1
|
Wang CH, Ji XY, Ji N, Yan QF, Xu HQ, Wang XQ, Chen XF, Lu CF. Association between glucose time-in-range and the severity of metabolic dysfunction-associated steatotic liver disease in Chinese adults with type 2 diabetes mellitus. BMC Endocr Disord 2025; 25:113. [PMID: 40269889 PMCID: PMC12016164 DOI: 10.1186/s12902-025-01937-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/15/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND This study investigated the correlation between glucose time-in-range (TIR) and hepatic steatosis severity or liver fibrosis risk in Chinese adults with type 2 diabetes mellitus (T2DM) comorbid with metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS Participants with T2DM were evaluated for hepatic steatosis and fibrosis using vibration-controlled transient elastography. TIR was calculated based on data from a retrospective continuous glucose monitoring system. RESULTS A total of 184 T2DM patients with MASLD were enrolled. The controlled attenuation parameter (CAP) and liver stiffness measurement (LSM) decreased with increasing TIR (p < 0.05). Spearman correlation showed negative correlations between CAP, LSM, and TIR (r = -0.824 and - 0.842, p < 0.05) and positive correlations with basal insulin resistance (HOMA-IR) (r = 0.205 and 0.208, p < 0.01). Multiple linear regression revealed TIR and HOMA-IR independently correlated with CAP (std. regression coefficients = -0.695 and 0.103, p < 0.05) and LSM (std. regression coefficients = -0.735 and 0.083, p < 0.05), wit0.34 h TIR having a stronger impact. Binary logistic regression showed TIR Groups 3 (70% ≥ TIR < 85%) and 4 (TIR ≥ 85%) were protective for MASLD (OR = 0.26 and 0.11, 95% CI 0.10-0.66 and 0.04-0.29, P = 0.005 and < 0.001) and liver fibrosis (OR = 0.29 and 0.13, 95% CI 0.12-0.74 and 0.05-0.36, P = 0.010 and < 0.001) compared to TIR Group 1 (lowest quartile). CONCLUSION In T2DM patients with coexisting MASLD, a significant and independent association existed between TIR and the severity of hepatic steatosis. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Chun-Hua Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, No. 666 Shengli Road, Nantong, 226001, China
| | - Xuan-Yao Ji
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, No. 666 Shengli Road, Nantong, 226001, China
| | - Nian Ji
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, No. 666 Shengli Road, Nantong, 226001, China
| | - Qing-Feng Yan
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, No. 666 Shengli Road, Nantong, 226001, China
| | - Hong-Qing Xu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, No. 666 Shengli Road, Nantong, 226001, China
| | - Xue-Qin Wang
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, No. 666 Shengli Road, Nantong, 226001, China
| | - Xiang-Fan Chen
- Bio-Bank Department, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, No. 666 Shengli Road, Nantong, 226001, China.
| | - Chun-Feng Lu
- Department of Endocrinology, Affiliated Hospital 2 of Nantong University and First People's Hospital of Nantong City, No. 666 Shengli Road, Nantong, 226001, China.
| |
Collapse
|
2
|
Shahsavari K, Ardekani SS, Ardekani MRS, Esfahani MM, Kazemizadeh H, Jamialahmadi T, Iranshahi M, Khanavi M, Hasanpour M. Are alterations needed in Silybum marianum (Silymarin) administration practices? A novel outlook and meta-analysis on randomized trials targeting liver injury. BMC Complement Med Ther 2025; 25:134. [PMID: 40221681 PMCID: PMC11992775 DOI: 10.1186/s12906-025-04886-y] [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: 08/05/2024] [Accepted: 04/04/2025] [Indexed: 04/14/2025] Open
Abstract
It is widely believed that Silybum marianum (Silymarin) alleviates liver injury arising from various etiologies with different degrees of damage through its anti-inflammatory and antioxidant activities. This meta-analysis investigated the effects of silymarin administration on serum levels of liver enzymes including AST, ALT and ALP. From inception to November, 2023, a comprehensive literature search was conducted. Inclusion criteria for this study were randomized trials that provided sufficient data for each group at the beginning and end of the follow-up period. Ultimately, 55 studies with a total of 3545 patients were included. Comprehensive Meta-Analysis (CMA) V4 software was used for meta-analysis. Begg's funnel plot symmetry status, Begg's rank correlation, and Egger's weighted regression tests were used to examine potential publication bias. According to the findings of this meta-analysis silymarin administration showed a significant reduction in AST (SMD [95% CI]: - 0.670 [- 0.931, - 0.408], p-value = 0.000), and ALT (SMD [95% CI]: - 0.912 [- 1.177, - 0.646], p-value = 0.000) levels. While it had no statistically significant effect on ALP level (SMD [95% CI]: - 0.236 [- 1.929, 1.458], p-value = 0.159). Meta-regression analysis showed that there is no significant association between dose, age, BMI, treatment duration and hepatoprotective effects of silymarin. In subgroup analysis, a greater reduction in liver enzymes levels was observed in patients under 50 years old. The subgroup analysis was also showed significant decrease in AST and ALT levels for patients with BMI less than 30, while silymarin treatment had no significant effects on AST and ALT levels in patients with BMI ≥ 30. Silymarin at a dose of less than 400 mg and treatment duration ≤ 2 months showed greater decreasing effects on AST and ALT levels compared to its high doses and longer treatment duration. AST and ALT levels significantly decreased in patients with NAFLD and viral hepatitis, while it had no significant hepatoprotective effects in patients with drugs induced liver injury and alcohol-related liver disease. Modifying the dose and treatment duration with silymarin is recommended in patients with various causes of liver damage.
Collapse
Affiliation(s)
- Kasra Shahsavari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Reza Shams Ardekani
- Department of Pharmacognosy, Faculty of Pharmacy, and Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, PO Box: 14155 - 6451, Tehran, Iran
| | - Majid Mokaber Esfahani
- Department of Chemistry, Faculty of Science, Gonbad Kavous University, Gonbad Kavous, Iran
| | - Hossein Kazemizadeh
- Thoracic Research Center, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Tannaz Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahnaz Khanavi
- Department of Pharmacognosy, Faculty of Pharmacy, and Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, PO Box: 14155 - 6451, Tehran, Iran.
| | - Maede Hasanpour
- Department of Pharmacognosy, Faculty of Pharmacy, and Persian Medicine and Pharmacy Research Center, Tehran University of Medical Sciences, PO Box: 14155 - 6451, Tehran, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Liu L, Deng Y, Yang L, Wang M, Lai Y. Comparison of efficacy and safety of pioglitazone and SGLT2 inhibitors in treating Asian patients in MASLD associated with type 2 diabetes: A meta-analysis. J Diabetes Complications 2025; 39:108998. [PMID: 40043473 DOI: 10.1016/j.jdiacomp.2025.108998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 01/09/2025] [Accepted: 03/02/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVE To comprehensively evaluate the therapeutic efficacy of pioglitazone and SGLT2 inhibitors (SGLT2i) in patients with MASLD and Type 2 Diabetes(T2DM). METHODS Electronic databases, including Web of Science, PubMed, the Cochrane Library, China National Knowledge Internet (CNKI), Wan-Fang Digital Database, and China Science and Technology Journal Database (VIP) were searched from inception to December 2024. Two reviewers independently assessed study eligibility, performed continuous data extraction, and independently evaluated bias risks. Liver ultrasonography, computed tomography (CT), and biochemical indices were utilized to determine the impact of treatment in both groups. Improvement in liver biomarkers and fibrosis as primary outcome indicators; Improvement in body composition, metabolic parameters, glucose parameters, and incidence of adverse effects as a secondary outcome indicator. For continuous variables, mean and standard deviation (SD) were extracted. RevMan 5.4 software was used to systematically analyze the literature, including heterogeneity testing, odds ratios (OR) calculation, and 95 % confidence intervals (CI) for each influencing factor. RESULTS Nine randomly controlled trials with 755 Asian participants were included. Our research showed that SGLT2i was more effective than pioglitazone in improving fibrosis-4 score (SMD 0.41 [95%CI 0.18,0.64] p = 0.005), visceral fat area (SMD 0.34 [95%CI 0.14,0.54] p = 0.0007), BMI (SMD 0.29 [95%CI 0.03,0.56] p = 0.03), and low-density lipoprotein levels (SMD 0.21 [95%CI 0.04,0.38] p = 0.01). In contrast, no statistically significant differences were observed in other outcomes. CONCLUSIONS Our study demonstrated that in patients with MASLD and T2DM, SGLT2i was more effective overall in improving liver fibrosis, blood lipids, liver fat, and body composition in the short term. These findings establish a theoretical basis for safe and rational drug use in clinical practice. Additionally, they may contribute to new insights into the pathogenesis of MASLD and type 2 diabetes and drug discovery and development efforts.
