/j.diabres.2021.108951] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 07/03/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Growing evidence is suggestive that intermittent fasting likely to improve liver function; however, still the evidences are controversial to draw a definitive conclusion. Therefore, we conducted a systematic review and meta-analysis to estimate the effect size for changes in liver function tests (LFT) in healthy people practicing Ramadan diurnal intermittent fasting (RDIF), and to examine the impact of different covariates using subgroup analysis and meta-regression. METHODS Scientific databases were searched from date of inception in 1950 to the end of July 2020. The liver function tests searched and analyzed were aspartate transaminase (AST), alanine transaminase (ALT), gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), bilirubin (BLU), L-lactate dehydrogenase (LDH) and prothrombin time (PT). RESULTS Twenty studies (601 adult participants in total, aged 18-57 years) conducted in 10 countries between 1987 and 2020 were identified. RDIF-induced effect sizes for the LFT expressed as standardized mean difference (SMD) [95% confidence interval] were: AST (no. of studies K = 16, number of subjects N = 502, SMD = -0.257 [-0.381, -0.133], I2 = 42%); ALT (K = 16, N = 502, SMD = -0.105 [-0.282, 0.07], I2 = 71%); GGT (K = 2, N = 46, SMD = -0.533 [-0.842, -0.224], I2 = 0%); ALP (K = 10, N = 312, SMD = -0.318 [-0.432, -0.204], I2 = 0.0%); BLU (K = 10, N = 325, SMD = -0.264 [-0.520, -0.007], I2 = 70.1%); LDH (K = 5, N = 145, SMD = -0.041 [-0.380, 0.298], I2 = 72%); PT (K = 2, N = 74, SMD = -0.027 [-0.732, 0.678], I2 = 87%). CONCLUSION RDIF induces significant but small (AST, ALP, BLU) to medium (GGT) positive changes on LFT, and may confer a transient, short-term protection against fatty liver disease in healthy subjects.
Collapse
Affiliation(s)
- MoezAlIslam Faris
- Department of Clinical Nutrition and Dietetics, College of Health Sciences/Research Institute for Medical and Health Sciences (RIMHS), University of Sharjah, Sharjah, United Arab Emirates.
| | - Haitham Jahrami
- Rehabilitation Services, Periphery Hospitals, Ministry of Health, Manama, Bahrain; College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Dana Abdelrahim
- Department of Nutrition and Food Technology, Faculty of Agriculture, The University of Jordan, Amman, Jordan
| | - Nicola Bragazzi
- Department of Mathematics and Statistics, Laboratory of Industrial and Applied Mathematics (LIAM), York University, Toronto, Canada
| | - Ahmed BaHammam
- Department of Medicine, College of Medicine, University Sleep Disorders Center, King Saud University, Riyadh, Saudi Arabia; The Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, Riyadh, Saudi Arabia
| |
Collapse
|
43
|
Omagari K, Koba C, Nagata A, Ngo LCT, Yamasaki M, Fukuda A, Yuasa M, Suruga K, Inada N, Ichimura-Shimizu M, Tsuneyama K. Olive leaf powder prevents nonalcoholic steatohepatitis in Sprague–Dawley rats fed a high-fat and high-cholesterol diet. CLINICAL NUTRITION OPEN SCIENCE 2021. [DOI: 10.1016/j.nutos.2021.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
44
|
Diabetic Kidney Disease, Cardiovascular Disease and Non-Alcoholic Fatty Liver Disease: A New Triumvirate? J Clin Med 2021; 10:jcm10092040. [PMID: 34068699 PMCID: PMC8126096 DOI: 10.3390/jcm10092040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease is a highly prevalent disease worldwide with a renowned relation to cardiovascular disease and chronic kidney disease. These diseases share a common pathophysiology including insulin resistance, oxidative stress, chronic inflammation, dysbiosis and genetic susceptibilities. Non-alcoholic fatty liver disease is especially prevalent and more severe in type 2 diabetes. Patients with non-alcoholic fatty liver disease should have liver fibrosis assessment in order to identify those at the highest risk of adverse outcomes so that appropriate management strategies can be implemented. Early diagnosis and treatment of non-alcoholic fatty liver disease could ameliorate the burden of cardiovascular disease and chronic kidney disease.
