1
|
Talari NK, Mattam U, Rahman AP, Hemmelgarn BK, Wyder MA, Sylvestre PB, Greis KD, Chella Krishnan K. Functional compartmentalization of hepatic mitochondrial subpopulations during MASH progression. Commun Biol 2025; 8:258. [PMID: 39966593 DOI: 10.1038/s42003-025-07713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
The role of peridroplet mitochondria (PDM) in diseased liver, such as during the progression of metabolic dysfunction-associated steatohepatitis (MASH), remains unknown. We isolated hepatic cytoplasmic mitochondria (CM) and PDM from a mouse model of diet-induced MASLD/MASH to characterize their functions from simple steatosis to advanced MASH, using chow-fed mice as controls. Our findings show an inverse relationship between hepatic CM and PDM levels from healthy to steatosis to advanced MASH. Proteomics analysis revealed these two mitochondrial populations are compositionally and functionally distinct. We found that hepatic PDM are more bioenergetically active than CM, with higher pyruvate oxidation capacity in both healthy and diseased liver. Higher respiration capacity of PDM was associated with elevated OXPHOS protein complexes and increased TCA cycle flux. In contrast, CM showed higher fatty acid oxidation capacity with MASH progression. Transmission electron microscopy revealed larger and elongated mitochondria during healthy and early steatosis, which appeared small and fragmented during MASH progression. These changes coincided with higher MFN2 protein levels in hepatic PDM and higher DRP1 protein levels in hepatic CM. These findings highlight the distinct roles of hepatic CM and PDM in MASLD progression towards MASH.
Collapse
Affiliation(s)
- Noble Kumar Talari
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ushodaya Mattam
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Afra P Rahman
- Medical Sciences Baccalaureate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brook K Hemmelgarn
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael A Wyder
- Department of Cancer Biology, Proteomics Laboratory, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Pamela B Sylvestre
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kenneth D Greis
- Department of Cancer Biology, Proteomics Laboratory, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Karthickeyan Chella Krishnan
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
2
|
Luthra R, Sheth A. Understanding MASH: An Examination of Progression and Clinical Outcomes by Disease Severity in the TARGET-NASH Database. Adv Ther 2025; 42:1165-1195. [PMID: 39739194 PMCID: PMC11787050 DOI: 10.1007/s12325-024-03085-4] [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: 10/18/2024] [Accepted: 12/04/2024] [Indexed: 01/02/2025]
Abstract
INTRODUCTION Metabolic dysfunction-associated steatohepatitis (MASH), the progressive form of metabolic dysfunction-associated steatotic liver disease (MASLD), is linked to cardiometabolic risk factors such as obesity and type 2 diabetes (T2D). The rising prevalence of MASH and risk of hepatic and extra-hepatic complications emphasize the need for a better understanding of disease progression and associated outcomes. This study aimed to evaluate the incidence of, and demographic and clinical characteristics associated with, progression to MASH-related complications by disease severity in patients with non-cirrhotic MASH or MASH cirrhosis. Alignment between noninvasive tests (NITs) and biopsy-determined fibrosis stage was also assessed. METHODS This analysis used data from the TARGET-NASH cohort that includes adults with MASH across academic and community sites in the United States. Patients with non-cirrhotic MASH or MASH cirrhosis were stratified by disease severity based on fibrosis stage or cirrhosis. Progression to MASH-related outcomes, including all-cause mortality, cirrhosis, and liver transplantation, was assessed. RESULTS Among the 2378 patients included in this analysis, 48% had MASH cirrhosis. Incidence of all-cause mortality increased with disease severity from 0.14/100 person-months (100PM) at fibrosis stage 0-1 (F0-F1) to 2.02/100PM with compensated cirrhosis and 4.62/100PM with decompensated cirrhosis. Compared with patients with F0-F1, risk of progression to cirrhosis was higher in patients with F3 [hazard ratio (HR), 95% confidence interval (CI); 18.66, 10.97-31.73] and F2 (HR, 95% CI; 3.74, 2.00-6.98). Among those who progressed to MASH-related outcomes, 67.9% had T2D and 73.9% had hypertension. Vibration-controlled transient elastography showed better alignment with biopsy-determined fibrosis stage than Fibrosis-4 Index (FIB-4). CONCLUSIONS Progression to all-cause mortality in patients with MASH was significantly associated with the presence of higher fibrosis stage and cirrhosis. Cardiometabolic comorbidities such as T2D and hypertension were prevalent in patients with MASH progression. Early identification and management of MASH may mitigate disease progression and liver-related complications.
Collapse
Affiliation(s)
- Rakesh Luthra
- Novo Nordisk Inc., 800 Scudders Mill Rd, Plainsboro, NJ, 08536, USA
| | - Aarth Sheth
- Novo Nordisk Inc., 800 Scudders Mill Rd, Plainsboro, NJ, 08536, USA.
| |
Collapse
|
3
|
Singh A, Sohal A, Batta A. GLP-1, GIP/GLP-1, and GCGR/GLP-1 receptor agonists: Novel therapeutic agents for metabolic dysfunction-associated steatohepatitis. World J Gastroenterol 2024; 30:5205-5211. [PMID: 39735270 PMCID: PMC11612699 DOI: 10.3748/wjg.v30.i48.5205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/24/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
The global prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is estimated at 32.4%, reflecting its growing clinical significance. MASLD, which includes MASLD and metabolic dysfunction-associated steatohepatitis (MASH) has been linked to increased metabolic, cardiovascular, and malignant morbidity. Progression into fibrotic stages of MASLD is also strongly associated with liver-related mortality. The past few years have seen a heightened focus on creating innovative therapeutic strategies for MASH management. GLP-1 receptor agonists (RA) have also emerged as a potential treatment option. Studies on GLP-1 agonists, such as liraglutide and semaglutide, have demonstrated efficacy in MASH management, albeit with limited histological improvement of fibrosis. However, recent investigations into GLP-1/GIP RA (tirzepatide) and Glucagon/GLP-1 RA (survodutide) have shown even more encouraging results, with higher rates of MASH resolution and fibrosis improvement. The tolerability of these medications due to their gastrointestinal side effects remains a major concern. Future research should focus on optimizing drug regimens, identifying patients most likely to benefit, and balancing efficacy with tolerability. The evolving landscape of MASH therapeutics suggests a bright future, with the potential for combination therapies to further enhance patient outcomes.
Collapse
Affiliation(s)
- Anmol Singh
- Department of Medicine, Tristar Centennial Medical Center, Nashville, TN 37203, United States
| | - Aalam Sohal
- Division of Gastroenterology, Creighton University School of Medicine, Phoenix, AZ 85012, United States
| | - Akash Batta
- Department of Cardiology, Dayanand Medical College and Hospital, Ludhiana 141001, Punjab, India
| |
Collapse
|
4
|
Charlton M, Tonnu-Mihara I, Teng CC, Zhou Z, Asefaha F, Luthra R, Articolo A, Hoovler A, Uzoigwe C. Evaluating the burden of illness of metabolic dysfunction-associated steatohepatitis in a large managed care population: The ETHEREAL Study. J Manag Care Spec Pharm 2024; 30:1414-1430. [PMID: 39331041 PMCID: PMC11607210 DOI: 10.18553/jmcp.2024.24106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH; formerly nonalcoholic steatohepatitis) is the inflammatory form of metabolic dysfunction-associated steatotic liver disease (formerly nonalcoholic fatty liver disease). MASH is a progressive disease associated with increased risk for many hepatic and extra-hepatic complications such as cirrhosis, hepatocellular carcinoma, the requirement for liver transplantation, and cardiovascular (CV)-related and kidney-related complications. It is important to understand the clinical and economic burden of MASH. OBJECTIVES To assess and compare the clinical and economic burdens of MASH in adults with the non-MASH population in a real-world setting. METHODS This observational, retrospective study used the Healthcare Integrated Research Database (HIRD), which contains health care claims data for commercially insured and Medicare Advantage health plan members across the United States. All-cause, CV-related, and liver-related medical costs and health care resource utilization were evaluated in patients with at least 2 diagnoses of MASH during the patient identification period (October 1, 2016, to April 30, 2022) and compared with a non-MASH cohort 1:1 matched on age, Quan Charlson Comorbidity Index, region of residence, and health plan type and length of enrollment. Generalized linear regression with negative binomial and γ distribution models were used to compare health care resource utilization and medical costs, respectively, while controlling for confounders. Covariate-adjusted all-cause, CV-related, and liver-related hospitalization rate ratios and medical cost ratios were assessed and compared for the MASH and matched non-MASH cohorts. RESULTS A total of 18,549 patients with MASH were compared with 18,549 matched patients in the non-MASH cohort. After adjusting for covariates, MASH was associated with significantly higher rates of hospitalization and higher medical costs compared with the non-MASH cohort. When compared with the non-MASH cohort, patients with MASH had 1.22 (95% CI = 1.15-1.30; P < 0.0001) times higher rates of all-cause hospitalization, 1.13 (95% CI = 1.03-1.24; P = 0.008) times higher rates of CV-related hospitalization, and 7.22 (95% CI = 4.91-10.61; P < 0.0001) times higher rates of liver-related hospitalization. Similarly, all-cause medical costs were 1.26 (95% CI = 1.22-1.30; P < 0.0001) times higher, CV-related medical costs were 1.66 (95% CI = 1.59-1.73; P < 0.0001) times higher, and liver-related medical costs were 7.79 (95% CI = 7.42-8.17; P < 0.0001) times higher among patients with MASH. CONCLUSIONS Compared with those of the non-MASH cohort with similar age, Quan Charlson Comorbidity Index, health plan, region of residence, and duration of enrollment, patients with MASH had significantly higher all-cause, CV-related, and liver-related hospitalizations and medical costs.
Collapse
Affiliation(s)
| | | | | | | | | | - Rakesh Luthra
- Health Economics and Outcomes Research, Novo Nordisk Inc., Plainsboro, NJ
| | - Amy Articolo
- Medical Affairs, Novo Nordisk Inc., Plainsboro, NJ
| | | | | |
Collapse
|
5
|
Newsome PN, Sanyal AJ, Engebretsen KA, Kliers I, Østergaard L, Vanni D, Bugianesi E, Rinella ME, Roden M, Ratziu V. Semaglutide 2.4 mg in Participants With Metabolic Dysfunction-Associated Steatohepatitis: Baseline Characteristics and Design of the Phase 3 ESSENCE Trial. Aliment Pharmacol Ther 2024; 60:1525-1533. [PMID: 39412509 PMCID: PMC11599791 DOI: 10.1111/apt.18331] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 07/18/2024] [Accepted: 09/26/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Semaglutide, a glucagon-like peptide-1 receptor agonist, has demonstrated potential beneficial effects in metabolic dysfunction-associated steatohepatitis (MASH). AIMS To describe the trial design and baseline characteristics of the 'Effect of Semaglutide in Subjects with Non-cirrhotic Non-alcoholic Steatohepatitis' (ESSENCE) trial (NCT04822181). METHODS ESSENCE is a two-part, phase 3, randomised, multicentre trial evaluating the effect of subcutaneous semaglutide 2.4 mg in participants with biopsy-proven MASH and fibrosis stage 2 or 3. The primary objective of Part 1 is to demonstrate that semaglutide improves liver histology compared with placebo. The two primary endpoints are: resolution of steatohepatitis and no worsening of liver fibrosis, and improvement in liver fibrosis and no worsening of steatohepatitis. The Part 2 objective is based on clinical outcomes. The current work reports baseline characteristics of the first 800 randomised participants which includes demographics, laboratory parameters, liver histology, non-invasive tests and presence of metabolic dysfunction-associated steatotic liver disease (MASLD) cardiometabolic criteria. RESULTS Of 800 participants, 250 (31.3%) had fibrosis stage 2 and 550 (68.8%) had fibrosis stage 3. In the overall population, mean (standard deviation [SD]) age was 56 (11.6) years, 57.1% were female, mean (SD) body mass index was 34.6 (7.2) kg/m2, 55.5% had type 2 diabetes and > 99% had at least one MASLD cardiometabolic criterion according to the published definition. CONCLUSION The ESSENCE baseline population includes participants with clinically significant fibrosis stages 2 and 3. Although MASLD cardiometabolic criteria were not a requirement for study enrolment, almost all participants (> 99%) had at least one MASLD cardiometabolic criterion. TRIAL REGISTRATION NCT04822181.
Collapse
Affiliation(s)
- Philip N. Newsome
- Roger Williams Institute of Liver Studies, Faculty of Life Sciences and MedicineKing's College London and King's College HospitalLondonUK
- College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Arun J. Sanyal
- VCU School of MedicineStravitz‐Sanyal Institute for Liver Disease and Metabolic HealthRichmondVirginiaUSA
| | | | | | | | | | | | - Mary E. Rinella
- Division of Gastroenterology, Hepatology and NutritionUniversity of ChicagoChicagoIllinoisUSA
| | - Michael Roden
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfDüsseldorfGermany
- Institute for Clinical Diabetology, German Diabetes CenterLeibniz Center for Diabetes Research at Heinrich Heine University DüsseldorfDüsseldorfGermany
- German Center for Diabetes Research, Partner DüsseldorfMünchen‐NeuherbergGermany
| | - Vlad Ratziu
- Sorbonne Université, Institute for Cardiometabolism and NutritionHospital Pitié‐Salpêtrière, INSERM UMRS 1138 CRCParisFrance
| |
Collapse
|
6
|
Kitsios K, Trakatelli CM, Antza C, Triantafyllou A, Sarigianni M, Kotsis V. Treatment of Metabolic (Dysfunction)-Associated Fatty Liver Disease: Evidence from Randomized Controlled Trials-A Short Review. Metab Syndr Relat Disord 2024; 22:703-708. [PMID: 39088384 DOI: 10.1089/met.2024.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024] Open
Abstract
Metabolic-associated fatty liver disease (MALFD) is a highly prevalent and progressive disease, strongly related to obesity, metabolic syndrome, and cardiovascular disease. It comprises a spectrum of liver pathology from steatosis (fat accumulation in the hepatocytes) to steatosis with inflammation (metabolic-associated steatohepatitis, MASH), fibrosis, cirrhosis, and hepatocellular carcinoma. There is currently only one medication, resmetirom, US Food and Drug Administration approved for the treatment of MALFD. Evidence from randomized trials supports the efficacy of hypocaloric diets and exercise in MASH resolution. Moreover, substantial weight loss after bariatric surgery can lead to significant and longitudinally sustained MASH resolution, improvement in liver fibrosis, and decrease in the risk of major cardiovascular adverse events. Pioglitazone, an insulin sensitizer, initiated at the early stages, before the progression to fibrosis, may be effective in resolution of MASH in patients with or without type 2 diabetes. Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs), semaglutide and liraglutide, may also be effective in resolution of MASH but not of fibrosis. Preliminary data from interventions with tirzepatide, a dual GLP-1 and glucose-dependent insulinotropic polypeptide RA, and sodium-glucose cotransporter 2 inhibitors are encouraging, but more data based on liver biopsy are needed.