Collapse
Affiliation(s)
- Lingyan Liu
- College of Pharmacy Dali University, Dali 671000, Yunnan Province, China
| | - Yongkun Deng
- Department of Medical Protection Center, The 926th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kaiyuan 661600, Yunnan Province, China.
| | - Lijuan Yang
- College of Pharmacy Dali University, Dali 671000, Yunnan Province, China
| | - Miaojiao Wang
- College of Pharmacy Dali University, Dali 671000, Yunnan Province, China
| | - Yong Lai
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali 671000, Yunnan Province, China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali 671000, Yunnan Province, China; College of Pharmacy Dali University, Dali 671000, Yunnan Province, China.
| |
Collapse
|
4
|
Shibamoto J, Otsuka S, Okawa Y, Ashida R, Ohgi K, Kato Y, Dei H, Uesaka K, Sugiura T. Prognostic Impact of Diabetes Mellitus and Extended Hepatectomy on Perihilar Cholangiocarcinoma. ANNALS OF SURGERY OPEN 2025; 6:e552. [PMID: 40134488 PMCID: PMC11932603 DOI: 10.1097/as9.0000000000000552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 03/27/2025] Open
Abstract
Objective To evaluate the prognostic impact of diabetes mellitus (DM) in patients who underwent resection for perihilar cholangiocarcinoma (PHCC) and the influence of remnant liver volumes on postoperative glycemic profiles and survival outcomes. Background The impact of DM and extended hepatectomy on survival outcomes of patients with PHCC remains unclear. Methods A total of 184 patients who underwent hepatectomy with extrahepatic bile duct resection for PHCC between 2002 and 2020 were retrospectively analyzed and divided into groups based on DM and future liver remnant (FLR) ≥40% or <40%. Survival outcomes and glycemic profiles were analyzed. Results Patients with DM (n = 34) had significantly worse overall survival compared with those without DM (n = 150; median survival time: 23.3 vs 46.7 months; P = 0.003) although cancer-specific survival was comparable (P = 0.894). Patients with DM had a higher incidence of death from infections (P < 0.001). Multivariate analysis identified DM as an independent prognostic factor (hazard ratio, 1.742; P = 0.021). DM with FLR <40% (n = 11) exhibited worse survival (median survival time: 13.7 vs 35.0 months; P = 0.026) and a higher incidence of death from infections (P = 0.016) compared with those with FLR ≥40% (n = 23). The median glucose fluctuation was larger in patients with DM and FLR <40% (80 vs 39 mg/dL; P = 0.023). Conclusions DM was an independent prognostic factor in patients with PHCC undergoing hepatectomy. DM and FLR <40% were associated with worse survival and larger glucose fluctuation postoperatively.
Collapse
Affiliation(s)
- Jun Shibamoto
- From the Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Shimpei Otsuka
- From the Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yuta Okawa
- Division of Endocrinology and Metabolism, Shizuoka Cancer Center, Shizuoka, Japan
| | - Ryo Ashida
- From the Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhisa Ohgi
- From the Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yoshiyasu Kato
- From the Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hideyuki Dei
- From the Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Katsuhiko Uesaka
- From the Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| | - Teiichi Sugiura
- From the Division of Hepato-Biliary-Pancreatic Surgery, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
5
|
Abdel Monem MS, Adel A, Abbassi MM, Abdelaziz DH, Hassany M, Raziky ME, Sabry NA. Efficacy and safety of dapagliflozin compared to pioglitazone in diabetic and non-diabetic patients with non-alcoholic steatohepatitis: A randomized clinical trial. Clin Res Hepatol Gastroenterol 2025; 49:102543. [PMID: 39884573 DOI: 10.1016/j.clinre.2025.102543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/21/2025] [Accepted: 01/26/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH) is a serious end-stage spectrum of non-alcoholic fatty liver disease (NAFLD) with associated high risk of hepatic and extrahepatic complications. Several studies showed the significant beneficial effect of dapagliflozin on body composition, hepatic and metabolic parameters on NAFLD/NASH patients. The study aimed to investigate the efficacy and safety of dapagliflozin in both diabetic and non-diabetic biopsy-proven NASH patients; compared to pioglitazone. METHODS This was a four-group, prospective, randomized, parallel, open label study in which 100 biopsy-proven NASH patients were selected, stratified to diabetics and non-diabetics and randomized with 1:1 allocation to either 30 mg pioglitazone or 10 mg dapagliflozin, once daily for 24 weeks. Histological evaluation, anthropometric measures, hepatic, metabolic biochemical markers, fibrosis non-invasive markers, quality of life (QOL) and medications adverse events were examined. RESULTS Dapagliflozin showed a comparable histological effect to pioglitazone in both diabetic and non-diabetic patients (P>0.05). As assessed by transient elastography, it also showed a comparable effect on liver fibrosis grade improvement from baseline in diabetics (P=0.287) versus a significant superiority in non-diabetics (P=0.018). Dapagliflozin showed a significant superiority in all anthropometric measures (P<0.001) and QOL (P<0.05) among both diabetics and non-diabetics. There was a significant interaction between interventions and diabetes status on change from baseline of hepatic and metabolic panel collectively (P=0.023) in favor to dapagliflozin among diabetics. CONCLUSION Compared to pioglitazone, dapagliflozin had a comparable effect histologically, superior effect biochemically among diabetics and superior effect on liver fibrosis, steatosis and insulin resistance among non-diabetics. TRIAL REGISTRATION The study was registered on clinicaltrials.gov, identifier number NCT05254626.
Collapse
Affiliation(s)
- Mona S Abdel Monem
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| | - Abdulmoneim Adel
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Maggie M Abbassi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| | - Doaa H Abdelaziz
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia/Department of Clinical Pharmacy, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Mohamed Hassany
- National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Maissa El Raziky
- Endemic Medicine and Hepatogastroenterology, Faculty of Medicine, Cairo University, Egypt.
| | - Nirmeen A Sabry
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Egypt.
| |
Collapse
|
6
|
Baghdadi G, Shidfar F, Mokhtare M, Sarbakhsh P, Agah S. Effect of Pomegranate Peel Consumption on Liver Enzymes, Lipid Profile, Liver Steatosis, and Hs-CRP in Patients With Non-alcoholic Fatty Liver Disease: A Double-Blind, Placebo-Controlled, Randomized Clinical Trial. Phytother Res 2025; 39:619-629. [PMID: 39608758 DOI: 10.1002/ptr.8404] [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: 12/17/2023] [Revised: 03/20/2024] [Accepted: 06/23/2024] [Indexed: 11/30/2024]
Abstract
The most prevalent chronic liver disease for which there is currently no proven treatment is non-alcoholic fatty liver disease (NAFLD). An incomplete understanding of the underlying mechanisms of NAFLD may be the cause. The onset and development of this illness appear to be influenced by problems with lipid metabolism, insulin resistance, oxidative stress, and inflammation. Considering the antioxidant properties of pomegranate peel extract, this study was conducted to determine the effects of pomegranate peel consumption on some metabolic features in patients with NAFLD. Our hypothesis is that pomegranate peel can improve the grade of fatty liver, liver enzymes, lipid profile, serum high-sensitivity C-reactive protein (hs-CRP), and anthropometric indices. The aim of this study was to investigate the efficacy of pomegranate peel extract in NAFLD patients. This double-blind randomized clinical trial was conducted on 46 patients with NAFLD. Patients were randomly assigned to intervention group (n = 23) and placebo group (n = 23). Patients in the pomegranate peel group consumed two capsules, each containing 500 mg pomegranate peel extract daily as a part of low-calorie diet (i.e., 500-deficit calorie diet) for 10 weeks. While patients in the control group followed the low calorie diet and two capsules containing 500 mg maltodextrin. At the beginning and end of the study, demographic information, anthropometric indices, food intake, physical activity level, grade of fatty liver, liver enzymes, lipid profile, and serum high-sensitivity C-reactive protein (hs-CRP) were measured. Food intake was measured by 24-h food recall questionnaires and physical activity was measured by the International Physical Activity Questionnaire (IPAQ). Analysis of food recall questionnaire was done using Nutritionist IV program. Statistical analysis was performed using SPSS software (version 22), and a p value < 0.05 was defined as statistically significant. Of 46 patients, 42 of them completed the trial. At the end of the trial, pomegranate peel group had significantly higher reduction in TG (triglycerides), ALT(alanine aminotransferase), AST(aspartate transferase), hs-CRP and also had higher significant increase in HDL-C(high-density lipoprotein cholesterol) compared to the control group (p = 0/02, p = 0/02, p = 0/01, p = 0/01, and p = 0/04, respectively). However, changes in LDL-C, TC, ALP, GGT, and fatty liver grade were not significantly different between the two groups at the end of the study. The current study indicates that pomegranate peel extract has a favorable effect on liver enzymes, lipid profile, and serum high-sensitivity C-reactive protein (hs-CRP) in patients with NAFLD. To support these results, trials examining various dosages over longer time periods are necessary.