Collapse
|
45
|
Stojsavljevic-Shapeski S, Duvnjak M, Virovic-Jukic L, Hrabar D, Smircic Duvnjak L. New Drugs on the Block-Emerging Treatments for Nonalcoholic Steatohepatitis. J Clin Transl Hepatol 2021; 9:51-59. [PMID: 33604255 PMCID: PMC7868699 DOI: 10.14218/jcth.2020.00057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 09/18/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
Patients with nonalcoholic steatohepatitis (NASH) are at higher risk of progression to advanced stages of fibrosis, cirrhosis, hepatocellular carcinoma and other end-stage liver disease complications. When addressing treatment of NASH, we have limited approved options, and the mainstay of therapy is lifestyle intervention. Extensive research and revelation in the field of pathogenesis of NASH has offered new possibilities of treatment and emerging new drugs that are being tested currently in numerous preclinical and clinical trials. These drugs target almost all steps in the pathogenesis of NASH to improve insulin sensitivity, glucose and lipid metabolism, to inhibit de novo lipogenesis and delivery of lipids to the liver, and to influence apoptosis, inflammation and fibrogenesis. Although NASH is a multifactorial disease, in the future we could identify the predominating pathological mechanism and, by choosing the most appropriate specific medication, tailor the treatment for every patient individually.
Collapse
Affiliation(s)
| | - Marko Duvnjak
- Polyclinic Duvnjak, Zagreb, Croatia
- University of Applied Health Science, Zagreb, Croatia
| | - Lucija Virovic-Jukic
- Department of Gastroenterology and Hepatology, Clinical Hospital Center Sestre Milosrdnice, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Davor Hrabar
- Department of Gastroenterology and Hepatology, Clinical Hospital Center Sestre Milosrdnice, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Lea Smircic Duvnjak
- University of Applied Health Science, Zagreb, Croatia
- Vuk Vrhovac University Clinic-UH Merkur, Zagreb, Croatia
| |
Collapse
|
46
|
Niederseer D, Wernly B, Aigner E, Stickel F, Datz C. NAFLD and Cardiovascular Diseases: Epidemiological, Mechanistic and Therapeutic Considerations. J Clin Med 2021; 10:467. [PMID: 33530440 PMCID: PMC7865665 DOI: 10.3390/jcm10030467] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Overwhelming evidence suggests an association of cardiovascular disease (CVD) with non-alcoholic fatty liver disease (NAFLD); however, the underlying mechanisms remain largely speculative. It is, however, likely that common mechanisms contribute to the development of CVD and NAFLD, with lifestyle factors such as smoking, sedentary lifestyle with poor nutrition habits and physical inactivity being major candidates. These behavioral factors, on a predisposing genetic background, trigger changes in gut microbiota, inflammation, dyslipidemia and oxidative stress, leading to metabolic syndrome, diabetes and obesity as well as atherosclerosis. Treatment options to counteract both the progression and development of CVD and NAFLD include lifestyle interventions, optimal medical therapy of comorbid conditions and, as final possibility, bariatric surgery. As no causal pharmacotherapy of NAFLD is available, further research is urgently needed to address the unmet need of a growing population with NAFLD and CVD.
Collapse
Affiliation(s)
- David Niederseer
- Department of Cardiology, University Heart Center Zurich, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Bernhard Wernly
- Department of Anaesthesiology, Perioperative Medicine and Intensive Care Medicine, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria;
- Center for Public Health and Healthcare Research, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
- Department of Cardiology, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Elmar Aigner
- First Department of Medicine, Paracelsus Medical University, 5020 Salzburg, Austria;
| | - Felix Stickel
- Department of Gastroenterology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Christian Datz
- Department of Internal Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, 5110 Oberndorf, Austria
| |
Collapse
|
47
|
Krolevets T, Livzan V. Non-alcoholic fatty liver disease: digest 2021. DOKAZATEL'NAYA GASTROENTEROLOGIYA 2021; 10:27. [DOI: 10.17116/dokgastro20211002127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
|
48
|
Raza S, Rajak S, Upadhyay A, Tewari A, Anthony Sinha R. Current treatment paradigms and emerging therapies for NAFLD/NASH. FRONT BIOSCI-LANDMRK 2021; 26:206-237. [PMID: 33049668 PMCID: PMC7116261 DOI: 10.2741/4892] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one the fastest emerging manifestations of the metabolic syndrome worldwide. Non-alcoholic steatohepatitis (NASH), the progressive form of NAFLD, may culminate into cirrhosis and hepatocellular cancer (HCC) and is presently a leading cause of liver transplant. Although a steady progress is seen in understanding of the disease epidemiology, pathogenesis and identifying therapeutic targets, the slowest advancement is seen in the therapeutic field. Currently, there is no FDA approved therapy for this disease and appropriate therapeutic targets are urgently warranted. In this review we discuss the role of lifestyle intervention, pharmacological agents, surgical approaches, and gut microbiome, with regard to therapy for NASH. In particular, we focus the role of insulin sensitizers, thyroid hormone mimetics, antioxidants, cholesterol lowering drugs, incretins and cytokines as therapeutic targets for NASH. We highlight these targets aiming to optimize the future for NASH therapy.