Collapse
Affiliation(s)
- Konstantinos Kitsios
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Christina-Maria Trakatelli
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Christina Antza
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Areti Triantafyllou
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Maria Sarigianni
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| | - Vasilios Kotsis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Thessaloniki, Greece
| |
Collapse
|
7
|
Shu YY, Hu LL, Ye J, Yang L, Jin Y. Rifaximin alleviates MCD diet-induced NASH in mice by restoring the gut microbiota and intestinal barrier. Life Sci 2024; 357:123095. [PMID: 39368771 DOI: 10.1016/j.lfs.2024.123095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/31/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024]
Abstract
AIMS Due to the increasing global incidence rate of nonalcoholic steatohepatitis (NASH) combined with the lack of effective treatment methods for this disease, there is an urgent need to find new treatment strategies. The aim of this study was to investigate the efficacy of rifaximin in preventing and treating NASH and the related mechanism. MATERIALS AND METHODS A NASH model was constructed by feeding male C57BL/6 mice a methionine-choline-deficient (MCD) diet for 4 weeks. Rifaximin was administered for 1 week before MCD diet feeding or during the last week of MCD diet feeding to investigate its preventive or therapeutic effects. Liver pathology, hepatic enzyme levels and metabolic indices were measured to evaluate the effects of rifaximin on NASH. Intestinal barrier integrity was measured via the Ussing chamber system and western blotting. 16S rDNA sequencing was conducted to investigate the fecal microbiota composition. Western blotting was performed to evaluate peroxisome proliferator activated receptor (PPAR)α and PPARγ protein levels. KEY FINDINGS Rifaximin effectively alleviated MCD diet-induced NASH. The microbiota composition in MCD diet-fed mice was significantly altered, and intestinal barrier integrity was disrupted. Dysbiosis and intestinal barrier dysfunction were reversed by rifaximin. In addition, rifaximin modulated PPARα and PPARγ expression in the liver. SIGNIFICANCE Rifaximin effectively alleviated MCD diet-induced NASH by restoring the gut microbiota and reversing intestinal barrier dysfunction, suggesting that rifaximin treatment is a new approach for preventing and treating NASH.
Collapse
Affiliation(s)
- Yan Yun Shu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Department of Ultrasound, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Li Lin Hu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Jin Ye
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Yu Jin
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| |
Collapse
|
8
|
Fishman JC, Qian C, Kim Y, Rochon H, Szabo SM, Sun R, Charlton M. Cost burden of cirrhosis and liver disease progression in metabolic dysfunction-associated steatohepatitis: A US cohort study. J Manag Care Spec Pharm 2024; 30:929-941. [PMID: 38845444 PMCID: PMC11365567 DOI: 10.18553/jmcp.2024.24069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH), formerly nonalcoholic steatohepatitis, is characterized by fat accumulation and inflammation of the liver and may result in progression to cirrhosis and liver-related events. OBJECTIVE To characterize the impact of cirrhosis and progression to liver-related events on costs and health care resource use (HCRU) among MASH patients in the United States. METHODS The study cohort included patients with diagnosed nonalcoholic steatohepatitis (International Classification of Diseases, Tenth Revision, Clinical Modification code K75.81) in Optum's deidentified Clinformatics Data Mart Database (October 2015 to December 2022) and were stratified by baseline cirrhosis status. Among those without cirrhosis at baseline, patients were further stratified by status of progression to cirrhosis during follow-up. Total HCRU and costs per-person per-year (PPPY) were estimated and compared descriptively between the cohorts. In addition, gamma generalized linear models were used to compare costs PPPY between those with vs without cirrhosis at baseline, as well as with vs without progression during follow-up, while adjusting for baseline patient and disease characteristics. Annual costs per person were also longitudinally modeled using gamma generalized linear mixed models to understand longitudinal changes in costs PPPY while accounting for time correlations within individual patients. Lastly, a series of sensitivity analyses were conducted to assess the impact of study design features and clinical variations of total costs PPPY. RESULTS A total of 28,576 adults were included, and 9,157 (32.0%) had baseline cirrhosis; of the 19,419 without baseline cirrhosis, a total of 4,235 (21.8%) progressed over follow-up. Mean (SD) HCRU and costs PPPY were higher among patients with cirrhosis ($110,403 [$226,037]) than without ($28,340 [$61,472]; P < 0.01) and among those with progression ($58,128 [$102,626]) than without ($20,031 [$39,740]; P < 0.01). Costs remained significantly greater when adjusted for covariates, with a risk ratio (95% CI) of 1.99 (1.89-2.09) when comparing with vs without baseline cirrhosis and 2.28 (2.15-2.42) when comparing with vs without progression over follow-up. Costs increased with each subsequent year, to 21% by year 6 among those with cirrhosis at baseline and 49% among those without baseline cirrhosis who progressed. CONCLUSIONS The financial burden of MASH is substantial and significantly greater among those with cirrhosis or disease progression. Although patients without cirrhosis incur lower burden, the increase over time is greater and associated with progression. Therapies that slow progression may help alleviate the financial burden, and strategies are needed to identify patients with MASH at risk of progressing to cirrhosis.
Collapse
Affiliation(s)
| | | | - Yestle Kim
- Madrigal Pharmaceuticals, Inc., West Conshohocken, PA
| | | | | | - Rosie Sun
- Broadstreet HEOR, Vancouver, BC, Canada
| | | |
Collapse
|
9
|
Huang M, Chen H, Wang H, Wang X, Wang D, Li Y, Zhou Q, Zhang D, Li M, Ma L. Worldwide burden of liver cancer due to metabolic dysfunction-associated steatohepatitis from 1990 to 2019: insights from the Global Burden of Disease study. Front Oncol 2024; 14:1424155. [PMID: 39267839 PMCID: PMC11390418 DOI: 10.3389/fonc.2024.1424155] [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: 05/09/2024] [Accepted: 08/02/2024] [Indexed: 09/15/2024] Open
Abstract
Introduction Metabolic dysfunction-associated steatohepatitis (MASH) is increasingly becoming a prevalent cause of hepatocellular carcinoma (HCC). Our study examines the burden of MASH-related HCC globally, regionally, and nationally, along with associated risk factors from 1990 to 2019, considering variables such as age, sex, and socioeconomic status. Objective We aimed to report the global, regional, and national burden of liver cancer due to MASH and its attributable risk factors between 1990 and 2019, by age, sex, and sociodemographic index (SDI). Methods Utilizing the Global Burden of Disease 2019 project, we analyzed data on prevalence, mortality, and disability-adjusted life years (DALYs) for liver cancer attributable to MASH across 204 countries. We provided counts and rates per 100,000 population, including 95% uncertainty intervals. Results In 2019, there were 46.8 thousand cases of MASH-related HCC, leading to 34.7 thousand deaths, and 795.8 thousand DALYs globally. While the prevalence increased by 19.8% since 1990, the death and DALY rates decreased by 5.3% and 15.1%, respectively. The highest prevalence was in High-income Asia Pacific, with the greatest increases observed in Australasia, Central Asia, and High-income North America. Southern Sub-Saharan Africa reported the highest death rate, while the lowest rates were in parts of Latin America, Central Sub-Saharan Africa, and Eastern Europe. DALY rates were the highest in Southern Sub-Saharan Africa and the lowest in Tropical Latin America. Discussion The burden of MASH-related HCC is expected to rise slightly over the next decade. This disease, which is not associated with the SDI, remains a major public health problem. In addition, the escalating rates of obesity, demographic shifts, and an aging population could position MASH as a leading factor in liver cancer cases, surpassing viral hepatitis. It is imperative, therefore, that the forthcoming years see the implementation of strategic interventions aimed at the early detection and prevention of liver cancer associated with MASH.
Collapse
Affiliation(s)
- Minshan Huang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| | - Hang Chen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| | - Hui Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| | - Xianmei Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| | - Da Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| | - Yu Li
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| | - Qingqing Zhou
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| | - Dan Zhang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| | - Mengwei Li
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| | - Lanqing Ma
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Kunming Medical University, Kunming, China
| |
Collapse
|
10
|
Younossi ZM, Mangla KK, Chandramouli AS, Lazarus JV. Estimating the economic impact of comorbidities in patients with MASH and defining high-cost burden in patients with noncirrhotic MASH. Hepatol Commun 2024; 8:e0488. [PMID: 39037377 PMCID: PMC11265778 DOI: 10.1097/hc9.0000000000000488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/21/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) is associated with high health care costs. This US study investigated the economic burden of MASH, particularly in patients without cirrhosis, and the impact of comorbidities on health care costs. METHODS This retrospective, observational study used data from patients diagnosed with MASH aged ≥18 years from October 2015 to March 2022 (IQVIA Ambulatory electronic medical record-US). Patients were stratified by the absence or presence of cirrhosis. Primary outcomes included baseline characteristics and annualized total health care cost after MASH diagnosis during follow-up. In addition, this study defined high costs for the MASH population and identified patient characteristics associated with increased health care costs among those without cirrhosis. RESULTS Overall, 16,919 patients (14,885 without cirrhosis and 2034 with cirrhosis) were included in the analysis. The prevalence of comorbidities was high in both groups; annual total health care costs were higher in patients with cirrhosis. Patients with a high-cost burden (threshold defined using the United States national estimated annual health care expenditure of $13,555) had a higher prevalence of comorbidities and were prescribed more cardiovascular medications. MASH diagnosis was associated with an increase in cost, largely driven by inpatient costs. In patients without cirrhosis, an increase in cost following MASH diagnosis was associated with the presence and burden of comorbidities and cardiovascular medication utilization. CONCLUSIONS Comorbidities, such as cardiovascular disease and type 2 diabetes, are associated with a higher cost burden and may be aggravated by MASH. Prioritization and active management may benefit patients without cirrhosis with these comorbidities. Clinical care should focus on preventing progression to cirrhosis and managing high-burden comorbidities.
Collapse
Affiliation(s)
- Zobair M. Younossi
- The Global NASH Council, Washington, District of Columbia, USA
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
| | - Kamal Kant Mangla
- Novo Nordisk Service Center India Pvt Ltd, Bangalore, Karnataka, India
| | | | - Jeffrey V. Lazarus
- The Global NASH Council, Washington, District of Columbia, USA
- City University of New York Graduate School of Public Health and Health Policy (CUNY SPH), New York, New York, USA
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic, University of Barcelona, Barcelona, Spain
| |
Collapse
|
11
|
Sanyal AJ, Bedossa P, Fraessdorf M, Neff GW, Lawitz E, Bugianesi E, Anstee QM, Hussain SA, Newsome PN, Ratziu V, Hosseini-Tabatabaei A, Schattenberg JM, Noureddin M, Alkhouri N, Younes R. A Phase 2 Randomized Trial of Survodutide in MASH and Fibrosis. N Engl J Med 2024; 391:311-319. [PMID: 38847460 DOI: 10.1056/nejmoa2401755] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
BACKGROUND Dual agonism of glucagon receptor and glucagon-like peptide-1 (GLP-1) receptor may be more effective than GLP-1 receptor agonism alone for treating metabolic dysfunction-associated steatohepatitis (MASH). The efficacy and safety of survodutide (a dual agonist of glucagon receptor and GLP-1 receptor) in persons with MASH and liver fibrosis are unclear. METHODS In this 48-week, phase 2 trial, we randomly assigned adults with biopsy-confirmed MASH and fibrosis stage F1 through F3 in a 1:1:1:1 ratio to receive once-weekly subcutaneous injections of survodutide at a dose of 2.4, 4.8, or 6.0 mg or placebo. The trial had two phases: a 24-week rapid-dose-escalation phase, followed by a 24-week maintenance phase. The primary end point was histologic improvement (reduction) in MASH with no worsening of fibrosis. Secondary end points included a decrease in liver fat content by at least 30% and biopsy-assessed improvement (reduction) in fibrosis by at least one stage. RESULTS A total of 293 randomly assigned participants received at least one dose of survodutide or placebo. Improvement in MASH with no worsening of fibrosis occurred in 47% of the participants in the survodutide 2.4-mg group, 62% of those in the 4.8-mg group, and 43% of those in the 6.0-mg group, as compared with 14% of those in the placebo group (P<0.001 for the quadratic dose-response curve as best-fitting model). A decrease in liver fat content by at least 30% occurred in 63% of the participants in the survodutide 2.4-mg group, 67% of those in the 4.8-mg group, 57% of those in the 6.0-mg group, and 14% of those in the placebo group; improvement in fibrosis by at least one stage occurred in 34%, 36%, 34%, and 22%, respectively. Adverse events that were more frequent with survodutide than with placebo included nausea (66% vs. 23%), diarrhea (49% vs. 23%), and vomiting (41% vs. 4%); serious adverse events occurred in 8% with survodutide and 7% with placebo. CONCLUSIONS Survodutide was superior to placebo with respect to improvement in MASH without worsening of fibrosis, warranting further investigation in phase 3 trials. (Funded by Boehringer Ingelheim; 1404-0043 ClinicalTrials.gov number, NCT04771273; EudraCT number, 2020-002723-11.).
Collapse
Affiliation(s)
- Arun J Sanyal
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Pierre Bedossa
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Mandy Fraessdorf
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Guy W Neff
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Eric Lawitz
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Elisabetta Bugianesi
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Quentin M Anstee
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Samina Ajaz Hussain
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Philip N Newsome
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Vlad Ratziu
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Azadeh Hosseini-Tabatabaei
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Jörn M Schattenberg
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Mazen Noureddin
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Naim Alkhouri
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| | - Ramy Younes
- From the Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond (A.J.S.); Liverpat and University of Paris (P.B.), and Sorbonne Université, Institute for Cardiometabolism and Nutrition, Assistance Publique-Hôpitaux de Paris, INSERM Unité Mixte de Recherche Scientifique 1138, Centre de Recherche des Cordeliers (V.R.) - all in Paris; Boehringer Ingelheim, Ingelheim am Rhein (M.F., S.A.H and R.Y.), Saarland University Medical Center, Homburg (J.M.S.), and University of the Saarland, Saarbrücken (J.M.S.) - all in Germany; Covenant Metabolic Specialists, Sarasota, FL (G.W.N.); Houston Methodist Hospital and Houston Research Institute, Houston (M.N.), and the Texas Liver Institute, University of Texas Health San Antonio, San Antonio (E.L. and N.A.) - all in Texas; the University of Turin, Turin, Italy (E.B.); the Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, and Newcastle National Institute for Health and Care Research Biomedical Research Centre, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne (Q.M.A.), the University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham (P.N.N.), and the Institute of Hepatology, Faculty of Life Sciences and Medicine, King's College London and King's College Hospital, London (P.N.N.) - all in the United Kingdom; Boehringer Ingelheim Pharmaceuticals, Ridgefield, CT (A.H.-T.); and Arizona Liver Health, Chandler (N.A.)
| |
Collapse
|
12
|
Su M, Yan Z, Wang Y, Cai J, Dong J, Luo J, Chen D, Liu A, Ye H. Depression Exacerbates Hepatic Steatosis in C57BL/6J Mice by Activating the Hypothalamic-Pituitary-Adrenal Axis. In Vivo 2024; 38:1677-1689. [PMID: 38936893 PMCID: PMC11215574 DOI: 10.21873/invivo.13618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 06/29/2024]
Abstract
BACKGROUND/AIM Depression is associated with metabolic disorders, including non-alcoholic fatty liver disease (NAFLD). However, the mechanisms underlying the interaction between them are still poorly known. MATERIALS AND METHODS In this study, mice on a choline deficiency, L-amino acid-defined, high-fat diet (CDAHFD) developing steatosis were challenged with chronic restraint stress (CRS), a protocol widely used to induce depression. The development of depression and steatosis was evaluated using histopathology analysis, ELISA, q-PCR and Western Blot. RESULTS The contribution of the activated HPA axis to hepatic steatosis progress was fully established, which was validated using a hepatocyte model. Histopathological and biochemical analysis indicated that steatosis was exacerbated by CRS challenge, and behavioral tests indicated that the mice developed depression. Among the screened endocrinal pathways, the hypothalamic-pituitary-adrenal (HPA) axis was significantly activated and the synergistic effect of CDAHFD and CRS in activating the HPA axis was observed. In the hypothalamus, expression of corticotropin-releasing hormone (CRH) was increased by 86% and the protein levels of hypothalamic CRH were upregulated by 25% to 33% by CRS treatment. Plasma CRH levels were elevated by 45-56% and plasma adrenocorticotropic hormone (ACTH) levels were elevated by 29-58% by CRS treatment. In the liver, target genes of the HPA axis were activated, accompanied by disruption of the lipid metabolism and progression of steatohepatitis. The lipid metabolism in the Hepa1-6 cell line treated with endogenous corticosterone (CORT) was in accordance with the aforementioned in vivo responses. CONCLUSION Depression aggravated hepatic steatosis in CDAHFD-fed mice by activating the HPA axis. The risk of NAFLD development should be fully considered in depressive patients and improvement of psychotic disorders could be an etiological treatment strategy for them.