Collapse
Affiliation(s)
- Ghazal Baghdadi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
- Nutritional Sciences Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Marjan Mokhtare
- Colo Rectal Research Center, Hazrat Rasool Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Parvin Sarbakhsh
- Department of Statistics and Epidemiology, School of Public Health, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahram Agah
- Nutritional Sciences Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Bhardwaj M, Mazumder PM. An insight on the additive impact of type 2 diabetes mellitus and nonalcoholic fatty liver disease on cardiovascular consequences. Mol Biol Rep 2025; 52:169. [PMID: 39873861 DOI: 10.1007/s11033-025-10249-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes mellitus (T2DM) are associated with a multifactorial complicated aetiology that is often coexisting and has a strong and distinct connection with cardiovascular diseases (CVDs). In order to accomplish effective and appropriate therapeutic strategies, a deeper understanding of the bidirectional interaction between NAFLD patients, NAFLD patients with T2DM, and NAFLD patients with CVDs is required to control the concomitant rise in prevalence of these conditions worldwide. This article also aims to shed light on the epidemiology and mechanisms behind the relationship between T2DM, NAFLD and the related cardiovascular consequences. METHOD Literature was collected from PubMed, Medline, Embase, Web of Science and Google scholar from inception to June, 2024. For surveying literature different combinations and formats of terms including NAFLD, NASH, T2DM and CVDs were used. RESULTS In the recent decade, clinical and epidemiological studies have been conducted and provide strong evidence that NAFLD is closely linked with CVD progression along with associated morbidity and mortality in both patients with and without T2DM. Several mechanistic approaches contribute to cardiovascular consequences and abnormalities in cardiac biomarkers in T2DM and NAFLD patients, including adipose tissue malfunction, mitochondrial dysfunction, the microbiota, genetic and epigenetic alterations contributing to insulin resistance, glucotoxicity and lipotoxicity. CONCLUSION The study reveals a complex interplay between diabetes, hepatic and cardiovascular complications, leading to significant morbidity and mortality in diabetic and NAFLD patients. This pandemic necessitates further research to identify mitigating variables and develop effective treatment approaches.
Collapse
Affiliation(s)
- Monika Bhardwaj
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India
| | - Papiya Mitra Mazumder
- Department of Pharmaceutical Sciences & Technology, BIT Mesra, Ranchi, 835215, India.
| |
Collapse
|
8
|
Sun X, Li F, Yan H, Chang X, Yao X, Yang X, Wu S, Suo Y, Zhu X, Wang C, Gao J, Wang H, Chen Y, Xia M, Bian H, Gao X. Intermittent compared with continuous calorie restriction for treatment of metabolic dysfunction-associated steatotic liver disease: a randomized clinical trial. Am J Clin Nutr 2025; 121:158-166. [PMID: 39447676 DOI: 10.1016/j.ajcnut.2024.10.012] [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: 06/16/2024] [Revised: 09/08/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Calorie restriction has been demonstrated to be effective in treating metabolic dysfunction-associated steatotic liver disease (MASLD). However, it has been limited by poor long-term adherence. OBJECTIVES This study aimed to compare intermittent calorie restriction (ICR) with traditional continuous calorie restriction (CCR) for the treatment of MASLD. METHODS We conducted a 12-wk, parallel-arm, randomized controlled trial that included 60 adults with MASLD and abnormal glucose metabolism. The participants were randomly assigned to either the ICR group (2 successive days of fasting [∼500 kcal/d] and 5 d of recovery per week) or the CCR group. The primary outcome was liver fat content (LFC) measured by 1H-proton magnetic resonance spectroscopy. The secondary and exploratory outcomes included weight, body composition, glucose, insulin, lipids, and liver stiffness. RESULTS The mean reduction in LFC was -20.5% [95% confidence interval (CI): -25.0, -15.9%] in the ICR group and -15.5% (95% CI: -20.3, -10.8%) in the CCR group. Changes in LFC were not significantly different between the 2 groups (P = 0.15), and were homogeneous among different liver segments. The analysis of exploratory endpoints provided clues that the ICR was associated with greater reductions in fat mass and glycosylated hemoglobin. There were no significant differences in changes of weight, lean mass, insulin resistance, triglyceride, and liver stiffness between the 2 groups. Participants showed high adherence to both the ICR and CCR schemes. CONCLUSIONS The ICR and CCR schemes had similar effects on reducing LFC, suggesting that the ICR 5:2 diet can be an effective alternative for treating MASLD with high adherence. TRIAL REGISTRATION NUMBER This trial was registered at clinicaltrials.gov as NCT04283942.
Collapse
Affiliation(s)
- Xiaoyang Sun
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Metabolic Disease, Fudan University, Shanghai, China
| | - Fan Li
- Department of Nutrition, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongmei Yan
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Metabolic Disease, Fudan University, Shanghai, China
| | - Xinxia Chang
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Metabolic Disease, Fudan University, Shanghai, China
| | - Xiuzhong Yao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinyu Yang
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Metabolic Disease, Fudan University, Shanghai, China
| | - Shasha Wu
- Department of Nutrition, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Suo
- School of Public Health, Fudan University, Shanghai, China
| | - Xiaopeng Zhu
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Metabolic Disease, Fudan University, Shanghai, China
| | - Chengyan Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Jian Gao
- Department of Nutrition, Zhongshan Hospital, Fudan University, Shanghai, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mingfeng Xia
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Metabolic Disease, Fudan University, Shanghai, China.
| | - Hua Bian
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Metabolic Disease, Fudan University, Shanghai, China.
| | - Xin Gao
- Department of Endocrinology, Zhongshan Hospital, Fudan University, Shanghai, China; Institute of Metabolic Disease, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Marginean CM, Pirscoveanu D, Cazacu SM, Popescu MS, Marginean IC, Iacob GA, Popescu M. Non-Alcoholic Fatty Liver Disease, Awareness of a Diagnostic Challenge—A Clinician’s Perspective. GASTROENTEROLOGY INSIGHTS 2024; 15:1028-1053. [DOI: 10.3390/gastroent15040071] [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] [Indexed: 01/03/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the main cause of chronic liver disease globally. NAFLD is a complex pathology, considered to be the hepatic expression of metabolic syndrome (MetS). It is supposed to become the main indication for liver transplantation in the coming years and is estimated to affect 57.5–74.0% of obese people, 22.5% of children and 52.8% of obese children, with 50% of individuals with type 2 diabetes being diagnosed with NAFLD. Recent research has proved that an increase in adipose tissue insulin resistance index is an important marker of liver injury in patients with NAFLD. Despite being the main underlying cause of incidental liver damage and a growing worldwide health problem, NAFLD is mostly under-appreciated. Currently, NAFLD is considered a multifactorial disease, with various factors contributing to its pathogenesis, associated with insulin resistance and diabetes mellitus, but also with cardiovascular, kidney and endocrine disorders (polycystic ovary syndrome, hypothyroidism, growth hormone deficiency). Hepatitis B and hepatitis C, sleep apnea, inflammatory bowel diseases, cystic fibrosis, viral infections, autoimmune liver diseases and malnutrition are some other conditions in which NAFLD can be found. The aim of this review is to emphasize that, from the clinician’s perspective, NAFLD is an actual and valuable key diagnosis factor for multiple conditions; thus, efforts need to be made in order to increase recognition of the disease and its consequences. Although there is no global consensus, physicians should consider screening people who are at risk of NAFLD. A large dissemination of current concepts on NAFLD and an extensive collaboration between physicians, such as gastroenterologists, internists, cardiologists, diabetologists, nutritionists and endocrinologists, is equally needed to ensure we have the knowledge and resources to address this public health challenge.