Collapse
Affiliation(s)
- Sana Raza
- Department of Endocrinology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Sangam Rajak
- Department of Endocrinology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Aditya Upadhyay
- Department of Endocrinology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Archana Tewari
- Department of Endocrinology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India,
| |
Collapse
|
49
|
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is a global epidemic without effective therapeutic agents in the clinic. This meta-analysis aimed to assess the efficacy of the marketed hepatoprotectant bicyclol at improving blood biomarkers in patients with NAFLD. DESIGN Electronic databases were searched for randomised controlled trials (RCTs) published up to August 2020 using bicyclol to treat NAFLD. The risk of bias, quality of evidence and publication bias were evaluated. Blood biomarkers, including alanine transaminase (ALT), aspartate aminotransferase (AST), total bilirubin (TBIL), triglyceride (TG) and total cholesterol (TC), were analysed using Review Manager V.5.3 software. Outcomes with significant heterogeneity (I2 ≥75%) were divided into the bicyclol monotherapy subgroup and combination treatment subgroup. RESULTS Twelve RCTs involving 1008 patients were finally included. No serious adverse events were reported in the bicyclol-treated groups. The total effective rate of bicyclol intervention for NAFLD was significantly higher than that of the control group. The decreases in the levels of AST (mean difference (MD) = -15.20; 95% CI -20.51 to -9.90; I2=74%), TBIL (MD = -1.72; 95% CI -2.72 to -0.72; I2=0%) and TC (MD = -0.52; 95% CI -0.70 to -0.34; I2=67%) treated by bicyclol were significantly higher than those in the control group. When a high heterogeneity existed (I2 ≥75%), subgroup analyses were conducted and revealed significantly decreased ALT levels (MD = -34.07; 95% CI -36.70 to -31.43; I2=0%) merely in the bicyclol monotherapy subgroup, while TG level (MD = -0.39; 95% CI -0.45 to -0.33; I2=0%) was decreased in the bicyclol combination therapy subgroup. CONCLUSIONS The study presents the evidence of bicyclol monotherapy and/or combination therapy for improving liver function and blood lipid biomarkers in patients with NAFLD. This preliminary study predicts that bicyclol might be an alternative drug for NAFLD therapy in the future.
Collapse
Affiliation(s)
- Hu Li
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan-Nan Liu
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zong-Gen Peng
- CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
50
|
Hepatocellular carcinoma in patients with non-alcoholic steatohepatitis - epidemiology, risk factors, clinical implications and treatment. Clin Exp Hepatol 2020; 6:170-175. [PMID: 33145423 PMCID: PMC7592090 DOI: 10.5114/ceh.2020.99506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022] Open
Abstract
In recent years, rapid growth of incidence of metabolic syndrome, obesity and diabetes has been noted worldwide. Concurrent non-alcoholic steatohepatitis (NASH) has become a dominant factor of hepatic cirrhosis and hepatocellular carcinoma (HCC). The most important risk factors of transition from NASH to HCC are the degree of liver fibrosis, diabetes, obesity, age and male gender. Body mass index (BMI) reduction and increase of physical activity limit the risk of occurrence of HCC. Also, treatment of diabetes with metformin and application of statins have potential anticancer effects. Patients with HCC due to NASH should be treated in line with BCLC staging. Distant results of HCC therapy in the course of non-alcoholic fatty liver disease (NAFLD) are similar to the results of cancer of different aetiologies. However, patients with the metabolic syndrome are at high perioperative risk, and thus require accurate preparation, especially cardiological, in order to avoid that risk.
Collapse
|