Collapse
Affiliation(s)
- Mingli Su
- Department of Gastroenterology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo, P.R. China
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Zheng Yan
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Ying Wang
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Jiacheng Cai
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Jia Dong
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Jia Luo
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Dahua Chen
- Department of Gastroenterology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo, P.R. China
| | - Aiming Liu
- Zhejiang Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, Health Science Center, Ningbo University, Ningbo, P.R. China
| | - Hua Ye
- Department of Gastroenterology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo, P.R. China;
| |
Collapse
|
13
|
Weber EJ, Younis IR, Wang L, Xiao D, Barchuk WT, Othman AA. Evaluation of the Effects of Meal Type and Acid-Reducing Agents on the Pharmacokinetics of Cilofexor, a Selective Nonsteroidal Farnesoid X Receptor Agonist. Clin Pharmacol Drug Dev 2024; 13:677-687. [PMID: 38346861 DOI: 10.1002/cpdd.1384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/17/2024] [Indexed: 06/02/2024]
Abstract
Cilofexor is a nonsteroidal farnesoid X receptor agonist being developed in combination with firsocostat/semaglutide for the treatment of nonalcoholic steatohepatitis. This phase 1 study evaluated the effects of food and acid-reducing agents (ARAs) on the pharmacokinetics of cilofexor (100- or 30-mg fixed-dose combination with firsocostat) in healthy participants. Cohorts 1 (n = 20, 100 mg) and 2 (n = 30, 30 mg) followed a 3-period, 2-sequence crossover design and evaluated effects of light-fat and high-fat meals. Cohort 3 (n = 30, 100 mg fasting) followed a 2-period, 2-sequence crossover design and evaluated the effects of a 40-mg single dose of famotidine. Cohort 4 (n = 18, 100 mg) followed a 3-period, 2-sequence crossover design and evaluated the effects of a 40-mg once-daily regimen of omeprazole administered under fasting conditions or following a light-fat meal. Administration with light-fat or high-fat meals resulted in no change and an ∼35% reduction in cilofexor AUC, respectively, relative to the fasting conditions. Under fasting conditions, famotidine increased cilofexor AUC by 3.2-fold and Cmax by 6.1-fold, while omeprazole increased cilofexor AUC by 3.1-fold and Cmax by 4.8-fold. With a low-fat meal, omeprazole increased cilofexor exposure to a lesser extent (Cmax 2.5-fold, AUC 2.1-fold) than fasting conditions. This study suggests that caution should be exercised when cilofexor is administered with ARAs under fed conditions; coadministration of cilofexor (100 or 30 mg) with ARAs under fasting conditions is not recommended with the current clinical trial formulations.
Collapse
Affiliation(s)
| | | | - Lulu Wang
- Gilead Sciences, Inc., Foster City, CA, USA
| | | | | | | |
Collapse
|
14
|
Wang B, Yu H, Gao J, Yang L, Zhang Y, Yuan X, Zhang Y. Machine learning deciphers the significance of mitochondrial regulators on the diagnosis and subtype classification in non-alcoholic fatty liver disease. Heliyon 2024; 10:e29860. [PMID: 38707433 PMCID: PMC11066337 DOI: 10.1016/j.heliyon.2024.e29860] [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: 09/21/2023] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is a highly prevalent liver disease worldwide and lack of research on the diagnostic utility of mitochondrial regulators in NAFLD. Mitochondrial dysfunction plays a pivotal role in the development and progression of NAFLD, especially oxidative stress and acidity β-oxidative overload. Thus, we aimed to identify and validate a panel of mitochondrial gene expression biomarkers for detection of NAFLD. Methods We selected the GSE89632 dataset and identified key mitochondrial regulators by intersecting DEGs, WGCNA modules, and MRGs. Classification of NAFLD subtypes based on these key mitochondrial regulatory factors was performed, and the pattern of immune system infiltration in different NAFLD subtypes were also investigated. RF, LASSO, and SVM-RFE were employed to identify possible diagnostic biomarkers from key mitochondrial regulatory factors and the predictive power was demonstrated through ROC curves. Finally, we validated these potential diagnostic biomarkers in human peripheral blood samples and a high-fat diet-induced NAFLD mouse model. Results We identified 25 key regulators of mitochondria and two NAFLD subtypes with different immune infiltration patterns. Four potential diagnostic biomarkers (BCL2L11, NAGS, HDHD3, and RMND1) were screened by three machine learning methods thereby establishing the diagnostic model, which showed favorable predictive power and achieved significant clinical benefit at certain threshold probabilities. Then, through internal and external validation, we identified and confirmed that BCL2L11 was significantly downregulated in NAFLD, while the other three were significantly upregulated. Conclusion The four MRGs, namely BCL2L11, NAGS, HDHD3, and RMND1, are novel potential biomarkers for diagnosing NAFLD. A diagnostic model constructed using the four MRGs may aid early diagnosis of NAFLD in clinics.
Collapse
Affiliation(s)
- Bingyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | | | - Jiawei Gao
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Liuxin Yang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yali Zhang
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
- Zhang Yali Famous Traditional Chinese Medicine Expert Studio, Harbin, China
| | - Xingxing Yuan
- Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Yang Zhang
- Department of Gastroenterology, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
15
|
Wang T, Wang D, Kuang G, Gong X, Zhang L, Wan J, Li K. Derlin-1 promotes diet-induced non-alcoholic fatty liver disease via increasing RIPK3-mediated necroptosis. Free Radic Biol Med 2024; 217:29-47. [PMID: 38522486 DOI: 10.1016/j.freeradbiomed.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/29/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND & AIMS Unrestricted endoplasmic reticulum (ER) stress and the continuous activation of ER associated protein degradation (ERAD) pathway might lead to the aggravation of non-alcoholic steatohepatitis (NASH). Derlin-1 has been considered to be an integral part of the ERAD pathway, which is involved in the regulation of the transport and excretion of protein degradation products within ER. However, the regulatory role and mechanism of Derlin-1 in NASH remains unclear. METHODS The expression of Derlin-1 was firstly detected in the liver of normal and NASH animal model and patient. Then, western diet (WD)-induced NASH mice were administrated with the lentivirus-mediated Derlin-1 knockdown or overexpression. Finally, RIPK3 knockout mice were used to explore the mechanism. The liver injury, hepatic steatosis, inflammation, and fibrosis as well as ER stress signal pathway were evaluated. RESULTS The levels of Derlin-1 were significantly elevated in the liver of WD-fed mice and NASH patients when compared to the control group. Furthermore, Derlin-1 knockdown attenuated WD-induced liver injury, lipid accumulation, inflammatory response, and fibrosis. Conversely, overexpression of Derlin-1 presented the completely opposite results. Mechanistically, Derlin-1 enhanced ER stress pathways and led to necroptosis, and RIPK3 knockout dramatically reduced Derlin-1 expression and reversed the progression of NASH aggravated by Derlin-1. CONCLUSIONS Notably, Derlin-1 is a critical modulator in NASH. It may accelerate the progression of NASH by regulating the activation of the ERAD pathway and further aggravating the ER stress, which might be involved in RIPK3-mediated necroptosis. Therefore, targeting Derlin-1 as a novel intervention point holds the potential to delay or even reverse NASH.
Collapse
Affiliation(s)
- Ting Wang
- Department of Orthopedics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Dehua Wang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Ge Kuang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China.
| | - Xia Gong
- Department of Anatomy, Chongqing Medical University, Chongqing, China.
| | - Li Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Jingyuan Wan
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China; Department of Pharmacology, Chongqing Medical University, Chongqing, China.
| | - Ke Li
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
16
|
Drapkina OM, Kontsevaya AV, Kalinina AM, Avdeev SN, Agaltsov MV, Alekseeva LI, Almazova II, Andreenko EY, Antipushina DN, Balanova YA, Berns SA, Budnevsky AV, Gainitdinova VV, Garanin AA, Gorbunov VM, Gorshkov AY, Grigorenko EA, Jonova BY, Drozdova LY, Druk IV, Eliashevich SO, Eliseev MS, Zharylkasynova GZ, Zabrovskaya SA, Imaeva AE, Kamilova UK, Kaprin AD, Kobalava ZD, Korsunsky DV, Kulikova OV, Kurekhyan AS, Kutishenko NP, Lavrenova EA, Lopatina MV, Lukina YV, Lukyanov MM, Lyusina EO, Mamedov MN, Mardanov BU, Mareev YV, Martsevich SY, Mitkovskaya NP, Myasnikov RP, Nebieridze DV, Orlov SA, Pereverzeva KG, Popovkina OE, Potievskaya VI, Skripnikova IA, Smirnova MI, Sooronbaev TM, Toroptsova NV, Khailova ZV, Khoronenko VE, Chashchin MG, Chernik TA, Shalnova SA, Shapovalova MM, Shepel RN, Sheptulina AF, Shishkova VN, Yuldashova RU, Yavelov IS, Yakushin SS. Comorbidity of patients with noncommunicable diseases in general practice. Eurasian guidelines. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2024; 23:3696. [DOI: 10.15829/1728-8800-2024-3996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Создание руководства поддержано Советом по терапевтическим наукам отделения клинической медицины Российской академии наук.
Collapse
|
17
|
Tamura K, Ito K, Kishimoto R, Yoshida K, Kishimoto T, Obata T, Yamaguchi T. The Effect of Steatosis on Shear-Wave Velocity and Viscoelastic Properties Related to Liver Fibrosis Progression in Rat Models. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:592-599. [PMID: 38238201 DOI: 10.1016/j.ultrasmedbio.2024.01.002] [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: 07/31/2023] [Revised: 12/26/2023] [Accepted: 01/01/2024] [Indexed: 02/17/2024]
Abstract
OBJECTIVE Hepatic fibrosis has recently been evaluated using ultrasonography or magnetic resonance elastography. Although the shear wave velocity (SWV) obtained using point shear wave elastography (pSWE) provides a valuable measure of fibrosis, underlying steatosis may affect its measurement. METHODS Using hepatic fibrosis samples, this study evaluated the effect of steatosis on the shear wave velocity of pSWE (Vs) and viscoelastic properties (assessed by dynamic mechanical analysis) of rat liver. Fifty rats with various grades of steatosis and fibrosis underwent open abdominal in vivo Vs measurements using a commercial ultrasound scanner. The mechanical properties of hepatic tissue were also characterized under ex vivo conditions using dynamic mechanical analysis and the Zener model of viscoelasticity. RESULTS Fibrosis and steatosis progression influenced Vs and elasticity. The SWV computed using the Zener model and Vs showed a substantial correlation (r > 0.8). Fibrosis progression increased SWV. Steatosis was also related to SWV. Steatosis progression obscured the SWV change associated with fibrosis progression. CONCLUSION We conclude that steatosis progression affects the evaluation of fibrosis progression. This finding could aid discrimination of non-alcoholic steatohepatitis from non-alcoholic fatty liver disease using SWV.
Collapse
Affiliation(s)
- Kazuki Tamura
- Preeminent Medical Photonics Education & Research, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan.
| | - Kazuyo Ito
- Institute of Engineering, Tokyo University of Agriculture and Technology, Koganei-shi, Tokyo 184-8588 Japan
| | - Riwa Kishimoto
- Applied MRI Research, Department of Molecular Imaging and Theranostics, National Institutes for Quantum and Radiological Science and Technology, Inage-ku, Chiba 263-0024, Japan
| | - Kenji Yoshida
- Center for Frontier Medical Engineering, Chiba University, Inage-ku, Chiba 263-8522, Japan
| | - Takashi Kishimoto
- Department of Molecular Pathology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | - Takayuki Obata
- Applied MRI Research, Department of Molecular Imaging and Theranostics, National Institutes for Quantum and Radiological Science and Technology, Inage-ku, Chiba 263-0024, Japan
| | - Tadashi Yamaguchi
- Center for Frontier Medical Engineering, Chiba University, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
18
|
Aller R, Calleja JL, Crespo J, Romero-Gómez M, Turnes J, Benmarzouk-Hidalgo OJ, Subirán R, Gil A. Advanced fibrosis associated with non-alcoholic steatohepatitis (NASH) in Spain: results of a Delphi study. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:337-346. [PMID: 37343722 DOI: 10.1016/j.gastrohep.2023.06.005] [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: 12/23/2022] [Revised: 05/18/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023]
Abstract
OBJECTIVE To describe in detail the epidemiology, diagnosis, clinical management, treatment options, impact on quality of life and unmet needs of patients with advanced liver fibrosis (F3-F4) associated with non-alcoholic steatohepatitis (NASH) in Spain. METHODOLOGY Delphi study of two rounds of consultation rounds with 41 expert hepatologists from 16 autonomous communities to collect their experience in clinical practice. RESULTS The estimated prevalence of adult patients diagnosed with F3-F4 fibrosis associated with NASH in Spain is 0.019% (95% confidence interval [CI]: 0.019-0.020%). Approximately 7,588 adults with this condition are currently diagnosed and managed in the Digestive System Services of Spanish hospitals, and around 1,881 new patients are diagnosed each year. Management is multidisciplinary and includes the specialties of Digestive System, Endocrinology and Internal Medicine, considering the frequently associated metabolic comorbidities (obesity, type 2 diabetes mellitus or dysmetabolic iron overload). Despite a clear impact on quality of life, this it is not routinely evaluated in clinical practice. The most widely used non-invasive diagnostic techniques are transitional elastography and liver fibrosis index 4 (FIB-4). The absence of effective and safe treatments appears as the main unmet need for the management of these patients. CONCLUSIONS This study provides a representation of the current situation of patients diagnosed with F3-F4 fibrosis associated with NASH in Spain, increasing the evidence available and contributing to informed decision-making by professionals and the health system.
Collapse
Affiliation(s)
- Rocío Aller
- Servicio de Gastroenterología, Hospital Clínico Universitario de Valladolid, Valladolid, España
| | - José Luis Calleja
- Servicio de Gastroenterología, Hospital Universitario Puerta de Hierro, Madrid, España
| | - Javier Crespo
- Servicio de Gastroenterología, Hospital Universitario Marqués de Valdecilla, Santander, España
| | - Manuel Romero-Gómez
- Servicio de Gastroenterología, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Juan Turnes
- Servicio de Gastroenterología, Complejo Hospitalario de Pontevedra, Pontevedra, España
| | | | | | - Alicia Gil
- Omakase Consulting S.L., Barcelona, España.
| |
Collapse
|
19
|
Ravaut G, Carneiro A, Mounier C. Exploring the impacts of ketogenic diet on reversible hepatic steatosis: initial analysis in male mice. Front Nutr 2024; 11:1290540. [PMID: 38577162 PMCID: PMC10991688 DOI: 10.3389/fnut.2024.1290540] [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: 09/07/2023] [Accepted: 02/27/2024] [Indexed: 04/06/2024] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is the most common chronic liver disease. Ketogenic diet (KD), a diet with very low intake in carbohydrates, gained popularity as a weight-loss approach. However, in mice models, it has been reported that an excess exposition of dietary fat induces hepatic insulin resistance and steatosis. However, data published is inconsistent. Herein, we investigated in a mouse model, the metabolic effects of KD and its contribution to the pathogenesis of NALFD. Mice were exposed to KD or CHOW diet for 12 weeks while a third group was exposed to KD for also 12 weeks and then switched to CHOW diet for 4 weeks to determine if we can rescue the phenotype. We evaluated the effects of diet treatments on fat distribution, glucose, and insulin homeostasis as well as hepatic steatosis. Mice fed with KD developed glucose intolerance but not insulin resistance accompanied by an increase of inflammation. KD-fed mice showed an increase of fat accumulation in white adipose tissue and liver. This effect could be explained by an increase in fat uptake by the liver with no changes of catabolism leading to MAFLD. Interestingly, we were able to rescue the phenotype by switching KD-fed mice for 4 weeks on a CHOW diet. Our studies demonstrate that even if mice develop hepatic steatosis and glucose intolerance after 12 weeks of KD, they do not develop insulin resistance and more importantly, the phenotype can be reversed by switching the mice from a KD to a CHOW.