Collapse
Affiliation(s)
- Cristina Maria Marginean
- Internal Medicine Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Denisa Pirscoveanu
- Neurology Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Sergiu Marian Cazacu
- Research Center of Gastroenterology and Hepatology, Gastroenterology Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Marian Sorin Popescu
- Internal Medicine Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | | | - George Alexandru Iacob
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Mihaela Popescu
- Endocrinology Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
10
|
Weinstein AA, de Avila L, Fadahunsi AI, Price JK, Golabi P, Escheik C, Gerber LH, Younossi ZM. Liver disease-linked metabolic and behavioral factors associated with cognitive performance in an observational study of community dwelling adults. Medicine (Baltimore) 2024; 103:e40448. [PMID: 39809142 PMCID: PMC11596524 DOI: 10.1097/md.0000000000040448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/22/2024] [Indexed: 01/16/2025] Open
Abstract
Modifiable risk factors associated with cognitive functioning are important for identifying potential targets for intervention development. Although there are a few recognized modifiable risk factors (e.g., diabetes mellitus, diet, physical activity), there are limitations in the conclusions that can be drawn due to limited data. Therefore, this study examined the relationship between modifiable liver disease-linked metabolic and behavioral factors in a sample of community dwelling adults who do not currently experience functional limitations due to cognitive abilities. Individuals aged 19 to 69 were recruited to participate in this cross-sectional study in the Washington, DC area. Participants were assessed using anthropometric measures, ultrasound of the liver, glycated hemoglobin A1C, self-reported fatigue, clinical history, and 7 domains of cognitive function: processing speed, short- and long-term visual memory, working memory, inhibition, shifting, and abstract reasoning. The study included 104 participants (44% female, 51.1 ± 13.5 years old). The modifiable factors that were most consistently related to cognitive performance were waist-to-height ratio, which was related to a decrease in performance in 4 of the domains (short-term and long-term visual memory, working memory, and abstract reasoning), and the presence of nonalcoholic fatty liver disease, which was related to an increase in performance in the same 4 domains. This study suggests that liver disease-linked modifiable factors are associated with cognitive performance, even in middle-aged individuals without self-reported cognitive dysfunction. Further research is needed to explore the mechanisms that impact cognitive performance in relation to these factors to establish early intervention targets for reducing future cognitive deficits.
Collapse
Affiliation(s)
- Ali A. Weinstein
- Beatty Liver and Obesity Research Program, Inova Fairfax Medical Campus, Falls Church, VA
- Department of Global and Community Health, George Mason University, Fairfax, VA
| | - Leyla de Avila
- Beatty Liver and Obesity Research Program, Inova Fairfax Medical Campus, Falls Church, VA
| | | | - Jillian K. Price
- Beatty Liver and Obesity Research Program, Inova Fairfax Medical Campus, Falls Church, VA
| | - Pegah Golabi
- Beatty Liver and Obesity Research Program, Inova Fairfax Medical Campus, Falls Church, VA
| | - Carey Escheik
- Beatty Liver and Obesity Research Program, Inova Fairfax Medical Campus, Falls Church, VA
| | - Lynn H. Gerber
- Beatty Liver and Obesity Research Program, Inova Fairfax Medical Campus, Falls Church, VA
| | - Zobair M. Younossi
- Beatty Liver and Obesity Research Program, Inova Fairfax Medical Campus, Falls Church, VA
| |
Collapse
|
11
|
Ahmad R, Haque M. Metformin: Beyond Type 2 Diabetes Mellitus. Cureus 2024; 16:e71730. [PMID: 39421288 PMCID: PMC11486535 DOI: 10.7759/cureus.71730] [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: 09/28/2024] [Accepted: 10/17/2024] [Indexed: 10/19/2024] Open
Abstract
Metformin was developed from an offshoot of Guanidine. It is known to be the first-line medication for type 2 diabetes mellitus, polycystic ovarian syndrome, and weight reduction. Metformin has also been shown to have effectiveness in the management of non-alcoholic fatty liver disease (NAFLD), liver cirrhosis, and various carcinomas like hepatocellular, colorectal, prostate, breast, urinary bladder, blood, melanoma, bone, skin, lung and so on. This narrative review focuses on the effect of metformin on non-alcoholic fatty liver disease, liver cirrhosis, and hepatocellular carcinoma. The search platforms for the topic were PubMed, Scopus, and Google search engine. Critical words for searching included 'Metformin,' AND 'Indications of Metformin,' AND 'Non-Alcoholic Fatty Liver Disease,' AND 'Metformin mechanism of action,' AND 'NAFLD management,' AND 'NAFLD and inflammation,' AND 'Metformin and insulin,' AND 'Metformin and inflammation,' AND 'Liver cirrhosis,' AND 'Hepatocellular carcinoma.' Lifestyle modification and the use of hypoglycemic agents can help improve liver conditions. Metformin has several mechanisms that enhance liver health, including reducing reactive oxygen species, nuclear factor kappa beta (NF-κB), liver enzymes, improving insulin sensitivity, and improving hepatic cell lipophagy. Long-term use of metformin may cause some adverse effects like lactic acidosis and gastrointestinal disturbance. Metformin long-term overdose may lead to a rise in hydrogen sulfide in liver cells, which calls for pharmacovigilance. Drug regulating authorities should provide approval for further research, and national and international guidelines need to be developed for liver diseases, perhaps with the inclusion of metformin as part of the management regime.
Collapse
Affiliation(s)
- Rahnuma Ahmad
- Department of Physiology, Medical College for Women and Hospital, Dhaka, BGD
| | - Mainul Haque
- Department of Pharmacology and Therapeutics, National Defence University of Malaysia, Kuala Lumpur, MYS
| |
Collapse
|
12
|
Dayanand Y, Pather R, Xulu N, Booysen I, Sibiya N, Khathi A, Ngubane P. Exploring the Biological Effects of Anti-Diabetic Vanadium Compounds in the Liver, Heart and Brain. Diabetes Metab Syndr Obes 2024; 17:3267-3278. [PMID: 39247428 PMCID: PMC11380877 DOI: 10.2147/dmso.s417700] [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: 05/28/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2024] Open
Abstract
The prevalence of diabetes mellitus and diabetes-related complications is rapidly increasing worldwide, placing a substantial financial burden on healthcare systems. Approximately 537 million adults are currently diagnosed with type 1 or type 2 diabetes globally. However, interestingly, the increasing morbidity rate is primarily influenced by the effects of long-term hyperglycemia on vital organs such as the brain, the liver and the heart rather than the ability of the body to use glucose effectively. This can be attributed to the summation of the detrimental effects of excessive glucose on major vascular systems and the harmful side effects attributed to the current treatment associated with managing the disease. These drugs have been implicated in the onset and progression of cardiovascular disease, hepatocyte injury and cognitive dysfunction, thereby warranting extensive research into alternative treatment strategies. Literature has shown significant progress in utilizing metal-based compounds, specifically those containing transition metals such as zinc, magnesium and vanadium, in managing hyperglycaemia. Amongst these metals, research carried out on vanadium reflected the most promising anti-diabetic efficacy in cell culture and animal studies. This was attributed to the ability to improve glucose management in the bloodstream by enhancing its uptake and metabolism in the kidney, brain, skeletal muscle, heart and liver. Despite this, organic vanadium was considered toxic due to its accumulative characteristics. To alleviate vanadium's toxic nature while subsequently manipulating its therapeutic properties, vanadium complexes were synthesized using either vanadate or vanadyl as a base compound. This review attempts to evaluate organic vanadium salts' therapeutic and toxic effects, highlight vanadium complexes' research and provide insight into the novel dioxidovanadium complex synthesized in our laboratory to alleviate hyperglycaemia-associated macrovascular complications in the brain, heart and liver.
Collapse
Affiliation(s)
- Yalka Dayanand
- School of Laboratory Medicine and Medical Science, University of Kwazulu-Natal, Durban, South Africa
| | - Reveshni Pather
- School of Laboratory Medicine and Medical Science, University of Kwazulu-Natal, Durban, South Africa
| | - Nombuso Xulu
- School of Laboratory Medicine and Medical Science, University of Kwazulu-Natal, Durban, South Africa
| | - Irvin Booysen
- School of Chemistry and Physics, University of Kwazulu-Natal, Pietermaritzburg, South Africa
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Grahamstown, South Africa
| | - Andile Khathi
- School of Laboratory Medicine and Medical Science, University of Kwazulu-Natal, Durban, South Africa
| | - Phikelelani Ngubane
- School of Laboratory Medicine and Medical Science, University of Kwazulu-Natal, Durban, South Africa
| |
Collapse
|
13
|
Lebwohl M, Merola JF, Strober B, Armstrong A, Yoshizaki A, Gisondi P, Szilagyi B, Peterson L, de Cuyper D, Cross N, Davies O, Gottlieb AB. Bimekizumab safety in moderate to severe plaque psoriasis: Rates of hepatic events and changes in liver parameters over 2 years in randomized phase 3/3b trials. J Am Acad Dermatol 2024; 91:281-289. [PMID: 38588819 DOI: 10.1016/j.jaad.2024.03.041] [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: 11/30/2023] [Revised: 02/23/2024] [Accepted: 03/12/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND Patients with psoriasis are at increased risk of liver function abnormalities. OBJECTIVE Explore rates of hepatic treatment-emergent adverse events (TEAEs) and changes in liver parameters in bimekizumab-treated patients with psoriasis. METHODS Data are reported from 5 phase 3/3b trials over 2 years. Hepatic TEAEs, laboratory elevations in alanine aminotransferase (ALT) or aspartate aminotransferase (AST), and changes in clinical markers of liver fibrosis (Fibrosis-4 [FIB-4] Index and AST to Platelet Ratio Index [APRI]) are reported. TEAEs are presented using exposure-adjusted incidence rates (EAIRs) per 100 patient-years (PY). RESULTS 2186 patients received ≥1 bimekizumab dose. Over 2 years, the EAIR of hepatic TEAEs was 3.5/100 PY and did not increase from first to second year. 2-year EAIRs of ALT/AST elevations >3x and >5x the upper limit of normal were 2.3 and 0.6/100 PY; rates were similar to placebo, adalimumab, secukinumab, and ustekinumab during controlled study periods. FIB-4 and APRI scores did not increase through 2 years, regardless of fibrosis risk at baseline. LIMITATIONS Obesity, diabetes, dyslipidemia, chronic alcohol consumption, and medication changes are confounding factors for hepatic dysfunction. CONCLUSION Rates of hepatic adverse events (AEs) with bimekizumab were consistent through 2 years; incidences of transaminase elevations were similar to comparators during phase 3/3b controlled study periods.