Collapse
Affiliation(s)
| | | | - Catherine Mounier
- CERMO-FC Research Center, Molecular Metabolism of Lipids Laboratory, Biological Sciences Department, University of Quebec in Montreal (UQAM), Montreal, QC, Canada
| |
Collapse
|
20
|
Xue Y, Wei Y, Cao L, Shi M, Sheng J, Xiao Q, Cheng Z, Luo T, Jiao Q, Wu A, Chen C, Zhong L, Zhang C. Protective effects of scutellaria-coptis herb couple against non-alcoholic steatohepatitis via activating NRF2 and FXR pathways in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116933. [PMID: 37482263 DOI: 10.1016/j.jep.2023.116933] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellaria-coptis herb couple (SC) is a classic herbal pair used in many Traditional Chinese Medicine (TCM) formulations in the treatment of endocrine and metabolic deseases. Diabetes mellitus and non-alcoholic steatohepatitis (NASH) are both endocrine and metabolic diseases. Previous studies have shown that SC has anti-diabetic effects. However, the effect and mechanism of SC against NASH remains unclear. AIM OF THE STUDY This study aimed to demonstrate the effect and mechanism of SC against NASH through the nuclear factor-erythroid 2-related factor 2 (Nrf2) and farnesoid X receptor (FXR) dual signaling pathways in vivo and in vitro. MATERIALS AND METHODS The high fat diet-fed rat model, and HepG2 and RAW264.7 cell models were used. Serum biochemical indexes and liver histopathological changes were examined. Metabolomics, transcriptomics, and flow cytometry were performed. RT-qPCR and western blot analysis were performed to provide expression of NRF2 and FXR pathway signal molecules during SC's anti-NASH treatment in vivo and in vitro. RESULTS SC had anti-NASH effects in vivo with significantly improvement of serum NASH biochemical index and hepatopathological structure; meanwhile, SC significantly elevated the expression levels of FXR protein in liver and intestinal tissues, and cholesterol 7a-hydroxylase (CYP7A1) protein in liver. The mRNA expression levels of Takeda G protein receptor 5 (TGR5), CYP7A1, fibroblast growth factor receptor-4 (FGFR4), FXR, small heterodimer partner (SHP), fibroblast growth factor 15/19 (FGF15/19) and glucagon-like peptide-1 (GLP-1) were significantly elevated by SC. SC reduced the levels of NorCA, isoLCA and α-MCA in the feces of NAFLD rats. In vitro, SC-containing serum (SC-CS) was found to significantly reduce intracellular lipid deposition, inhibit ROS production, reduce intracellular Malondialdehyde (MDA) and IL-1β levels, and enhance the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Six differential genes closely related to oxidative stress and Nrf2 were identified by transcriptomic analysis. SC-CS up-regulated the expression of NRF2, and reduced the expression of TXNIP and Caspase-1 genes in RAW264.7 cells. In addition, SC-CS reduced the expression of Keap1 and NF-κB, and up-regulated the expression of Nrf2, heme oxygenase-1 (HO-1), quinone oxidoreductase 1 (NQO1), and SOD; SC-CS elevated the protein level of NRF2, and reduced the protein level of TXNIP in HepG2 cells. CONCLUSIONS the mechanisms of SC action against NASH was closely related to the simultaneous activations of both NRF2 and FXR signaling pathways. These findings provide a new insight into the anti-NASH application of SC in clinical settings and demonstrate the potential of SC in the treatment of NASH.
Collapse
Affiliation(s)
- Yanan Xue
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Yue Wei
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Lan Cao
- Research Center of Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine, PR China
| | - Min Shi
- College of Life Science, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Junqing Sheng
- College of Life Science, Nanchang University, Nanchang, 330031, PR China
| | - Qin Xiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Ziwen Cheng
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Tao Luo
- First Affiliated Hospital of Nanchang University, 330006, PR China
| | - Quanhui Jiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Ailan Wu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Lingyun Zhong
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Changhua Zhang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China; Nanchang Research Institute, Sun Yat-sen University, Jiangxi, 330096, PR China.
| |
Collapse
|
21
|
Lakkis NA, Mokalled NM, Osman MH, Musharrafieh UM, Eljammal M. Liver Cancer and Risk Factors in the MENA Region: Epidemiology and Temporal Trends Based on the 2019 Global Burden of Disease Data. Cancer Control 2024; 31:10732748241297346. [PMID: 39487107 PMCID: PMC11531006 DOI: 10.1177/10732748241297346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/24/2024] [Accepted: 10/10/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Liver cancer (LivCa) is a growing concern in the MENA region, driven by diverse factors, including viral hepatitis, lifestyle-related risks, and other causes. METHODS Utilizing GBD 2019 data, we assessed LivCa patterns, emphasizing chronic viral hepatitis, non-viral factors, and health care disparities across the MENA region. RESULTS Rising LivCa rates, particularly related to chronic viral hepatitis, highlight the region's health challenges. Lifestyle factors, such as obesity and diabetes, contribute significantly. Disparities in health care access and cancer registration hinder accurate assessments. CONCLUSION A comprehensive strategy is vital, encompassing vaccination promotion, health care enhancements, and lifestyle awareness. Urgent coordinated efforts are needed to address disparities, implement evidence-based interventions, and alleviate the escalating LivCa burden in the MENA region.
Collapse
Affiliation(s)
- Najla A. Lakkis
- Department of Family Medicine, American University of Beirut Medical Center (AUBMC), Beirut, Lebanon
| | - Nour M. Mokalled
- Department of Internal Medicine, Hematology-Oncology, American University of Beirut (AUB), Beirut, Lebanon
| | - Mona H. Osman
- Department of Family Medicine, American University of Beirut Medical Center (AUBMC), Beirut, Lebanon
| | - Umayya M. Musharrafieh
- Department of Family Medicine, American University of Beirut Medical Center (AUBMC), Beirut, Lebanon
| | - Mohammad Eljammal
- Faculty of Medicine, American University of Beirut Medical Center (AUBMC), Beirut, Lebanon
| |
Collapse
|
22
|
Mac Curtain BM, O'Brien L, El Sherif O, Mc Cormack A, Carolan E, Ryan JD, O'Shea D, Gallagher TK. Biguanides and glucagon like peptide 1 receptor agonists in the amelioration of post liver transplant weight gain; a scoping review of the mechanism of action, safety and efficacy. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2024; 17:17-27. [PMID: 38737926 PMCID: PMC11080689 DOI: 10.22037/ghfbb.v17i1.2899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/02/2023] [Indexed: 05/14/2024]
Abstract
Weight gain post-liver transplant can lead to adverse patient outcomes in the post-transplant period. Pharmacotherapy and other measures can be utilised to reduce the burden and occurrence of weight gain in this population. We explored the mechanism of action, safety, and efficacy of these medications, specifically GLP-1 receptor agonists and metformin, focusing on liver transplant patients. This scoping review was conducted in line with the scoping review structure as outlined by the PRISMA guidelines. Metformin and GLP-1 receptor agonists have been observed to be safe and effective in liver transplant patients. Experimental models have found liver-centric weight loss mechanisms in this drug cohort. There is a paucity of evidence about the use of antihyperglycemics in a post-transplant population for weight loss purposes. However, some small studies have shown strong safety and efficacy data. The evidence in relation to using these medications in patients with metabolic syndrome for weight loss warrants further study in a transplant population.
Collapse
Affiliation(s)
| | - Luke O'Brien
- Hepatopancreatobiliary Group, St Vincent's University Hospital, Dublin 4, Ireland
| | - Omar El Sherif
- National Liver Transplant Unit, St Vincent's University Hospital, Dublin 4, Ireland
| | - Aidan Mc Cormack
- National Liver Transplant Unit, St Vincent's University Hospital, Dublin 4, Ireland
| | - Emer Carolan
- Department of Hepatology, Beaumont Hospital, Dublin 9, Ireland
| | - John D Ryan
- Department of Hepatology, Beaumont Hospital, Dublin 9, Ireland
| | - Donal O'Shea
- Department of Endocrinology, St Vincent's University Hospital, Dublin 4, Ireland
| | - Tom K Gallagher
- Hepatopancreatobiliary Group, St Vincent's University Hospital, Dublin 4, Ireland
- National Liver Transplant Unit, St Vincent's University Hospital, Dublin 4, Ireland
| |
Collapse
|
23
|
Xue Y, Wei Y, Cao L, Shi M, Sheng J, Xiao Q, Cheng Z, Luo T, Jiao Q, Wu A, Chen C, Zhong L, Zhang C. Protective effects of scutellaria-coptis herb couple against non-alcoholic steatohepatitis via activating NRF2 and FXR pathways in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116933. [DOI: https:/doi.org/10.1016/j.jep.2023.116933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2024]
|
24
|
Gal P, Feldmajer G, Augusto M, Gani R, Hook E, Bullement A, Philips Z, Smith I. De Novo Cost-Effectiveness Model Framework for Nonalcoholic Steatohepatitis-Modeling Approach and Validation. PHARMACOECONOMICS 2023; 41:1629-1639. [PMID: 37505423 PMCID: PMC10635953 DOI: 10.1007/s40273-023-01298-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/18/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Nonalcoholic steatohepatitis (NASH) is a chronic liver disease associated with hepatic morbidity and mortality and extra-hepatic comorbidities. Published NASH cost-effectiveness models (CEMs) are heterogeneous and consistently omit comorbid conditions that frequently co-exist alongside NASH. We aimed to develop a de novo CEM framework that incorporates extra-hepatic disease states and outcomes alongside hepatic components to enable future estimation of the cost-effectiveness of NASH interventions. METHODS Patient-level simulation and cohort-level Markov models were implemented in the same framework. Model inputs included fibrosis progression; late-stage liver disease outcomes; comorbidity outcomes for cardiovascular disease, type 2 diabetes, and obesity; mortality; health-related quality of life; and direct medical costs. The prototype analysis assessed the cost-effectiveness of obeticholic acid versus standard of care from a US payer perspective over a lifetime horizon with costs and effects discounted at 3% per annum. However, the CEM was designed for easy adaptation to other countries, time horizons, and other considerations. Efficacy and adverse event parameters were obtained from the 18-month interim analysis of the REGENERATE trial. Outputs include total and incremental costs, total life years, and quality-adjusted life years. RESULTS In this model, total costs, total life years, and quality-adjusted life years were all higher with obeticholic acid compared with standard of care. Cross-validation of this model with the 2016 and 2020 Institute for Clinical and Economic Review models revealed marked differences, mainly driven by mortality inputs, transition probability estimates, and incorporation of the effect of treatment and comorbidities. CONCLUSION This is the first CEM in NASH to incorporate the clinical consequences of several comorbidities. The flexible yet standardized framework permits estimation of the cost-effectiveness of NASH interventions in a variety of settings. The model currently includes several assumptions and will be further developed as more relevant data become available.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Inger Smith
- White Box Health Economics Ltd, Amelia House, Crescent Road, Worthing, West Sussex, UK
| |
Collapse
|
25
|
Paine‐Cabrera D, Harvey LK, Robarts DR, Pritchard MT, Thyfault J, Weinman SA, Apte U, Slowik V. Leukocyte cell-derived chemotaxin 2 correlates with pediatric non-alcoholic fatty liver disease. Clin Transl Sci 2023; 16:2719-2728. [PMID: 37877453 PMCID: PMC10719457 DOI: 10.1111/cts.13666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 09/07/2023] [Accepted: 10/03/2023] [Indexed: 10/26/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), newly renamed metabolic dysfunction-associated liver disease (MASLD), is a leading cause of liver disease in children and adults. There is a paucity of data surrounding potential biomarkers and therapeutic targets, especially in pediatric NAFLD. Leukocyte cell-derived chemotaxin 2 (LECT2) is a chemokine associated with both liver disease and skeletal muscle insulin resistance. Our aim was to determine associations between LECT2 and common clinical findings of NAFLD in pediatric patients. Enzyme-linked immunosorbent assay (ELISA) was used to measure serum LECT2 concentrations in children (aged 2-17 years) with and without NAFLD. LECT2 concentrations were then correlated to clinical parameters in NAFLD. Mean LECT2 was significantly elevated in children with NAFLD versus healthy controls (n = 63 vs. 42, 5.83 ± 1.98 vs. 4.02 ± 2.02 ng/mL, p < 0.005). Additionally, LECT2 had strong correlations with body mass index (BMI) (Pearson r = 0.301, p = 0.002). A LECT2 concentration of 3.76 mg/mL predicts NAFLD with a sensitivity of 90.5% and specificity of 54.8%. Principal component analysis and logistic regression models further confirmed associations between LECT2 and NAFLD status. This study demonstrates increased serum LECT2 concentrations in pediatric NAFLD, which correlates with BMI and shows strong predictive value within these patients. Our data indicate that LECT2 is a potential diagnostic biomarker of disease and should be further investigated in pediatric as well as adult NAFLD.
Collapse
Affiliation(s)
- Diego Paine‐Cabrera
- Department of Pharmacology, Toxicology and TherapeuticsUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Lisa K. Harvey
- Division of Gastroenterology, Hepatology, and NutritionChildren's Mercy – Kansas CityKansas CityMissouriUSA
| | - Dakota R. Robarts
- Department of Pharmacology, Toxicology and TherapeuticsUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Michele T. Pritchard
- Department of Pharmacology, Toxicology and TherapeuticsUniversity of Kansas Medical CenterKansas CityKansasUSA
- University of Kansas Liver CenterKansas CityKansasUSA
| | - John Thyfault
- Department of Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityKansasUSA
- Children's Center for Healthy Lifestyles and NutritionKansas CityMissouriUSA
| | - Steven A. Weinman
- University of Kansas Liver CenterKansas CityKansasUSA
- Department of Internal MedicineUniversity of Kansas Medical CenterKansas CityKansasUSA
| | - Udayan Apte
- Department of Pharmacology, Toxicology and TherapeuticsUniversity of Kansas Medical CenterKansas CityKansasUSA
- University of Kansas Liver CenterKansas CityKansasUSA
- Children's Center for Healthy Lifestyles and NutritionKansas CityMissouriUSA
| | - Voytek Slowik
- Division of Gastroenterology, Hepatology, and NutritionChildren's Mercy – Kansas CityKansas CityMissouriUSA
- Children's Center for Healthy Lifestyles and NutritionKansas CityMissouriUSA
| |
Collapse
|
26
|
Nadolsky K, Cryer DR, Articolo A, Fisher T, Schneider J, Rinella M. Nonalcoholic steatohepatitis diagnosis and treatment from the perspective of patients and primary care physicians: a cross-sectional survey. Ann Med 2023; 55:2211349. [PMID: 37171239 PMCID: PMC10184582 DOI: 10.1080/07853890.2023.2211349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND The global prevalence of nonalcoholic steatohepatitis (NASH) is rising. Despite this, NASH is underdiagnosed and does not yet have approved pharmacological treatments. We sought to understand the path to diagnosis, patient interactions with healthcare professionals, treatment regimens, and disease management for patients with NASH. METHODS Cross-sectional online surveys of patients with a self-reported diagnosis of NASH and healthcare professionals treating patients with NASH were conducted from 10th November 2020, to 1st January 2021. This manuscript focuses on responses from 152 patients with NASH and 101 primary care physicians (PCPs). RESULTS Patients (n = 152, mean age = 40, SD = 11) and healthcare professionals (n = 226) were located throughout the US. In the most common patient journey, 72% of patients had initial discussions about symptoms with a PCP but only 30% report receiving their NASH diagnosis from a PCP. Almost half of PCPs (47%) were not aware of any clinical practice guidelines for diagnosis and management of NASH. For ongoing management of NASH, PCPs most frequently prescribed lifestyle changes such as exercise (89%), lifestyle changes focused on diet (79%), and/or metformin (57%). Other healthcare professionals rarely referred patients to PCPs for treatment, but when they did, the primary reasons were patients struggling with lifestyle modifications (58%), needing to lose weight (46%), and needing treatment of comorbidities (42%). CONCLUSIONS PCPs may benefit from greater awareness of NASH and guidelines for its diagnosis and treatment. Given the absence of pharmacological treatments approved for NASH, PCPs can offer support in obesity management, comorbidity management, and risk stratification for liver disease progression.