Collapse
Affiliation(s)
- Mark Lebwohl
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Joseph F Merola
- Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts
| | - Bruce Strober
- Department of Dermatology, Yale University, New Haven, Connecticut; Central Connecticut Dermatology Research, Cromwell, Connecticut
| | - April Armstrong
- University of California Los Angeles (UCLA), Los Angeles, California
| | - Ayumi Yoshizaki
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan; Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Paolo Gisondi
- Section of Dermatology and Venereology, Department of Medicine, University of Verona, Verona, Italy
| | | | | | | | | | | | - Alice B Gottlieb
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
14
|
Bridi L, Agrawal S, Tesfai K, Madamba E, Bettencourt R, Richards LM, Khera AV, Loomba R, Ajmera V. The impact of genetic risk on the prevalence of advanced fibrosis and cirrhosis in prospectively assessed patients with type 2 diabetes. Aliment Pharmacol Ther 2024; 60:369-377. [PMID: 38825972 PMCID: PMC11236495 DOI: 10.1111/apt.18099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/20/2024] [Accepted: 05/17/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Genetic factors contribute to the risk and severity of metabolic dysfunction-associated steatotic liver disease (MASLD). However, the utility of genetic testing in risk stratification remains poorly characterised. AIMS To examine the influence of genetic risk on advanced fibrosis and cirrhosis in patients with type 2 diabetes mellitus (T2DM) and the utility of a polygenic risk score (PRS) in screening guidelines. METHODS We prospectively enrolled adults aged ≥50 years with T2DM recruited from clinics. PRS was the sum of risk alleles in PNPLA3, TM6SF2 and SERPINA1 minus the protective variant in HSD17B13. We performed magnetic resonance elastography and vibration-controlled transient elastography to assess for advanced fibrosis and cirrhosis. RESULTS Of 382 included patients, the mean age and BMI were 64.8 (±8.4) years and 31.7 (±6.2) kg/m2 respectively. The prevalence of advanced fibrosis and cirrhosis were 12.3% and 5.2% respectively; higher PRS was associated with increased risk of cirrhosis (2.7% vs. 7.5%, p = 0.037). High PRS was associated with increased risk of advanced fibrosis among those with fibrosis-4 index (FIB-4) index <1.3 (9.6% vs. 2.3%, p = 0.036) but was not significantly different in other FIB-4 categories. Incorporating PRS determination into the American Gastroenterological Association and American Association for the Study of Liver Diseases screening guidelines prevented approximately 20% of all participants with advanced fibrosis from being inappropriately classified as low risk. CONCLUSIONS Utilising a well-phenotyped, prospective cohort of adults with T2DM, we found that adding an assessment of genetic risk to recommendations to screen at-risk populations may improve risk prediction.
Collapse
Affiliation(s)
- Lana Bridi
- MASLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Saaket Agrawal
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Kaleb Tesfai
- MASLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Egbert Madamba
- MASLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Ricki Bettencourt
- MASLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Lisa M Richards
- MASLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Amit V Khera
- Division of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Verve Therapeutics, Boston, Massachusetts, USA
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| | - Veeral Ajmera
- MASLD Research Center, Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
- Division of Gastroenterology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
15
|
Ferenc K, Jarmakiewicz-Czaja S, Sokal-Dembowska A, Stasik K, Filip R. Common Denominator of MASLD and Some Non-Communicable Diseases. Curr Issues Mol Biol 2024; 46:6690-6709. [PMID: 39057041 PMCID: PMC11275402 DOI: 10.3390/cimb46070399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Currently, steatohepatitis has been designated as metabolic dysfunction-associated steatohepatitis (MASLD). MASLD risk factors mainly include metabolic disorders but can also include genetic, epigenetic, and environmental factors. Disease entities such as obesity, diabetes, cardiovascular disease, and MASLD share similar pathomechanisms and risk factors. Moreover, a bidirectional relationship is observed between the occurrence of certain chronic diseases and MASLD. These conditions represent a global public health problem that is responsible for poor quality of life and high mortality. It seems that paying holistic attention to these problems will not only help increase the chances of reducing the incidence of these diseases but also assist in the prevention, treatment, and support of patients.
Collapse
Affiliation(s)
- Katarzyna Ferenc
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (K.F.)
| | - Sara Jarmakiewicz-Czaja
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Katarzyna Stasik
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (K.F.)
- IBD Unit, Department of Gastroenterology, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| | - Rafał Filip
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (K.F.)
- IBD Unit, Department of Gastroenterology, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
16
|
Rinaldi R, De Nucci S, Donghia R, Donvito R, Cerabino N, Di Chito M, Penza A, Mongelli FP, Shahini E, Zappimbulso M, Pesole PL, Coletta S, Triggiani V, Cozzolongo R, Giannelli G, De Pergola G. Gender Differences in Liver Steatosis and Fibrosis in Overweight and Obese Patients with Metabolic Dysfunction-Associated Steatotic Liver Disease before and after 8 Weeks of Very Low-Calorie Ketogenic Diet. Nutrients 2024; 16:1408. [PMID: 38794646 PMCID: PMC11123918 DOI: 10.3390/nu16101408] [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: 04/09/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/26/2024] Open
Abstract
Obesity and metabolic syndrome are linked to steatotic liver disease (SLD), the most common form of chronic liver disease. Lifestyle modifications and dieting are strategies that can prevent metabolic dysfunction-associated steatotic liver disease (MASLD). The very low-calorie ketogenic diet (VLCKD) is a helpful treatment for MASLD and has been recommended for people affected by obesity; we evaluated the effect of gender on steatosis and fibrosis in a cohort of 112 overweight or obese patients undergoing an eight-week treatment with a VLCKD. Differences between the genders in terms of anthropometric measures, body composition, and metabolic indicators were examined before, during, and after the nutritional intervention. At baseline, there were significant differences between men and women in terms of anthropometric parameters, blood pressure, Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), fasting insulin, hepatic markers, and lipid profile. Men had considerably higher levels of liver steatosis (measured by CAP) and liver stiffness (measured by E) under basal conditions than women. After the VLCKD, there were reductions in both genders of controlled attenuation parameter (CAP), body weight, body mass index (BMI), waist circumference, systolic and diastolic blood pressure, insulin resistance, fat mass (FM), free fat mass (FFM), and fasting blood glucose, insulin, glycated hemoglobin (HbA1c), triglycerides, total cholesterol, low-density lipoprotein (LDL) cholesterol, alanine transaminase (ALT), gamma-glutamyl transferase (γGT), and uric acid levels. Only in men, liver stiffness, aspartate aminotransferase (AST), creatinine, and C-reactive protein (CRP) levels significantly decreased. Moreover, men had significantly greater levels of liver steatosis: the male gender featured an increase of 23.96 points of the Fibroscan CAP. Men exhibited higher levels of steatosis and fibrosis than women, and these differences persist despite VLCKD. These gender-specific variations in steatosis and fibrosis levels could be caused by hormonal and metabolic factors, suggesting that different therapeutic strategies might be required depending on the gender.