Collapse
Affiliation(s)
- Karl Nadolsky
- MI State University College of Human Medicine, Holland Hospital Endocrinology, Obesity & Diabetes, Holland, MI, USA
| | | | | | | | | | - Mary Rinella
- Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| |
Collapse
|
27
|
Lin YP, Fang QL, Fu SN, Li XP, Shi R, Du CH, Qiao X, Yin XQ, Zeng YC, Zhao XJ, Hua Y. The alleviating effect of Scutellaria amoena extract on the regulation of gut microbiota and its metabolites in NASH rats by inhibiting the NLRP3/ASC/caspase-1 axis. Front Pharmacol 2023; 14:1143785. [PMID: 38026986 PMCID: PMC10660680 DOI: 10.3389/fphar.2023.1143785] [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: 02/01/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Scutellaria amoena (SA) is the root of S. amoena C.H. Wright of Labiatae, also known as Scutellaria southwestern. This is mainly distributed in Sichuan, Yunnan, and Guizhou in China. In southwest China, SA is used as an alternative method to genuine medicine for the treatment of allergy, diarrhea, inflammation, hepatitis, and bronchitis. Thus far, studies on the effects of SA on non-alcoholic steatohepatitis (NASH) are lacking. This paper investigated the effect of SA on the regulation of gut microbiota and its metabolites in NASH rats by inhibiting the NOD-like receptor 3 (NLRP3)/apoptosis-associated speck-like protein (ASC)/caspase-1 axis. Methods: A NASH rat model was induced by a high-fat diet (HFD) for 12 weeks, and rats were orally given different doses of SA extracts (150 and 300 mg/kg/d) for 6 weeks. Changes in histological parameters, body weight, organ indexes, cytokines, and biochemical parameters related to NLRP3 in NASH rats were checked. 16S rRNA gene sequencing and UPLC-MS/MS technology were used to analyze the changes in the gut microbiota composition and its metabolites in NASH rats. Results: SA significantly inhibited the HFD-induced increase in body weight, lipid levels, and inflammatory infiltration. SA notably inhibited the HFD-induced increase in the upper and lower factors of NLRP3, such as transforming growth factor (TGF)-β, tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-18, pro-IL-18, IL-1β, pro-IL-1β, NLRP3, ASC, and caspase-1. Additionally, mRNA expressions of caspase-1, NLRP3, and ASC were significantly downregulated after SA treatment. The results of the intestinal flora showed that SA could increase the diversity of flora and change its structure and composition in NASH rats by reducing Firmicutes/Bacteroidetes (F/B) ratio, Blautia (genus), Lachospiraceae (family), and Christensenellaceae R-7 group (genus), and increasing Muribaculaceae (family) and Bacteroides (genus). The metabolomics revealed that 24 metabolites were possibly the key metabolites for SA to regulate the metabolic balance of NASH rats, including chenodeoxycholic acid, xanthine, and 9-OxoODE. Nine metabolic pathways were identified, including primary bile acid biosynthesis, bile secretion, purine metabolism, and secondary bile acid biosynthesis. Conclusion: SA can regulate the intestinal microbial balance and metabolic disorder by inhibiting the NLRP3/ASC/caspase-1 axis to relieve NASH.
Collapse
Affiliation(s)
- Yu-Ping Lin
- Key Laboratory for Forest Resources Conservation and Use in the Southwest Mountains of China, Southwest Forestry University, Kunming, China
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Qiong-Lian Fang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Sheng-Nan Fu
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Xin-Ping Li
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Rui Shi
- Key Laboratory for Forest Resources Conservation and Use in the Southwest Mountains of China, Southwest Forestry University, Kunming, China
| | - Cheng-Hong Du
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Xue Qiao
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Xun-Qing Yin
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Yong-Cheng Zeng
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiu-Juan Zhao
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China
| | - Yan Hua
- Key Laboratory for Forest Resources Conservation and Use in the Southwest Mountains of China, Southwest Forestry University, Kunming, China
| |
Collapse
|
28
|
Stender S, Davey Smith G, Richardson TG. Genetic variation and elevated liver enzymes during childhood, adolescence and early adulthood. Int J Epidemiol 2023; 52:1341-1349. [PMID: 37105232 PMCID: PMC10555681 DOI: 10.1093/ije/dyad048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Genetic factors influence the risk of fatty liver disease (FLD) in adults. The aim of this study was to test if, and when, genetic risk factors known to affect FLD in adults begin to exert their deleterious effects during childhood, adolescence and early adulthood. METHODS We included up to 4018 British children and adolescents from the Avon Longitudinal Study of Parents and Children (ALSPAC) cohort. Three genetic variants known to associate robustly with FLD in adults (PNPLA3 rs738409, TM6SF2 rs58542926 and HSD17B13 rs72613567) were tested for association with plasma levels of alanine transaminase (ALT) and aspartate transaminase (AST) during childhood (mean age: 9.9 years), early adolescence (15.5 years), late adolescence (17.8 years), and early adulthood (24.5 years). We also tested the associations of a 17-variant score and whole-genome polygenic risk scores (PRS) derived from associations in adults with plasma ALT and AST at the same four time points. Associations with elastography-derived liver steatosis and fibrosis were tested in early adulthood. RESULTS Genetic risk factors for FLD (individually, combined into a 3-variant score, a 17-variant score and as a genome-wide PRS), were associated with higher liver enzymes, beginning in childhood and throughout adolescence and early adulthood. The ALT-increasing effects of the genetic risk variants became larger with increasing age. The ALT-PRS was associated with liver steatosis in early adulthood. No genetic associations with fibrosis were observed. CONCLUSIONS Genetic factors that promote FLD in adults associate with elevated liver enzymes already during childhood, and their effects get amplified with increasing age.
Collapse
Affiliation(s)
- Stefan Stender
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - George Davey Smith
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, University of Bristol, Bristol, UK
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, University of Bristol, Bristol, UK
| |
Collapse
|
29
|
Cao Y, Fang X, Sun M, Zhang Y, Shan M, Lan X, Zhu D, Luo H. Preventive and therapeutic effects of natural products and herbal extracts on nonalcoholic fatty liver disease/nonalcoholic steatohepatitis. Phytother Res 2023; 37:3867-3897. [PMID: 37449926 DOI: 10.1002/ptr.7932] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common condition that is prevalent in patients who consume little or no alcohol, and is characterized by excessive fat accumulation in the liver. The disease is becoming increasingly common with the rapid economic development of countries. Long-term accumulation of excess fat can lead to NAFLD, which represents a global health problem with no effective therapeutic approach. NAFLD is a complex, multifaceted pathological process that has been the subject of extensive research over the past few decades. Herbal medicines have gained attention as potential therapeutic agents to prevent and treat NAFLD due to their high efficacy and low risk of side effects. Our overview is based on a PubMed and Web of Science database search as of Dec 22 with the keywords: NAFLD/NASH Natural products and NAFLD/NASH Herbal extract. In this review, we evaluate the use of herbal medicines in the treatment of NAFLD. These natural resources have the potential to inform innovative drug research and the development of treatments for NAFLD in the future.
Collapse
Affiliation(s)
- Yiming Cao
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Xiaoxue Fang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Mingyang Sun
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Yegang Zhang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Mengyao Shan
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Xintian Lan
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Difu Zhu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Haoming Luo
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| |
Collapse
|
30
|
Yan M, Li H, Xu S, Wu J, Li J, Xiao C, Mo C, Ding BS. Targeting Endothelial Necroptosis Disrupts Profibrotic Endothelial-Hepatic Stellate Cells Crosstalk to Alleviate Liver Fibrosis in Nonalcoholic Steatohepatitis. Int J Mol Sci 2023; 24:11313. [PMID: 37511074 PMCID: PMC10379228 DOI: 10.3390/ijms241411313] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Chronic liver diseases affect over a billion people worldwide and often lead to fibrosis. Nonalcoholic steatohepatitis (NASH), a disease paralleling a worldwide surge in metabolic syndromes, is characterized by liver fibrosis, and its pathogenesis remains largely unknown, with no effective treatment available. Necroptosis has been implicated in liver fibrosis pathogenesis. However, there is a lack of research on necroptosis specific to certain cell types, particularly the vascular system, in the context of liver fibrosis and NASH. Here, we employed a mouse model of NASH in combination with inducible gene knockout mice to investigate the role of endothelial necroptosis in NASH progression. We found that endothelial cell (EC)-specific knockout of mixed lineage kinase domain-like protein (MLKL), a critical executioner involved in the disruption of cell membranes during necroptosis, alleviated liver fibrosis in the mouse NASH model. Mechanistically, EC-specific deletion of Mlkl mitigated the activation of TGFβ/Smad 2/3 pathway, disrupting the pro-fibrotic crosstalk between endothelial cells and hepatic stellate cells (HSCs). Our findings highlight endothelial MLKL as a promising molecular target for developing therapeutic interventions for NASH.
Collapse
Affiliation(s)
- Mengli Yan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Hui Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Shiyu Xu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Jinyan Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Jiachen Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Chengju Xiao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
- Fibrosis Research Program, Division of Pulmonary and Critical Care Medicine, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Regenerative Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
31
|
Ma N, Hou A, Pan X, Sun F, Xu X, Yu C, Lai R, Huang R, Gong L, Xie Q, Chen J, Ren J. MiR-552-3p Regulates Multiple Fibrotic and Inflammatory genes Concurrently in Hepatic Stellate Cells Improving NASH-associated Phenotypes. Int J Biol Sci 2023; 19:3456-3471. [PMID: 37496991 PMCID: PMC10367551 DOI: 10.7150/ijbs.80760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 05/13/2023] [Indexed: 07/28/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a chronic liver disease characterized by hepatic steatosis, inflammation, and progressive fibrosis. Our previous study demonstrated that microRNA-552-3p (miR-552-3p) was down-regulated in the livers of patients with NASH and alleviated hepatic glycolipid metabolic disorders. However, whether miR-552-3p affects NASH progression remains unclear. In this current study, we found that hepatic miR-552-3p expression was negatively correlated with the degree of liver fibrosis and inflammation of NASH patients. Interestingly, the level of miR-552-3p was decreased during hepatic stellate cell (HSC) activation in vitro. Overexpression of miR-552-3p could not only inhibit the expression of fibrotic and inflammatory genes, but also restrain the activation of TGF-β1/Smad3 signaling pathway by down-regulating the expression of TGFBR2 and SMAD3 in HSCs, finally suppressing HSC activation. More importantly, overexpression of miR-552-3p ameliorated liver fibrosis and inflammation in two murine models: high fat/high fructose/high cholesterol diet-induced NASH model and carbon tetrachloride (CCl4)-treated liver fibrosis model. In conclusion, miR-552-3p plays a crucial role in the pathogenesis of NASH by limiting multiple fibrotic and inflammatory pathways in HSCs, which may shed light on its therapeutic potential in NASH.
Collapse
Affiliation(s)
- Ningning Ma
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road Beijing 100049, China
| | - Aijun Hou
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road Beijing 100049, China
| | - Xiangyu Pan
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Fuguang Sun
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road Beijing 100049, China
| | - Xiaoding Xu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Chuwei Yu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road Beijing 100049, China
| | - Rongtao Lai
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Ruimin Huang
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road Beijing 100049, China
| | - Likun Gong
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road Beijing 100049, China
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Jing Chen
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road Beijing 100049, China
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
- School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai 201210, China
- University of Chinese Academy of Sciences, No.19A Yuquan Road Beijing 100049, China
| |
Collapse
|
32
|
Roesch-Dietlen F, Pérez-Morales A, Grube-Pagola P, González-Santes M, Díaz-Roesch F, Triana-Romero A, Roesch-Ramos L, Remes-Troche J, Cruz-Aguilar M. Prevalencia de la esteatosis hepática metabólica (EHMet) en pacientes con litiasis vesicular. Estudio de una cohorte de casos en el sur-sureste de México. REVISTA DE GASTROENTEROLOGÍA DE MÉXICO 2023; 88:225-231. [DOI: 10.1016/j.rgmx.2021.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
33
|
Kim AD, Kui L, Kaufmann B, Kim SE, Leszczynska A, Feldstein AE. Myeloid-specific deletion of chitinase-3-like 1 protein ameliorates murine diet-induced steatohepatitis progression. J Mol Med (Berl) 2023; 101:813-828. [PMID: 37166517 PMCID: PMC10300183 DOI: 10.1007/s00109-023-02325-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/12/2023]
Abstract
Chitinase-3-like 1 protein (CHI3L1) is a secreted glycoprotein, strongly correlated with fibrosis severity in chronic liver diseases including non-alcoholic steatohepatitis (NASH). However, the mechanisms by which CHI3L1 contributes to fibrogenesis remain undefined. Here, we showed that infiltrating monocyte-derived liver macrophages represent the main source of CHI3L1 in murine NASH. We developed a floxed CHI3L1 knock-out (KO) mouse to further study the cell-specific role of CHI3L1 ablation. Wildtype (WT) and myeloid cell-specific CHI3L1 KO mice (CreLyz) were challenged with a highly inflammatory and fibrotic dietary model of NASH by administering choline-deficient high-fat diet for 10 weeks. Macrophage accumulation and inflammatory cell recruitment were significantly ameliorated in the CreLyz group compared to WT (F4/80 IHC p < 0.0001, CD11b IHC p < 0.0001). Additionally, hepatic stellate cell (HSC) activation and fibrosis were strongly decreased in this group (α-SMA IHC p < 0.0001, picrosirius red staining p < 0.0001). In vitro studies were performed stimulating bone marrow derived macrophages, THP-1 (human monocytes) and LX2 (human HSCs) cells with recombinant CHI3L1 to dissect its relationship with fibrosis development. Results showed an important role of CHI3L1 regulating fibrosis-promoting factors by macrophages (TGFB1 p < 0.05, CTGF p < 0.01) while directly activating HSCs (ACTA2 p < 0.01, COL1A1 p < 0.01), involving IL13Rα2 as the potential mediator. Our findings uncovered a novel role of CHI3L1 derived from liver macrophages in NASH progression and identifies this protein as a potential anti-fibrotic therapeutic target. KEY MESSAGES: We showed that CHI3L1 expression is increased in murine CDAA-HFAT diet NASH model, and that infiltrating macrophages are a key source of CHI3L1 production. Myeloid cell-specific CreLyz CHI3L1 knock-out in mice fed with CDAA-HFAT diet improved the NASH phenotype, with significantly reduced accumulation of pro-inflammatory macrophages and neutrophils compared with WT group. DEG and qPCR analysis of genes in CreLyz CHI3L1 knock-out mouse liver showed the mechanistic role of CHI3L1 in cellular chemotaxis. HSC is directly activated by CHI3L1 via receptor IL13Rα2, leading to upregulation of collagen deposition and pro-fibrotic gene, TIMP-1 and TIMP-2 release in whole liver. Direct stimulation of macrophages with CHI3L1 leads to upregulated expression of HSC-activation factors, suggesting its role in modulating macrophage-HSC crosstalk.
Collapse
Affiliation(s)
- Andrea D Kim
- Department of Pediatrics, University of California San Diego, 3020 Children's Way, MC 5030, La Jolla, San Diego, CA, 92103-8450, USA
| | - Lin Kui
- Department of Pediatrics, University of California San Diego, 3020 Children's Way, MC 5030, La Jolla, San Diego, CA, 92103-8450, USA
| | - Benedikt Kaufmann
- Department of Pediatrics, University of California San Diego, 3020 Children's Way, MC 5030, La Jolla, San Diego, CA, 92103-8450, USA
- Department of Surgery, TUM School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Sung Eun Kim
- Department of Pediatrics, University of California San Diego, 3020 Children's Way, MC 5030, La Jolla, San Diego, CA, 92103-8450, USA
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Republic of Korea
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Republic of Korea
| | - Aleksandra Leszczynska
- Department of Pediatrics, University of California San Diego, 3020 Children's Way, MC 5030, La Jolla, San Diego, CA, 92103-8450, USA
| | - Ariel E Feldstein
- Department of Pediatrics, University of California San Diego, 3020 Children's Way, MC 5030, La Jolla, San Diego, CA, 92103-8450, USA.
| |
Collapse
|
34
|
Barreby E, Strunz B, Nock S, Naudet L, Shen JX, Johansson H, Sönnerborg I, Ma J, Urgard E, Pallett LJ, Hu Y, Fardellas A, Azzimato V, Vankova A, Levi L, Morgantini C, Maini MK, Stål P, Rosshart SP, Coquet JM, Nowak G, Näslund E, Lauschke VM, Ellis E, Björkström NK, Chen P, Aouadi M. Human resident liver myeloid cells protect against metabolic stress in obesity. Nat Metab 2023; 5:1188-1203. [PMID: 37414931 PMCID: PMC10365994 DOI: 10.1038/s42255-023-00834-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 06/05/2023] [Indexed: 07/08/2023]
Abstract
Although multiple populations of macrophages have been described in the human liver, their function and turnover in patients with obesity at high risk of developing non-alcoholic fatty liver disease (NAFLD) and cirrhosis are currently unknown. Herein, we identify a specific human population of resident liver myeloid cells that protects against the metabolic impairment associated with obesity. By studying the turnover of liver myeloid cells in individuals undergoing liver transplantation, we find that liver myeloid cell turnover differs between humans and mice. Using single-cell techniques and flow cytometry, we determine that the proportion of the protective resident liver myeloid cells, denoted liver myeloid cells 2 (LM2), decreases during obesity. Functional validation approaches using human 2D and 3D cultures reveal that the presence of LM2 ameliorates the oxidative stress associated with obese conditions. Our study indicates that resident myeloid cells could be a therapeutic target to decrease the oxidative stress associated with NAFLD.
Collapse
Affiliation(s)
- Emelie Barreby
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Benedikt Strunz
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sebastian Nock
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Léa Naudet
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
| | - Helene Johansson
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet (CLINTEC), Huddinge, Sweden
| | - Isabella Sönnerborg
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet (CLINTEC), Huddinge, Sweden
| | - Junjie Ma
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Egon Urgard
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Laura J Pallett
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Yizhou Hu
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Achilleas Fardellas
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Valerio Azzimato
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- BioPharmaceuticals R&D, Clinical Pharmacology and Safety Sciences, Translational Hepatic Safety, AstraZeneca, Gothenburg, Sweden
| | - Ana Vankova
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Laura Levi
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Morgantini
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Cardio Metabolic Unit, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mala K Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Per Stål
- Division of Gastroenterology, Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Stephan P Rosshart
- Department of Microbiome Research, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Department of Medicine II, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Jonathan M Coquet
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Greg Nowak
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet (CLINTEC), Huddinge, Sweden
| | - Erik Näslund
- Division of Surgery, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Ewa Ellis
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet (CLINTEC), Huddinge, Sweden
| | - Niklas K Björkström
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ping Chen
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Myriam Aouadi
- Center for Infectious Medicine (CIM), Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
35
|
Roesch-Dietlen F, Pérez-Morales AG, Grube-Pagola P, González-Santes M, Díaz-Roesch F, Triana-Romero A, Roesch-Ramos L, Remes-Troche JM, Cruz-Aguilar M. Prevalence of metabolic associated fatty liver disease (MAFLD) in patients with gallstone disease. Study on a cohort of cases in South-Southeastern Mexico. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2023; 88:225-231. [PMID: 37258385 DOI: 10.1016/j.rgmxen.2021.09.006] [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: 03/15/2021] [Accepted: 09/07/2021] [Indexed: 06/02/2023]
Abstract
INTRODUCTION Metabolic (dysfunction) associated fatty liver disease (MAFLD) and gallstone disease are entities that share similar risk factors. Numerous publications confirm their elevated frequency, but few studies have considered their prevalence and possible association. AIMS To determine the prevalence of MAFLD in patients with gallstone disease and the usefulness of liver biopsy for diagnosing the liver disease. MATERIALS AND METHODS A prospective study was conducted on patients that underwent laparoscopic cholecystectomy, in whom liver biopsy was performed. VARIABLES ANALYZED Anthropometric characteristics, biochemical profile, conventional ultrasound, risk factors, and histopathologic study of the liver biopsy. STATISTICAL ANALYSIS Descriptive statistics were carried out for the quantitative variables and the Student's t test and multivariate analysis through binary logistic regression were employed for the continuous variables, utilizing IBM-SPSS, 25.0 (Windows) software. RESULTS A total of 136 patients were classified into 2 groups: 40 (29.41%) with normal liver and 96 (70.59%) with MAFLD. Of the 136 patients, 71 patients (52.21%) corresponded to hepatic steatosis, 21 (15.44%) to steatohepatitis, and 4 (2.94%) to cirrhosis. Perisinusoidal inflammation was found in 39 cases (28.68%) and fibrosis was found in 10 (7.35%). The risk factors for both groups were age, diabetes, high blood pressure, and obesity. Glucose, triglyceride, and aminotransferase levels were significantly higher in the MAFLD group and conventional ultrasound demonstrated moderate concordance for its detection. DISCUSSION AND CONCLUSIONS The results confirmed the elevated frequency of MAFLD associated with gallstone disease, justifying liver biopsy during cholecystectomy for diagnosing MAFLD.
Collapse
Affiliation(s)
- F Roesch-Dietlen
- Departamento de Gastroenterología, Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico.
| | - A G Pérez-Morales
- Profesor de la Facultad de Medicina y Vicerrector, Universidad Veracruzana, Región Veracruz-Boca del Río, Mexico
| | - P Grube-Pagola
- Anatomopatólogo, Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico
| | - M González-Santes
- Profesor de la Facultad de Bioanálisis, Universidad Veracruzana, Veracruz, Mexico
| | | | - A Triana-Romero
- Médico en Servicio Social CONACyT, Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico
| | - L Roesch-Ramos
- Profesora y Directora, Facultad de Odontología, Universidad Veracruzana, Mexico
| | - J M Remes-Troche
- Departamento de Neurogastroenterología, Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico
| | - M Cruz-Aguilar
- Profesor de la Facultad de Bioanálisis, Universidad Veracruzana, Veracruz, Mexico
| |
Collapse
|
36
|
Sakamoto S, Nagasaki A, Shrestha M, Shintani T, Watanabe A, Furusho H, Chayama K, Takata T, Miyauchi M. Porphyromonas gingivalis-odontogenic infection is the potential risk for progression of nonalcoholic steatohepatitis-related neoplastic nodule formation. Sci Rep 2023; 13:9350. [PMID: 37291206 PMCID: PMC10250332 DOI: 10.1038/s41598-023-36553-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 06/06/2023] [Indexed: 06/10/2023] Open
Abstract
Porphyromonas gingivalis (P.g.), a major periodontal pathogen is a known risk factor for various systemic diseases. However, the relationship between P.g. and nonalcoholic steatohepatitis (NASH)-related hepatocellular carcinoma (HCC) is unclear. Thus, we aimed to elucidate whether P.g.-odontogenic infection promotes NASH-related HCC development/progression and to clarify its mechanism. Using high-fat diet (HFD)-induced NASH mouse model, P.g. was infected odontogenically. After 60 weeks of infection, tumor profiles were examined. Chow diet (CD) groups were also prepared at 60 weeks. Nodule formation was only seen in HFD-mice. P.g.-odontogenic infection significantly increased the mean nodule area (P = 0.0188) and tended to promote histological progression score after 60 weeks (P = 0.0956). Interestingly, P.g. was detected in the liver. HFD-P.g. (+) showed numerous TNF-α positive hepatic crown-like structures and 8-OHdG expression in the non-neoplastic liver. In P.g.-infected hepatocytes, phosphorylation of integrin β1 signaling molecules (FAK/ERK/AKT) was upregulated in vitro. In fact, total AKT in the liver of HFD-P.g. (+) was higher than that of HFD-P.g. (-). P.g.-infected hepatocytes showed increased cell proliferation and migration, and decreased doxorubicin-mediated apoptosis. Integrin β1 knockdown inhibited these phenotypic changes. P.g.-odontogenic infection may promote the progression of neoplastic nodule formation in an HFD-induced NASH mouse model via integrin signaling and TNF-α induced oxidative DNA damage.
Collapse
Affiliation(s)
- Shinnichi Sakamoto
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, Hiroshima, 734-8551, Japan
- Division of Pathology, Department of Diagnostic and Therapeutic Sciences, Meikai University School of Dentistry, Sakado, Japan
| | - Atsuhiro Nagasaki
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, Hiroshima, 734-8551, Japan
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Madhu Shrestha
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, Hiroshima, 734-8551, Japan
- Department of Diagnostic Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| | - Tomoaki Shintani
- Center of Oral Clinical Examination, Hiroshima University Hospital, Hiroshima, Japan
| | - Atsushi Watanabe
- Laboratory of Research Advancement, National Center for Geriatrics and Gerontology, Research Institute, Obu, Japan
| | - Hisako Furusho
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, Hiroshima, 734-8551, Japan
| | - Kazuaki Chayama
- Collaborative Research Laboratory of Medical Innovation, Hiroshima University, Hiroshima, Japan
- Research Center for Hepatology and Gastroenterology, Hiroshima University, Hiroshima, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takashi Takata
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, Hiroshima, 734-8551, Japan.
- Shunan University, 843-4-2 Gakuendai, Shunan, Japan.
| | - Mutsumi Miyauchi
- Department of Oral and Maxillofacial Pathobiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima, Hiroshima, 734-8551, Japan.
| |
Collapse
|
37
|
Hosseini SP, Farivar S, Rezaei R, Tokhanbigli S, Hatami B, Zali MR, Baghaei K. Fibroblast growth factor 2 reduces endoplasmic reticulum stress and apoptosis in in-vitro Non-Alcoholic Fatty Liver Disease model. Daru 2023; 31:29-37. [PMID: 37156902 PMCID: PMC10238349 DOI: 10.1007/s40199-023-00459-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/08/2023] [Indexed: 05/10/2023] Open
Abstract
PURPOSE Non-Alcoholic fatty liver disease is characterized by the accumulation of excess fat in the liver, chronic inflammation, and cell death, ranging from simple steatosis to fibrosis, and finally leads to cirrhosis and hepatocellular carcinoma. The effect of Fibroblast growth factor 2 on apoptosis and ER stress inhibition has been investigated in many studies. In this study, we aimed to investigate the effect of FGF2 on the NAFLD in-vitro model in the HepG2 cell line. METHODS The in-vitro NAFLD model was first induced on the HepG2 cell line using oleic acid and palmitic acid for 24 h and evaluated by ORO staining and Real-time PCR. The cell line was then treated with various concentrations of fibroblast growth factor 2 for 24 h, total RNA was extracted and cDNA was consequently synthesized. Real-time PCR and flow cytometry was applied to evaluate gene expression and apoptosis rate, respectively. RESULTS It was shown that fibroblast growth factor 2 ameliorated apoptosis in the NAFLD in-vitro model by reducing the expression of genes involved in the intrinsic apoptosis pathway, including caspase 3 and 9. Moreover, endoplasmic reticulum stress was decreased following upregulating the protective ER-stress genes, including SOD1 and PPARα. CONCLUSIONS FGF2 significantly reduced ER stress and intrinsic apoptosis pathway. Our data suggest that FGF2 treatment could be a potential therapeutic strategy for NAFLD.
Collapse
Affiliation(s)
- Seyedeh Parisa Hosseini
- Department of Molecular and Cell Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Shirin Farivar
- Department of Molecular and Cell Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Ramazan Rezaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samaneh Tokhanbigli
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
38
|
Chen X, Shi J, Lai Y, Xue Y, Ung COL, Hu H. Systematic analysis of randomised controlled trials of Chinese herb medicine for non-alcoholic steatohepatitis (NASH): implications for future drug development and trial design. Chin Med 2023; 18:58. [PMID: 37208742 DOI: 10.1186/s13020-023-00761-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 04/28/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a liver disease currently lacking an approved therapy, resulting in significant clinical demand. Traditional Chinese medicines (TCMs) have been commonly used to manage NASH. This study aimed to systematically analyse the randomised controlled trials (RCTs) using TCMs for NASH management. METHODS A systematic literature review was performed by following PRISMA guidelines 2020 in six electronic databases: PubMed, Web of Science, Scopus, Embase, the Cochrane Library, and China National Knowledge Infrastructure, from inception until August 2022. RCTs using TCMs for NASH were included in the analysis, irrespective of language or blinding. RESULTS 112 RCTs were included in this review, with 10,573 NASH participants. 108 RCTs were conducted in China, and 4 RCTs were in other countries. Herbal medicine decoction was the major dosage form used for treating NASH (82/112). 11 TCMs products have been approved for NASH treatment (8 in China, 2 in Iran, and 1 in Japan). Classic prescriptions, such as "Huang Lian Jie Du decoction", "Yin Chen Hao decoction", and "Yi Guan Jian" were used in some studies. The TCMs treatment of NASH involved the use of 199 different plants, with the top 5 herbs being Salviae Miltiorrhizae Radix Et Rhizoma, Alismatis Rhizoma, Bupleuri Radix, Poria, and Curcumae Radix. "Salviae Miltiorrhizae Radix Et Rhizoma + Bupleuri Radix/Alismatis Rhizoma" were the mostly common drug-pair in the herbs network analysis. Nowadays, "Bupleuri Radix/Alismatis Rhizoma + Atractylodis Macrocephalae Rhizoma" are increasingly applied in herbal formulas for NASH. Based on the PICOS principles, the included studies varied in terms of the population, intervention, comparator, outcomes, and study design. However, some studies reported unstandardised results and failed to report diagnostic standards, inclusion or exclusion criteria, or sufficient patient information. CONCLUSION Adopting Chinese classic prescriptions or drug-pair may provide a basis for developing new drugs of NASH management. Further research is needed to refine the clinical trial design and obtain more convincing evidence for using TCMs to treat NASH.
Collapse
Affiliation(s)
- Xianwen Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Junnan Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Yunfeng Lai
- School of Public Health and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan Xue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Carolina Oi Lam Ung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China.
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Taipa, Macao, China.
| | - Hao Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China.
- Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Taipa, Macao, China.
| |
Collapse
|
39
|
Mak LY, Gane E, Schwabe C, Yoon KT, Heo J, Scott R, Lee JH, Lee JI, Kweon YO, Weltman M, Harrison SA, Neuschwander-Tetri BA, Cusi K, Loomba R, Given BD, Christianson DR, Garcia-Medel E, Yi M, Hamilton J, Yuen MF. A phase I/II study of ARO-HSD, an RNA interference therapeutic, for the treatment of non-alcoholic steatohepatitis. J Hepatol 2023; 78:684-692. [PMID: 36513186 DOI: 10.1016/j.jhep.2022.11.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Loss-of-function HSD17β13 mutations protect against the development of chronic liver disease. HSD17β13 inhibition represents a potential approach to treat liver diseases, such as non-alcoholic steatohepatitis (NASH). ARO-HSD is an RNA interference (RNAi) therapeutic designed to selectively reduce expression of HSD17β13 mRNA in hepatocytes. In this study, we evaluated the effects of ARO-HSD in normal healthy volunteers (NHVs) and patients with confirmed or clinically suspected NASH. METHODS The safety, tolerability, and pharmacodynamics of ARO-HSD were evaluated in 32 NHVs and 18 patients with confirmed/clinically suspected NASH. Double-blind NHV cohorts received single escalating doses of ARO-HSD (25, 50, 100, or 200 mg) or placebo subcutaneously on Day 1. Open-label patient cohorts received ARO-HSD (25, 100, or 200 mg) subcutaneously on Days 1 and 29. Liver biopsy was performed pre-dose and on Day 71 to evaluate expression levels of HSD17β13 mRNA and protein. RESULTS ARO-HSD treatment was well tolerated with no treatment-related serious adverse events or drug discontinuations. The most frequently reported treatment-emergent adverse events were mild injection site reactions, which were short in duration. Mean changes in hepatic HSD17β13 mRNA from baseline to Day 71 were: -56.9% (25 mg), -85.5% (100 mg), and -93.4% (200 mg). The mean HSD17β13 mRNA reduction was 78.6% (p <0.0001) across pooled cohorts. Hepatic HSD17β13 protein levels were similarly reduced across doses. In patients, mean changes in alanine aminotransferase from baseline to Day 71 were -7.7% (25 mg), -39.3% (100 mg), and -42.3% (200 mg) (p <0.001 for pooled cohorts). CONCLUSIONS ARO-HSD was well tolerated at doses ≤200 mg. This proof-of-concept study demonstrated that short-term treatment with ARO-HSD reduces hepatic HSD17β13 mRNA and protein expression, which is accompanied by reductions in alanine aminotransferase. CLINICALTRIALS GOV NUMBER NCT04202354. IMPACTS AND IMPLICATIONS There is an unmet medical need for new therapies to treat alcohol-related and non-alcoholic liver disease. ARO-HSD is a small-interfering RNA designed to silence HSD17β13 expression and hence to phenocopy the protective effect seen in individuals with HSD17β13 loss-of-function. The reductions in HSD17β13 expression and in transaminases seen with ARO-HSD administration represent an initial step towards clinical validation of HSD17β13, a drug target with substantial genetic validation, as an important modulator of human liver disease.