Collapse
Affiliation(s)
- Roberta Rinaldi
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.R.); (S.D.N.); (R.D.); (N.C.); (M.D.C.); (A.P.); (F.P.M.)
| | - Sara De Nucci
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.R.); (S.D.N.); (R.D.); (N.C.); (M.D.C.); (A.P.); (F.P.M.)
| | - Rossella Donghia
- Laboratory of Epidemiology and Statistics, National Institute of Gastroenterology “Saverio de Bellis”, IRCCS Hospital, Castellana Grotte, 70013 Bari, Italy;
| | - Rosanna Donvito
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.R.); (S.D.N.); (R.D.); (N.C.); (M.D.C.); (A.P.); (F.P.M.)
| | - Nicole Cerabino
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.R.); (S.D.N.); (R.D.); (N.C.); (M.D.C.); (A.P.); (F.P.M.)
| | - Martina Di Chito
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.R.); (S.D.N.); (R.D.); (N.C.); (M.D.C.); (A.P.); (F.P.M.)
| | - Alice Penza
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.R.); (S.D.N.); (R.D.); (N.C.); (M.D.C.); (A.P.); (F.P.M.)
| | - Francesco Pio Mongelli
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.R.); (S.D.N.); (R.D.); (N.C.); (M.D.C.); (A.P.); (F.P.M.)
| | - Endrit Shahini
- Department of Gastroenterology, National Institute of Gastroenterology “Saverio de Bellis”, IRCCS Hospital, Castellana Grotte, 70013 Bari, Italy; (E.S.); (M.Z.); (R.C.)
| | - Marianna Zappimbulso
- Department of Gastroenterology, National Institute of Gastroenterology “Saverio de Bellis”, IRCCS Hospital, Castellana Grotte, 70013 Bari, Italy; (E.S.); (M.Z.); (R.C.)
| | - Pasqua Letizia Pesole
- Core Facility Biobank, National Institute of Gastroenterology “Saverio de Bellis”, IRCCS Hospital, Castellana Grotte, 70013 Bari, Italy; (P.L.P.); (S.C.)
| | - Sergio Coletta
- Core Facility Biobank, National Institute of Gastroenterology “Saverio de Bellis”, IRCCS Hospital, Castellana Grotte, 70013 Bari, Italy; (P.L.P.); (S.C.)
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Raffaele Cozzolongo
- Department of Gastroenterology, National Institute of Gastroenterology “Saverio de Bellis”, IRCCS Hospital, Castellana Grotte, 70013 Bari, Italy; (E.S.); (M.Z.); (R.C.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology “Saverio de Bellis”, IRCCS Hospital, Castellana Grotte, 70013 Bari, Italy;
| | - Giovanni De Pergola
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (R.R.); (S.D.N.); (R.D.); (N.C.); (M.D.C.); (A.P.); (F.P.M.)
| |
Collapse
|
17
|
Ekpor E, Akyirem S, Adade Duodu P. Prevalence of metabolic dysfunction-associated fatty liver disease and its association with glycemic control in persons with type 2 diabetes in Africa: A systematic review and meta-analysis. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0002835. [PMID: 38709759 PMCID: PMC11073701 DOI: 10.1371/journal.pgph.0002835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/11/2024] [Indexed: 05/08/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) and type 2 diabetes (T2D) are interconnected metabolic disorders with significant health implications. However, a comprehensive understanding of the extent of their co-occurrence in Africa is lacking. The aim of this review was to determine the prevalence of MAFLD and its association with glycemic control (HbA1c) in persons with T2D in Africa. A systematic search was conducted on PubMed, Medline, Embase, Scopus, Global Health, and Web of Science from their inception to December 6, 2023. Data on MAFLD prevalence and correlation coefficients regarding its association with glycemic control were pooled through random effect meta-analyses. Potential sources of heterogeneity were investigated using subgroup analysis and meta-regression. A total of 10 studies were included in the meta-analysis of MAFLD prevalence, while 2 were incorporated in the analysis of the association between MAFLD and glycemic control. The pooled prevalence of MAFLD in persons with T2D was 48.1% (95% CI: 36.1-60.3). The subgroup analysis revealed regional variations in MAFLD prevalence, with rates of 44.7% (95% CI: 28.7-62.0) in sub-Saharan Africa and 55.3% (95% CI: 36.2-73.0) in Northern Africa. Additionally, we observed an increasing trend in MAFLD prevalence, recording 55.1% (95% CI: 43.6-66.1) in the recent five years. There was a weak positive correlation between MAFLD and HbA1c (r = 0.33, 95% CI: 0.18-0.47). The findings of this study highlight a high prevalence of MAFLD in persons with T2D in Africa, with a suggested link between MAFLD and suboptimal glycemic control. Therefore, healthcare providers should prioritize the screening and management of MAFLD in individuals with T2D to enhance their metabolic health.
Collapse
Affiliation(s)
- Emmanuel Ekpor
- School of Nursing and Midwifery, University of Ghana, Accra, Ghana
- Christian Health Association of Ghana, Accra, Ghana
| | - Samuel Akyirem
- Yale School of Nursing, Yale University, New Haven, Connecticut, United States of America
| | - Precious Adade Duodu
- Department of Nursing, School of Human and Health Sciences, University of Huddersfield, Huddersfield, England, United Kingdom
| |
Collapse
|
18
|
Yasin A, Nguyen M, Sidhu A, Majety P, Spitz J, Asgharpour A, Siddiqui MS, Sperling LS, Quyyumi AA, Mehta A. Liver and cardiovascular disease outcomes in metabolic syndrome and diabetic populations: Bi-directional opportunities to multiply preventive strategies. Diabetes Res Clin Pract 2024; 211:111650. [PMID: 38604447 DOI: 10.1016/j.diabres.2024.111650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/13/2024]
Abstract
The incidence and prevalence of metabolic syndrome (MetS) and type 2 diabetes mellitus (T2DM) are rising globally. MetS and T2DM are associated with significant morbidity and mortality, which is partly related to liver and cardiovascular disease. Insulin resistance is central to MetS and T2DM pathophysiology, and drives ectopic fat deposition in the liver, also known as metabolic dysfunction-associated steatotic liver disease (MASLD). MetS and T2DM are not only risk factors for developing MASLD but are also independently associated with disease progression to steatohepatitis, cirrhosis, and hepatocellular carcinoma. In addition to the risk of liver disease, MetS and T2DM are independent risk factors for cardiovascular disease (CVD), including coronary artery disease (CAD) and heart failure (HF). Importantly, there is a bidirectional relationship between liver and CVD due to shared disease pathophysiology in patients with MetS and T2DM. In this review, we have described studies exploring the relationship of MetS and T2DM with MASLD and CVD, independently. Following this we discuss studies evaluating the interplay between liver and cardiovascular risk as well as pragmatic risk mitigation strategies in this patient population.
Collapse
Affiliation(s)
| | | | - Angad Sidhu
- Virginia Commonwealth University, Richmond, VA, US
| | | | - Jared Spitz
- Inova Heart and Vascular Institute, Fairfax, VA, US
| | | | | | | | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Atlanta, Georgia
| | - Anurag Mehta
- Virginia Commonwealth University, Richmond, VA, US.
| |
Collapse
|
19
|
Younossi ZM, Henry L. Epidemiology of NAFLD - Focus on diabetes. Diabetes Res Clin Pract 2024; 210:111648. [PMID: 38569945 DOI: 10.1016/j.diabres.2024.111648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 04/01/2024] [Indexed: 04/05/2024]
Abstract
There is increasing appreciation of the complex interaction between nonalcoholic fatty liver disease (NAFLD) with type 2 diabetes (T2D) and insulin resistance. Not only is the prevalence of NAFLD disease high among patients with T2D, the liver disease is also more progressive. Currently, the global prevalence of NAFLD in the general population (2016-2019) is 38 %. The prevalence of T2D among those with NAFLD is approximately 23 % while the prevalence of NAFLD among those with T2D can be as high as 70 %. The prevalence of nonalcoholic steatohepatitis (NASH) is approximately 7 % in the general population and 37 % among patients with T2D. Globally, the MENA and Latin America regions of the world appear to have the highest burden of both NAFLD and T2D. Compared to those with NAFLD but without T2D, those with NAFLD and T2D are at a much higher risk for disease progression to cirrhosis and for decompensated cirrhosis, hepatocellular carcinoma, and all-cause mortality. Given that highly effective new treatments are available for T2D, high risk NAFLD with T2D should be considered for these regimens. This requires implementation of risk stratification algorithms in the primary care and endocrinology practices to identify those patients at highest risk for adverse outcomes.
Collapse
Affiliation(s)
- Zobair M Younossi
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, United States; Center for Liver Disease, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, United States; Center for Outcomes Research In Liver Diseases, Washington, DC, United States.
| | - Linda Henry
- Betty and Guy Beatty Center for Integrated Research, Inova Health System, Falls Church, VA, United States; Center for Liver Disease, Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA, United States; Center for Outcomes Research In Liver Diseases, Washington, DC, United States
| |
Collapse
|
20
|
Tarantino G, Citro V. What are the common downstream molecular events between alcoholic and nonalcoholic fatty liver? Lipids Health Dis 2024; 23:41. [PMID: 38331795 PMCID: PMC10851522 DOI: 10.1186/s12944-024-02031-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Liver fat storage, also called hepatic steatosis, is increasingly common and represents a very frequent diagnosis in the medical field. Excess fat is not without consequences. In fact, hepatic steatosis contributes to the progression toward liver fibrosis. There are two main types of fatty liver disease, alcoholic fatty liver disease (AFLD) and nonalcoholic fatty liver disease (NAFLD). Although AFLD and NAFLD are similar in their initial morphological features, both conditions involve the same evolutive forms. Moreover, there are various common mechanisms underlying both diseases, including alcoholic liver disease and NAFLD, which are commonalities. In this Review, the authors explore similar downstream signaling events involved in the onset and progression of the two entities but not completely different entities, predominantly focusing on the gut microbiome. Downstream molecular events, such as the roles of sirtuins, cytokeratins, adipokines and others, should be considered. Finally, to complete the feature, some new tendencies in the therapeutic approach are presented.