Collapse
Affiliation(s)
- Lung-Yi Mak
- Department of Medicine, School of Clinical Medicine, Hong Kong; State Key Laboratory of Liver Research, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Ed Gane
- University of Auckland, Auckland, New Zealand
| | | | - Ki Tae Yoon
- Department of Internal Medicine, Pusan National University College of Medicine and Liver Center, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Jeong Heo
- Department of Internal Medicine, College of Medicine, Pusan National University and Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Russell Scott
- Lipid and Diabetes Research, New Zealand Clinical Research, Christchurch, New Zealand
| | - Jeong-Hoon Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung Il Lee
- Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | - Kenneth Cusi
- University of Florida, Gainesville, FL, United States
| | - Rohit Loomba
- NAFLD Research Center, UCSD, Division of Gastroenterology, La Jolla, CA, United States
| | - Bruce D Given
- Arrowhead Pharmaceuticals, Inc., Pasadena, CA, United States
| | | | | | - Min Yi
- Arrowhead Pharmaceuticals, Inc., Pasadena, CA, United States
| | - James Hamilton
- Arrowhead Pharmaceuticals, Inc., Pasadena, CA, United States
| | - Man-Fung Yuen
- Department of Medicine, School of Clinical Medicine, Hong Kong; State Key Laboratory of Liver Research, Queen Mary Hospital, The University of Hong Kong, Hong Kong.
| |
Collapse
|
40
|
Swain MG, Pettersson B, Meyers O, Venerus M, Oscarsson J. A qualitative patient interview study to understand the experience of patients with nonalcoholic steatohepatitis. Hepatol Commun 2023; 7:e0036. [PMID: 36757391 PMCID: PMC9915959 DOI: 10.1097/hc9.0000000000000036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/26/2022] [Indexed: 02/10/2023] Open
Abstract
NASH is a potentially progressive form of NAFLD characterized by hepatocyte injury and liver inflammation which can cause fibrosis. Currently, there are limited data on the patient experience of NASH. Our aim was to use both literature review and patient interviews to understand the signs/symptoms and life impacts of NASH fibrosis stages F1-F4 that are important to patients, as well as begin to investigate the applicability of an instrument (ie, questionnaire) that may be used to capture patients' experiences. The literature review identified concepts (signs/symptoms and impacts) related to NASH fibrosis stages F1-F4 and the NASH-specific patient-reported outcome instrument (NASH-CHECK) for reporting patient experience of NASH. Interviews with 22 patients from Canada and the USA with NASH fibrosis stages F1-F4 revealed 27 signs/symptoms and 32 impacts that they felt were important, including fatigue, pain in the abdomen, worry, and frustration. Three concepts reported during patient interviews were not identified in the literature review. No concepts appeared to be exclusive to a specific fibrosis stage or presence/absence of obesity and no linear trends were identified between fibrosis stage or presence/absence of obesity and level of disturbance reported for concepts. The patient interviews supported the concepts included in the NASH-CHECK overall, demonstrating that it could be used to report the patient experience of NASH fibrosis stages F1-F4. Interviews with patients with NASH fibrosis stages F1-F4 revealed patients can self-report and elaborate on signs/symptoms and impacts related to the disease regardless of fibrosis stage. The NASH-CHECK was identified as a suitable instrument that could be used by patients with fibrosis stages F1-F4.
Collapse
Affiliation(s)
- Mark G. Swain
- Liver Unit, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Billie Pettersson
- Patient Centered Science, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals Medical, AstraZeneca, Gothenburg, Sweden
| | - Oren Meyers
- Patient Centered Endpoints, IQVIA, New York, New York, USA
| | | | - Jan Oscarsson
- Late-stage Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
41
|
Zijlstra MK, Gampa A, Joseph N, Sonnenberg A, Fimmel CJ. Progressive changes in platelet counts and Fib-4 scores precede the diagnosis of advanced fibrosis in NASH patients. World J Hepatol 2023; 15:225-236. [PMID: 36926233 PMCID: PMC10011908 DOI: 10.4254/wjh.v15.i2.225] [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: 06/27/2022] [Revised: 08/02/2022] [Accepted: 01/13/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Cirrhosis and its complications develop in a subgroup of patients with non-alcoholic fatty liver disease (NASH). Early detection of liver fibrosis represents an important goal of clinical care.
AIM To test the hypothesis that the development of cirrhosis in nonalcoholic fatty liver disease patients is preceded by the long-term trends of platelet counts and Fib-4 scores.
METHODS We identified all patients in our healthcare system who had undergone fibrosis staging by liver biopsy or magnetic resonance elastography (MRE) for non-alcoholic fatty liver disease during the past decade (n = 310). Platelet counts, serum glutamic-pyruvic transaminase and serum glutamic oxalacetic transaminase values preceding the staging tests were extracted from the electronic medical record system, and Fib-4 scores were calculated. Potential predictors of advanced fibrosis were evaluated using multivariate regression analysis.
RESULTS Significant decreases in platelet counts and increases in Fib-4 scores were observed in all fibrosis stages, particularly in patients with cirrhosis. In the liver biopsy group, the presence of cirrhosis was best predicted by the combination of the Fib-4 score at the time closest to staging (P < 0.0001), the presence of diabetes (P = 0.0001), and the correlation coefficient of the preceding time-dependent drop in platelet count (P = 0.044). In the MRE group, Fib4 score (P = 0.0025) and platelet drop (P = 0.0373) were significant predictors. In comparison, the time-dependent rise of the Fib-4 score did not contribute in a statistically significant way.
CONCLUSION Time-dependent changes in platelet counts and Fib-4 scores contribute to the prediction of cirrhosis in NASH patients with biopsy- or MRE-staged fibrosis. Their incorporation into predictive algorithms may assist in the earlier identification of high-risk patients.
Collapse
Affiliation(s)
- Michael K Zijlstra
- Department of Internal Medicine, NorthShore University Health System, Evanston, IL 60201, United States
| | - Anuhya Gampa
- Division of Gastroenterology and Hepatology, University of Chicago Pritzker School of Medicine, Chicago, IL 60637, United States
| | - Nora Joseph
- Department of Pathology, NorthShore University Health System, Evanston, IL 60201, United States
| | - Amnon Sonnenberg
- Portland VA Medical Center, Portland, OR 97239, United States
- Department of Gastroenterology, Oregon Health Sciences University, Portland, OR 97201, United States
| | - Claus J Fimmel
- Division of Gastroenterology, Department of Internal Medicine, NorthShore University Health System, Evanston, IL 60201, United States
| |
Collapse
|
42
|
Rajendran L, Murillo Perez CF, Ivanics T, Claasen MPAW, Hansen BE, Wallace D, Yoon PD, Sapisochin G. Outcomes of liver transplantation in non-alcoholic steatohepatitis (NASH) versus non-NASH associated hepatocellular carcinoma. HPB (Oxford) 2023; 25:556-567. [PMID: 36828740 DOI: 10.1016/j.hpb.2023.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/26/2023] [Accepted: 01/30/2023] [Indexed: 02/26/2023]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH)-associated hepatocellular carcinoma (HCC) is a rising indication for liver transplantation. This unique population, with multiple comorbidities, has potential for worse post-transplant outcomes. We compared post-transplant survival of NASH and non-NASH HCC patients using a large cohort. METHODS Adults transplanted for HCC between 2008 and 2018, from United Network for Organ Sharing (UNOS) and University Health Network (UHN) databases were divided into two populations: NASH and non-NASH. Recipient characteristics and post-transplant survival were compared. Subgroup analyses were performed within and beyond Milan criteria. RESULTS 2071 of 20,672 (10.0%) patients underwent transplantation for NASH HCC, with annual proportional increase of 1.2%UHN (p = 0.02) and 1.3%UNOS (p < 0.001). The 1-,3-,5-year post-transplant survival were 90.8%, 83.9%, 76.3% NASH HCC versus 91.9%, 82.1%, 74.9% non-NASH HCC (p = 0.94). No survival differences were observed in populations within or beyond Milan. Competing-risk analysis demonstrated no differences in risk for cardiovascular-related death (HR1.24, 95%CI 0.87-1.55, p = 0.16), or HCC recurrence-related death (HR1.21, 95%CI 0.89-1.65, p = 0.23). NASH HCC patients had lower risk of liver-related deaths (HR0.57, 95%CI 0.34-0.98, p = 0.04). DISCUSSION NASH HCC is a rising indication for liver transplantation. Despite demographic differences, no post-transplantation survival differences were observed between NASH and non-NASH HCC. This justifies equivalent organ allocation, irrespective of NASH status.
Collapse
Affiliation(s)
- Luckshi Rajendran
- Department of Surgery, Division of General Surgery, University of Toronto, Toronto, Canada. https://twitter.com/luckseee
| | - Carla F Murillo Perez
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada; Toronto Centre for Liver Disease, University Health Network, Toronto, Canada; Institute of Health Policy, Management & Evaluation, University of Toronto, Toronto, Canada
| | - Tommy Ivanics
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada; Department of Surgery, Henry Ford Hospital, Detroit, MI, USA; Department of Surgical Sciences, Akademiska Sjukhuset, Uppsala University, Uppsala, Sweden. https://twitter.com/ivanics_t
| | - Marco P A W Claasen
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada; Department of Surgery, Division of HPB & Transplant Surgery, Erasmus MC Transplant Institute, University Medical Centre Rotterdam, Rotterdam, the Netherlands. https://twitter.com/claasen_m
| | - Bettina E Hansen
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada; Toronto Centre for Liver Disease, University Health Network, Toronto, Canada; Institute of Health Policy, Management & Evaluation, University of Toronto, Toronto, Canada
| | - David Wallace
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, United Kingdom; Institute of Liver Studies, Kings College Hospital, Denmark Hill, London, United Kingdom
| | - Peter D Yoon
- Multi-Organ Transplant Program, University Health Network, Toronto, Canada; Department of Surgery, Westmead Hospital, Sydney, Australia
| | - Gonzalo Sapisochin
- Department of Surgery, Division of General Surgery, University of Toronto, Toronto, Canada; Multi-Organ Transplant Program, University Health Network, Toronto, Canada.
| |
Collapse
|
43
|
Zhi G, Shao B, Zheng T, Mu J, Li J, Feng Y, Zhu S, Dang Y, Liu F, Wang D. Exploring the molecular mechanism of Gan Shuang granules for the treatment of non-alcoholic steatohepatitis using network pharmacology, molecular docking, and experimental verification. Front Pharmacol 2023; 14:1082451. [PMID: 36762105 PMCID: PMC9902723 DOI: 10.3389/fphar.2023.1082451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
Background: With the gradual increase in prevalence in recent years, non-alcoholic steatohepatitis (NASH) has become one of the significant health problems that urgently needs to be addressed worldwide. GanShuang Granules (GSG) is derived from the classical Chinese formula Xiaoyao San and mainly used in the clinical treatment of chronic liver diseases. Objective: In this study, we aim to gain a deeper insight into the inhibiting effects of GSG on non-alcoholic fatty liver disease (NAFLD) rats and preliminarily elucidate the underlying intervention mechanisms. Methods: First, High performance liquid chromatography (UHPLC-Q/Orbitrap-MS/MS) was used for the active compounds prediction in GSG. Then the data was mapped to mzCloud database. The targets corresponding to GSG compounds were collected from public databases, along with disease genes for NAFLD. The core targets and molecular mechanisms of GSG for NAFLD treatment were predicted by protein-protein interaction (PPI) network, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses. Molecular docking of the core target-component interactions was simulated using AutoDock Vina software. The effect of GSG on NASH rats was evaluated by pathological staining and analysis of various index results. Finally, the candidate targets were further validated by ELISA and western blot (WB) analyses. Results: Combining UHPLC-Q/Orbitrap-MS/MS data analysis and public database data, a total of 346 cross-targets were obtained, corresponding to 81 compounds. The subnetwork with an MCODE score of 53.623 is a potential core target group for this study. GO and KEGG enrichment analyses showed that the targets of GSG in NAFLD were mostly related to oxidative stress, the NF-κB signaling pathway, and the apoptosis signaling pathway. By integrating the results of network pharmacology analysis, the core objectives of this study mainly include AKT1, CASP9, TNF, and CASP8. The core ingredients are related to resveratrol and fisetin. The molecular docking results indicated key binding activity between AKT1-fisetin, AKT1-Resveratrol, and CASP8-fisetin. Moreover, GSG could improve the inflammatory status and restore the abnormal lipid accumulation of NAFLD/NASH liver, and these levels are further verified by pathological staining and detection of related indicators. Mechanistically, GSG could regulate protein expression levels in the liver for P65, p-P65, IKB, p-IKB, IKK, caspase-3, -8, -9, and cytochrome C, etc. It reflects the inhibitory effect of GSG on the NF-κB/IκB signaling pathway. Conclusion: Our results suggested that GSG demonstrated therapeutic effects on NAFLD/NASH rats, and these may be mainly reflected in the inhibitory effects on the NF-κB/IκB signaling pathway and its downstream inflammation and apoptosis signals.
Collapse
Affiliation(s)
- Guoguo Zhi
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Bingjie Shao
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Tianyan Zheng
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jie Mu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jingwei Li
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yiyuan Feng
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Sha Zhu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yanni Dang
- Shanxi Buchang Pharmaceutical Company Limited, Xi’an, Shanxi, China
| | - Feng Liu
- Shanxi Buchang Pharmaceutical Company Limited, Xi’an, Shanxi, China,*Correspondence: Feng Liu, ; Dong Wang,
| | - Dong Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China,*Correspondence: Feng Liu, ; Dong Wang,
| |
Collapse
|
44
|
Akl MG, Widenmaier SB. Immunometabolic factors contributing to obesity-linked hepatocellular carcinoma. Front Cell Dev Biol 2023; 10:1089124. [PMID: 36712976 PMCID: PMC9877434 DOI: 10.3389/fcell.2022.1089124] [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: 11/03/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major public health concern that is promoted by obesity and associated liver complications. Onset and progression of HCC in obesity is a multifactorial process involving complex interactions between the metabolic and immune system, in which chronic liver damage resulting from metabolic and inflammatory insults trigger carcinogenesis-promoting gene mutations and tumor metabolism. Moreover, cell growth and proliferation of the cancerous cell, after initiation, requires interactions between various immunological and metabolic pathways that provide stress defense of the cancer cell as well as strategic cell death escape mechanisms. The heterogenic nature of HCC in addition to the various metabolic risk factors underlying HCC development have led researchers to focus on examining metabolic pathways that may contribute to HCC development. In obesity-linked HCC, oncogene-induced modifications and metabolic pathways have been identified to support anabolic demands of the growing HCC cells and combat the concomitant cell stress, coinciding with altered utilization of signaling pathways and metabolic fuels involved in glucose metabolism, macromolecule synthesis, stress defense, and redox homeostasis. In this review, we discuss metabolic insults that can underlie the transition from steatosis to steatohepatitis and from steatohepatitis to HCC as well as aberrantly regulated immunometabolic pathways that enable cancer cells to survive and proliferate in the tumor microenvironment. We also discuss therapeutic modalities targeted at HCC prevention and regression. A full understanding of HCC-associated immunometabolic changes in obesity may contribute to clinical treatments that effectively target cancer metabolism.
Collapse
Affiliation(s)
- May G. Akl
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Physiology, University of Alexandria, Alexandria, Egypt
| | - Scott B. Widenmaier
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
45
|
(Pro)Renin Receptor Antagonism Attenuates High-Fat-Diet-Induced Hepatic Steatosis. Biomolecules 2023; 13:biom13010142. [PMID: 36671527 PMCID: PMC9855393 DOI: 10.3390/biom13010142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/20/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) comprises a spectrum of liver damage directly related to diabetes, obesity, and metabolic syndrome. The (pro)renin receptor (PRR) has recently been demonstrated to play a role in glucose and lipid metabolism. Here, we test the hypothesis that the PRR regulates the development of diet-induced hepatic steatosis and fibrosis. C57Bl/6J mice were fed a high-fat diet (HFD) or normal-fat diet (NFD) with matching calories for 6 weeks. An 8-week methionine choline-deficient (MCD) diet was used to induce fibrosis. Two weeks following diet treatment, mice were implanted with a subcutaneous osmotic pump delivering either the peptide PRR antagonist, PRO20, or scrambled peptide for 4 or 6 weeks. Mice fed a 6-week HFD exhibited increased liver lipid accumulation and liver triglyceride content compared with NFD-fed mice. Importantly, PRO20 treatment reduced hepatic lipid accumulation in HFD-fed mice without affecting body weight or blood glucose. Furthermore, PRR antagonism attenuated HFD-induced steatosis, particularly microvesicular steatosis. In the MCD diet model, the percentage of collagen area was reduced in PRO20-treated compared with control mice. PRO20 treatment also significantly decreased levels of liver alanine aminotransferase, an indicator of liver damage, in MCD-fed mice compared with controls. Mechanistically, we found that PRR antagonism prevented HFD-induced increases in PPARγ and glycerol-3-phosphate acyltransferase 3 expression in the liver. Taken together, our findings establish the involvement of the PRR in liver triglyceride synthesis and suggest the therapeutic potential of PRR antagonism for the treatment of liver steatosis and fibrosis in NAFLD.
Collapse
|
46
|
Wang W, Yang L, Hu M, Yang Y, Ma Q, Chen J. Network Pharmacology to Reveal the Molecular Mechanisms of Rutaceous Plant-derived Limonin Ameliorating Non-alcoholic Steatohepatitis. Crit Rev Immunol 2023; 43:11-23. [PMID: 37831520 DOI: 10.1615/critrevimmunol.2023050080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
BACKGROUND Limonin shows promise in alleviating non-alcoholic fatty liver disease. We investigated the mechanisms of limonin against non-alcoholic steatohepatitis (NASH) using network pharmacology and molecular docking. METHODS Public databases provided NASH- and limonin-associated targets. VennDiagram identified potential limonin targets for NASH. Enrichment analysis explored the limonin-NASH relationship. PPI network analysis, CytoHubba models, and bioinformatics identified hub genes for NASH treatment. Molecular docking assessed limonin's binding ability to hub targets. RESULTS We found 37 potential limonin targets in NASH, involved in oxidative stress, inflammation, and signaling pathways. PPI network analysis revealed seven hub genes (STAT3, NFKBIA, MTOR, TLR4, CASP8, PTGS2, NFKB1) as NASH treatment targets. Molecular docking confirmed limonin's binding to STAT3, CASP8, and PTGS2. Animal experiments on high-fat diet mice showed limonin reduced hepatic steatosis, lipid accumulation, and expression of p-STAT3/STAT3, CASP8, and PTGS2. CONCLUSION Limonin's therapeutic effects in NASH may stem from its antioxidant and anti-inflammatory properties. STAT3, CASP8, and PTGS2 are potential key targets for NASH treatment, warranting further investigation.
Collapse
Affiliation(s)
- Wei Wang
- Department of Gastroenterology, 940th Hospital of Joint Support Force, Lanzhou, Gansu, China
| | - Li Yang
- Northwest Minzu University, Lanzhou, Gansu, China
| | - Minjie Hu
- Department of Gastroenterology, 940th Hospital of Joint Support Force, Lanzhou, Gansu, China
| | - Yonglin Yang
- Department of Gastroenterology, 940th Hospital of Joint Support Force, Lanzhou, Gansu, China
| | - Qiang Ma
- Department of Gastroenterology, 940th Hospital of Joint Support Force, Lanzhou, Gansu, China
| | - Jiayu Chen
- Department of Gastroenterology, 940th Hospital of Joint Support Force, Lanzhou, Gansu, China
| |
Collapse
|
47
|
Anstee QM, Hallsworth K, Lynch N, Hauvespre A, Mansour E, Kozma S, Bottomley J, Milligan G, Piercy J, Higgins V. Alignment of Physician-Stated vs Clinically Derived Reference Fibrosis Score in Patients with Non-Alcoholic Steatohepatitis: A Real-World European Survey. Pragmat Obs Res 2023; 14:13-27. [PMID: 36873793 PMCID: PMC9974948 DOI: 10.2147/por.s392320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Objective Stratifying disease severity in patients with non-alcoholic steatohepatitis (NASH) is essential for appropriate treatment and long-term management. Liver biopsy is the reference standard for fibrosis severity in NASH, but less invasive methods are used, eg, Fibrosis-4 Index (FIB-4) and vibration-controlled transient elastography (VCTE), for which reference thresholds for no/early fibrosis and advanced fibrosis are available. We compared subjective physician assessment of NASH fibrosis versus reference thresholds to understand classification in a real-world setting. Methods Data were drawn from Adelphi Real World NASH Disease Specific ProgrammeTM conducted in France, Germany, Italy, Spain and UK in 2018. Physicians (diabetologists, gastroenterologists, hepatologists) completed questionnaires for five consecutive NASH patients presenting for routine care. Physician-stated fibrosis score (PSFS) based on available information was compared with clinically defined reference fibrosis stage (CRFS) determined retrospectively using VCTE and FIB-4 data and eight reference thresholds. Results One thousand two hundred and eleven patients had VCTE (n = 1115) and/or FIB-4 (n = 524). Depending on thresholds, physicians underestimated severity in 16-33% (FIB-4) and 27-50% of patients (VCTE). Using VCTE ≥12.2, diabetologists, gastroenterologists and hepatologists underestimated disease severity in 35%, 32%, and 27% of patients, respectively, and overestimated fibrosis in 3%, 4%, and 9%, respectively (p = 0.0083 across specialties). Hepatologists and gastroenterologists had higher liver biopsy rates than diabetologists (52%, 56%, 47%, respectively). Conclusion PSFS did not consistently align with CRFS in this NASH real-world setting. Underestimation was more common than overestimation, potentially leading to undertreatment of patients with advanced fibrosis. More guidance on interpreting test results when classifying fibrosis is needed, thereby improving management of NASH.
Collapse
Affiliation(s)
- Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle Upon Tyne, UK.,Liver Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Kate Hallsworth
- Translational & Clinical Research Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle Upon Tyne, UK.,Liver Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Niall Lynch
- Global Value and Access, Gilead Sciences, Hayes, Uxbridge, UK
| | - Adrien Hauvespre
- Market Access and Reimbursement, Gilead Sciences, Hayes, Uxbridge, UK
| | - Eid Mansour
- Pricing and Market Access (Middle East), Gilead Sciences, Dubai, United Arab Emirates
| | - Sam Kozma
- Pricing and Market Access (Middle East), Gilead Sciences, Dubai, United Arab Emirates
| | | | - Gary Milligan
- Statistics Department, Adelphi Real World, Bollington, UK
| | - James Piercy
- Scientific Franchise, Adelphi Real World, Bollington, UK
| | - Victoria Higgins
- Scientific and NASH Franchise, Adelphi Real World, Bollington, UK
| |
Collapse
|
48
|
Chen J, Liu B, Xie X, Li W. Comparative molecular analysis of oral submucous fibrosis and other organ fibrosis based on weighted gene co-expression network analysis. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:1663-1672. [PMID: 36748376 PMCID: PMC10930262 DOI: 10.11817/j.issn.1672-7347.2022.220452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Indexed: 02/08/2023]
Abstract
OBJECTIVES There is currently a lack of economic and suitable animal models that can accurately recapitulate the oral submucous fibrosis (OSF) disease state for indepth study. This is one of the primary reasons for the limited therapeutic methods available for OSF. Based on the underlying logic of pan-cancer analysis, this study systematically compares OSF and the other four types of organ fibrosis from the aspects of molecules, signaling pathways, biological processes, etc. A comprehensive analysis of the similarities and differences between OSF and other organ fibrosis is helpful for researchers to discover some general rules of fibrosis disease and may provide new ideas for studying OSF. METHODS Microarray data of the GSE64216, GSE76882, GSE171294, GSE92592, and GSE90051 datasets were downloaded from GEO. Differentially expressed mRNAs (DEmRNAs) of each type of fibrosis were identified by Limma package. Weighted gene co-expression network analysis (WGCNA) was used to identify each type of fibrosis-related module. The similarities and differences of each fibrosis-related-module genes were analyzed by function and pathway enrichment analysis. RESULTS A total of 6 057, 10 910, 27 990, 10 480, and 4 801 DEmRNAs were identified in OSF, kidney intestinal fibrosis (KIF), liver fibrosis (LF), idiopathic pulmonary fibrosis (IPF), and skin fibrosis (SF), respectively. By using WGCNA, each type of fibrosis-related module was identified. The co-expression networks for each type of fibrosis were constructed respectively. Except that KIF and LF have 5 common hub genes, other fibrotic diseases have no common hub genes with each other. The common pathways of OSF, KIF, LF, IPF, and SF mainly focus on immune-related pathways. CONCLUSIONS OSF and the other 4 types of fibrotic diseases are tissue- and organ-specific at the molecular level, but they share many common signaling pathways and biological processes, mainly in inflammation and immunity.
Collapse
Affiliation(s)
- Jun Chen
- Hunan Key Laboratory of Oral Health Research; Hunan 3D Printing Engineering Research Center of Oral Care; Hunan Clinical Research Center of Oral Major Diseases and Oral Health; Xiangya School of Stomatology, Central South University, Changsha 410008.
- Department of Periodontics and Oral Medicine, Xiangya Stomatological Hospital, Central South University, Changsha 410008.
| | - Binjie Liu
- Hunan Key Laboratory of Oral Health Research; Hunan 3D Printing Engineering Research Center of Oral Care; Hunan Clinical Research Center of Oral Major Diseases and Oral Health; Xiangya School of Stomatology, Central South University, Changsha 410008
- Department of Periodontics and Oral Medicine, Xiangya Stomatological Hospital, Central South University, Changsha 410008
| | - Xiaoli Xie
- Hunan Key Laboratory of Oral Health Research; Hunan 3D Printing Engineering Research Center of Oral Care; Hunan Clinical Research Center of Oral Major Diseases and Oral Health; Xiangya School of Stomatology, Central South University, Changsha 410008
| | - Wenjie Li
- Hunan Key Laboratory of Oral Health Research; Hunan 3D Printing Engineering Research Center of Oral Care; Hunan Clinical Research Center of Oral Major Diseases and Oral Health; Xiangya School of Stomatology, Central South University, Changsha 410008.
- Department of Orthodontics, Xiangya Stomatological Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
49
|
Da BL, He AR, Shetty K, Suchman KI, Yu H, Lau L, Wong LL, Rabiee A, Amdur RL, Crawford JM, Fox SS, Grimaldi GM, Shah PK, Weinstein J, Bernstein D, Satapathy SK, Chambwe N, Xiang X, Mishra L. Pathogenesis to management of hepatocellular carcinoma. Genes Cancer 2022; 13:72-87. [DOI: 10.18632/genesandcancer.226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022] Open
Affiliation(s)
- Ben L. Da
- Department of Internal Medicine, Division of Hepatology, Sandra Atlas Bass Center for Liver Diseases and Transplantation, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Manhasset, NY 11030, USA
| | - Aiwu Ruth He
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington DC 20007, USA
| | - Kirti Shetty
- Division of Gastroenterology and Hepatology, University of Maryland, Baltimore, MD 21201, USA
| | - Kelly I. Suchman
- Department of Internal Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Manhasset, NY 11030, USA
| | - Herbert Yu
- Department of Epidemiology, University of Hawaii Cancer Center, Honolulu, HI 96813-5516, USA
| | - Lawrence Lau
- Department of Surgery, North Shore University Hospital, Northwell Health, Manhasset, NY 11030, USA
| | - Linda L. Wong
- Department of Surgery, University of Hawaii, Honolulu, HI 96813-5516, USA
| | - Atoosa Rabiee
- Department of Gastroenterology and Hepatology, VA Medical Center, Washington DC 20422, USA
| | - Richard L. Amdur
- Quantitative Intelligence, The Institutes for Health Systems Science and Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Northwell Health, NY 10022, USA
| | - James M. Crawford
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Sharon S. Fox
- Department of Pathology and Laboratory Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Gregory M. Grimaldi
- Department of Radiology, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - Priya K. Shah
- Department of Radiology, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - Jonathan Weinstein
- Division of Vascular and Interventional Radiology, Department of Radiology, Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY 11030, USA
| | - David Bernstein
- Department of Internal Medicine, Division of Hepatology, Sandra Atlas Bass Center for Liver Diseases and Transplantation, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Manhasset, NY 11030, USA
| | - Sanjaya K. Satapathy
- Department of Internal Medicine, Division of Hepatology, Sandra Atlas Bass Center for Liver Diseases and Transplantation, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell Health, Manhasset, NY 11030, USA
| | - Nyasha Chambwe
- The Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, NY 11030, USA
| | - Xiyan Xiang
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY 11030, USA
| | - Lopa Mishra
- The Institute for Bioelectronic Medicine, The Feinstein Institutes for Medical Research and Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY 11030, USA
| |
Collapse
|
50
|
Lu K, Fan Q, Zou X. Antisense oligonucleotide is a promising intervention for liver diseases. Front Pharmacol 2022; 13:1061842. [PMID: 36569303 PMCID: PMC9780395 DOI: 10.3389/fphar.2022.1061842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
As the body's critical metabolic organ, the liver plays an essential role in maintaining proper body homeostasis. However, as people's living standards have improved and the number of unhealthy lifestyles has increased, the liver has become overburdened. These have made liver disease one of the leading causes of death worldwide. Under the influence of adverse factors, liver disease progresses from simple steatosis to hepatitis, to liver fibrosis, and finally to cirrhosis and cancer, followed by increased mortality. Until now, there has been a lack of accepted effective treatments for liver disease. Based on current research, antisense oligonucleotide (ASO), as an alternative intervention for liver diseases, is expected to be an effective treatment due to its high efficiency, low toxicity, low dosage, strong specificity, and additional positive characteristics. In this review, we will first introduce the design, modification, delivery, and the mechanisms of ASO, and then summarize the application of ASO in liver disease treatment, including in non-alcoholic fatty liver disease (NAFLD), hepatitis, liver fibrosis, and liver cancer. Finally, we discuss challenges and perspectives on the transfer of ASO drugs into clinical use. This review provides a current and comprehensive understanding of the integrative and systematic functions of ASO for its use in liver disease.
Collapse
Affiliation(s)
- Kailing Lu
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qijing Fan
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Xiaoju Zou
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming, Yunnan, China,*Correspondence: Xiaoju Zou,
| |
Collapse
|