Collapse
Affiliation(s)
| | - Vincenzo Citro
- Department of General Medicine, Umberto I Hospital, Nocera Inferiore, SA, 84014, Italy
| |
Collapse
|
21
|
Kaneva AM, Bojko ER. Fatty liver index (FLI): more than a marker of hepatic steatosis. J Physiol Biochem 2024; 80:11-26. [PMID: 37875710 DOI: 10.1007/s13105-023-00991-z] [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: 07/21/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023]
Abstract
Fatty liver index (FLI) was developed as a simple and accurate marker of hepatic steatosis. FLI is derived from an algorithm based on body mass index, waist circumference, and levels of triglycerides and gamma-glutamyltransferase, and it is widely used in clinical and epidemiological studies as a screening tool for discriminating between healthy and nonalcoholic fatty liver disease (NAFLD) subjects. However, a systematic review of the literature regarding FLI revealed that this index has more extensive relationships with biochemical and physiological parameters. FLI is associated with key parameters of lipid, protein and carbohydrate metabolism, hormones, vitamins and markers of inflammation, or oxidative stress. FLI can be a predictor or risk factor for a number of metabolic and nonmetabolic diseases and mortality. FLI is also used as an indicator for determining the effects of health-related prevention interventions, medications, and toxic substances on humans. Although in most cases, the exact mechanisms underlying these associations have not been fully elucidated, they are most often assumed to be mediated by insulin resistance, inflammation, and oxidative stress. Thus, FLI may be a promising marker of metabolic health due to its multiple associations with parameters of physiological and pathological processes. In this context, the present review summarizes the data from currently available literature on the associations between FLI and biochemical variables and physiological functions. We believe that this review will be of interest to researchers working in this area and can provide new perspectives and directions for future studies on FLI.
Collapse
Affiliation(s)
- Anastasiya M Kaneva
- Institute of Physiology of Кomi Science Centre of the Ural Branch of the Russian Academy of Sciences, FRC Komi SC UB RAS, 50 Pervomayskaya str., 167982, Syktyvkar, Russia.
| | - Evgeny R Bojko
- Institute of Physiology of Кomi Science Centre of the Ural Branch of the Russian Academy of Sciences, FRC Komi SC UB RAS, 50 Pervomayskaya str., 167982, Syktyvkar, Russia
| |
Collapse
|
22
|
Yang CR, Lin WJ, Shen PC, Liao PY, Dai YC, Hung YC, Lai HC, Mehmood S, Cheng WC, Ma WL. Phenotypic and metabolomic characteristics of mouse models of metabolic associated steatohepatitis. Biomark Res 2024; 12:6. [PMID: 38195587 PMCID: PMC10777576 DOI: 10.1186/s40364-023-00555-9] [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: 09/30/2023] [Accepted: 12/29/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Metabolic associated steatohepatitis (MASH) is metabolic disease that may progress to cirrhosis and hepatocellular carcinoma. Mouse models of diet-induced MASH, which is characterized by the high levels of fats, sugars, and cholesterol in diets, are commonly used in research. However, mouse models accurately reflecting the progression of MASH in humans remain to be established. Studies have explored the potential use of serological metabolites as biomarkers of MASH severity in relation to human MASH. METHODS We performed a comparative analysis of three mouse models of diet-induced MASH in terms of phenotypic and metabolomic characteristics; MASH was induced using different diets: a high-fat diet; a Western diet; and a high-fat, high-cholesterol diet. Liver cirrhosis was diagnosed using standard clinical approaches (e.g., METAVIR score, hyaluronan level, and collagen deposition level). Mouse serum samples were subjected to nuclear magnetic resonance spectroscopy-based metabolomic profiling followed by bioinformatic analyses. Metabolomic analysis of a retrospective cohort of patients with hepatocellular carcinoma was performed; the corresponding cirrhosis scores were also evaluated. RESULTS Using clinically relevant quantitative diagnostic methods, the severity of MASH was evaluated. Regarding metabolomics, the number of lipoprotein metabolites increased with both diet and MASH progression. Notably, the levels of very low-density lipoprotein (VLDL) and low-density lipoprotein (LDL) significantly increased with fibrosis progression. During the development of diet-induced MASH in mice, the strongest upregulation of expression was noted for VLDL receptor. Metabolomic analysis of a retrospective cohort of patients with cirrhosis indicated lipoproteins (e.g., VLDL and LDL) as predominant biomarkers of cirrhosis. CONCLUSIONS Our findings provide insight into the pathophysiology and metabolomics of experimental MASH and its relevance to human MASH. The observed upregulation of lipoprotein expression reveals a feedforward mechanism for MASH development that may be targeted for the development of noninvasive diagnosis.
Collapse
Affiliation(s)
- Cian-Ru Yang
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Jen Lin
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Chun Shen
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan
| | - Pei-Yin Liao
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan
| | - Yuan-Chang Dai
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi City, Taiwan
| | - Yao-Ching Hung
- Department of Gynecology and Obstetrics, Asia University Hospital, Taichung, Taiwan
| | - Hsueh-Chou Lai
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Shiraz Mehmood
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chung Cheng
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan.
| | - Wen-Lung Ma
- Program for Health Science and Industry, Graduate Institute of Biomedical Sciences, and Department of Medicine, and Tumor Biology Center, School of Medicine, China Medical University, Taichung, Taiwan.
- Department of Medical Research, Department of Gynecology and Obstetrics, and Department of Gastroenterology, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
23
|
Scurt FG, Ganz MJ, Herzog C, Bose K, Mertens PR, Chatzikyrkou C. Association of metabolic syndrome and chronic kidney disease. Obes Rev 2024; 25:e13649. [PMID: 37783465 DOI: 10.1111/obr.13649] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/15/2023] [Indexed: 10/04/2023]
Abstract
The prevalence of kidney disease is increasing rapidly worldwide, reflecting rising rates of obesity, diabetes, and associated metabolic syndrome (MetS). Chronic kidney disease and related comorbidities such as obesity, diabetes, and hypertension place a significant financial burden on healthcare systems. Despite the widespread use of RAAS inhibitors, intensive blood pressure and glycemic control, and newer therapeutic options consisting of sodium/glucose cotransporter-2 (SGLT-2) inhibitors or glucagon-like peptide-1 (GLP-1) receptor agonists, a significant risk of progression to end-stage renal disease remains in the high-risk obese and diabetic population. The MetS is a cluster of cardiovascular risk factors that adversely affect the development and progression of chronic kidney failure. According to the criteria of the World Health Organization, it is defined by visceral adiposity, impaired glucose tolerance or insulin resistance, atherogenic dyslipidemia, raised blood pressure, and microalbuminuria with a albumin-to-creatinine ratio ≥30 mg/g. At molecular level MetS is marked by a proinflammatory state and increased oxidative stress leading to various pathophysiological changes causing endothelial dysfunction and a hypercoagulable state. Because the kidney is a highly vascularized organ, it is especially susceptible for those microvascular changes. Therefore, the MetS and its individual components are associated with the premature development, acceleration, and progression of chronic kidney disease. Therefore, it is becoming increasingly important to elucidate the underlying mechanisms of MetS-associated chronic kidney disease in order to develop new strategies for preventing and slowing the progression of renal disease. In this review, we will elucidate (i) the renal structural, hemodynamic, and metabolic changes that occur in obesity and obesity-related kidney injury; (ii) the clinicopathological characteristics of obesity-related kidney injury, primarily focusing on obesity-associated glomerulopathy; (iii) the potential additional factors or predisposing factors that may turn patients more susceptible to renal structural or functional compensatory failure and subsequent injury.
Collapse
Affiliation(s)
- Florian G Scurt
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, Medical Faculty, Otto-von Guericke University Magdeburg, Magdeburg, Germany
| | - Maximilian J Ganz
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, Medical Faculty, Otto-von Guericke University Magdeburg, Magdeburg, Germany
| | - Carolin Herzog
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, Medical Faculty, Otto-von Guericke University Magdeburg, Magdeburg, Germany
| | - Katrin Bose
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Magdeburg, Magdeburg, Germany
| | - Peter R Mertens
- University Clinic for Nephrology and Hypertension, Diabetology and Endocrinology, Medical Faculty, Otto-von Guericke University Magdeburg, Magdeburg, Germany
| | | |
Collapse
|
24
|
Cheung JTK, Zhang X, Wong GLH, Yip TCF, Lin H, Li G, Leung HHW, Lai JCT, Mahadeva S, Nik Mustapha NR, Wang XD, Liu WY, Wong VWS, Chan WK, Zheng MH. MAFLD fibrosis score: Using routine measures to identify advanced fibrosis in metabolic-associated fatty liver disease. Aliment Pharmacol Ther 2023; 58:1194-1204. [PMID: 37724633 DOI: 10.1111/apt.17722] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/06/2023] [Accepted: 09/07/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Early screening may prevent fibrosis progression in metabolic-associated fatty liver disease (MAFLD). AIMS We developed and validated MAFLD fibrosis score (MFS) for identifying advanced fibrosis (≥F3) among MAFLD patients. METHODS This cross-sectional, multicentre study consecutively recruited MAFLD patients receiving tertiary care (Malaysia as training cohort [n = 276] and Hong Kong and Wenzhou as validation cohort [n = 431]). Patients completed liver biopsy, vibration-controlled transient elastography (VCTE), and clinical and laboratory assessment within 1 week. We used machine learning to select 'highly important' predictors of advanced fibrosis, followed by backward stepwise regression to construct MFS formula. RESULTS MFS was composed of seven variables: age, body mass index, international normalised ratio, aspartate aminotransferase, gamma-glutamyl transpeptidase, platelet count, and history of type 2 diabetes. MFS demonstrated an area under the receiver-operating characteristic curve of 0.848 [95% CI 0.800-898] and 0.823 [0.760-0.886] in training and validation cohorts, significantly higher than aminotransferase-to-platelet ratio index (0.684 [0.603-0.765], 0.663 [0.588-0.738]), Fibrosis-4 index (0.793 [0.735-0.854], 0.737 [0.660-0.814]), and non-alcoholic fatty liver disease fibrosis score (0.785 [0.731-0.844], 0.750 [0.674-0.827]) (DeLong's test p < 0.05). MFS could include 92.3% of patients using dual cut-offs of 14 and 15, with a correct prediction rate of 90.4%, resulting in a larger number of patients with correct diagnosis compared to other scores. A two-step MFS-VCTE screening algorithm demonstrated positive and negative predictive values and overall diagnostic accuracy of 93.4%, 89.5%, and 93.2%, respectively, with only 4.0% of patients classified into grey zone. CONCLUSION MFS outperforms conventional non-invasive scores in predicting advanced fibrosis, contributing to screening in MAFLD patients.
Collapse
Affiliation(s)
- Johnny T K Cheung
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Xinrong Zhang
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Grace Lai-Hung Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Terry Cheuk-Fung Yip
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Huapeng Lin
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Guanlin Li
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Howard Ho-Wai Leung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, China
| | - Jimmy Che-To Lai
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Sanjiv Mahadeva
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Xiao-Dong Wang
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Wen-Yue Liu
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Vincent Wai-Sun Wong
- Medical Data Analytics Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Wah-Kheong Chan
- Gastroenterology and Hepatology Unit, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ming-Hua Zheng
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
- MAFLD Research Centre, Department of Hepatology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
25
|
Lowe KO, Tanase CE, Maghami S, Fisher LE, Ghaemmaghami AM. Inflammatory Network of Liver Fibrosis and How It Can Be Targeted Therapeutically. IMMUNO 2023; 3:375-408. [DOI: 10.3390/immuno3040023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Liver fibrosis is a complex, dynamic process associated with a broad spectrum of chronic liver diseases and acute liver failure, characterised by the dysregulated intrahepatic production of extracellular matrix proteins replacing functional liver cells with scar tissue. Fibrosis progresses due to an interrelated cycle of hepatocellular injury, triggering a persistent wound-healing response. The accumulation of scar tissue and chronic inflammation can eventually lead to cirrhosis and hepatocellular carcinoma. Currently, no therapies exist to directly treat or reverse liver fibrosis; hence, it remains a substantial global disease burden. A better understanding of the intricate inflammatory network that drives the initiation and maintenance of liver fibrosis to enable the rationale design of new intervention strategies is required. This review clarifies the most current understanding of the hepatic fibrosis cellular network with a focus on the role of regulatory T cells, and a possible trajectory for T cell immunotherapy in fibrosis treatment. Despite good progress in elucidating the role of the immune system in liver fibrosis, future work to better define the function of different immune cells and their mediators at different fibrotic stages is needed, which will enhance the development of new therapies.
Collapse
Affiliation(s)
- Kirstin O. Lowe
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK
| | - Leanne E. Fisher
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | |
Collapse
|
26
|
En Li Cho E, Ang CZ, Quek J, Fu CE, Lim LKE, Heng ZEQ, Tan DJH, Lim WH, Yong JN, Zeng R, Chee D, Nah B, Lesmana CRA, Bwa AH, Win KM, Faulkner C, Aboona MB, Lim MC, Syn N, Kulkarni AV, Suzuki H, Takahashi H, Tamaki N, Wijarnpreecha K, Huang DQ, Muthiah M, Ng CH, Loomba R. Global prevalence of non-alcoholic fatty liver disease in type 2 diabetes mellitus: an updated systematic review and meta-analysis. Gut 2023; 72:2138-2148. [PMID: 37491159 DOI: 10.1136/gutjnl-2023-330110] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease, with type 2 diabetes mellitus (T2DM) as a major predictor. Insulin resistance and chronic inflammation are key pathways in the pathogenesis of T2DM leading to NAFLD and vice versa, with the synergistic effect of NAFLD and T2DM increasing morbidity and mortality risks. This meta-analysis aims to quantify the prevalence of NAFLD and the prevalence of clinically significant and advanced fibrosis in people with T2DM. METHODS MEDLINE and Embase databases were searched from inception until 13 February 2023. The primary outcomes were the prevalence of NAFLD, non-alcoholic steatohepatitis (NASH) and fibrosis in people with T2DM. A generalised linear mixed model with Clopper-Pearson intervals was used for the analysis of proportions with sensitivity analysis conducted to explore heterogeneity between studies. RESULTS 156 studies met the inclusion criteria, and a pooled analysis of 1 832 125 patients determined that the prevalence rates of NAFLD and NASH in T2DM were 65.04% (95% CI 61.79% to 68.15%, I2=99.90%) and 31.55% (95% CI 17.12% to 50.70%, I2=97.70%), respectively. 35.54% (95% CI 19.56% to 55.56%, I2=100.00%) of individuals with T2DM with NAFLD had clinically significant fibrosis (F2-F4), while 14.95% (95% CI 11.03% to 19.95%, I2=99.00%) had advanced fibrosis (F3-F4). CONCLUSION This study determined a high prevalence of NAFLD, NASH and fibrosis in people with T2DM. Increased efforts are required to prevent T2DM to combat the rising burden of NAFLD. PROSPERO REGISTRATION NUMBER CRD42022360251.
Collapse
Affiliation(s)
- Elina En Li Cho
- Department of Medicine, National University Hospital, Singapore
| | - Chong Zhe Ang
- Department of Medicine, National University Hospital, Singapore
| | - Jingxuan Quek
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Clarissa Elysia Fu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lincoln Kai En Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zane En Qi Heng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Darren Jun Hao Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wen Hui Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jie Ning Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Rebecca Zeng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Douglas Chee
- Department of Medicine, National University Hospital, Singapore
| | - Benjamin Nah
- Department of Medicine, National University Hospital, Singapore
| | | | - Aung Hlaing Bwa
- Department of Medical Research, Union of Myanmar, Naypyidaw, Myanmar
| | - Khin Maung Win
- Department of Medical Research, Union of Myanmar, Naypyidaw, Myanmar
| | - Claire Faulkner
- Department of Medicine, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Majd B Aboona
- Department of Medicine, University of Arizona College of Medicine, Phoenix, Arizona, USA
| | - Mei Chin Lim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Diagnostic Imaging, National University Health System, Singapore
| | - Nicholas Syn
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anand V Kulkarni
- Hepatology, Asian Institute of Gastroenterology, Hyderabad, Telangana, India
| | - Hiroyuki Suzuki
- Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | | | - Nobuharu Tamaki
- Department of Medicine, University of California San Diego, La Jolla, California, USA
- Department of Medicine, Musashino Red Cross Hospital, Musashino, Japan
| | - Karn Wijarnpreecha
- Division of Gastroenterology and Hepatology, University of Michigan, Michigan, Michigan, USA
| | - Daniel Q Huang
- Department of Medicine, National University Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Gastroenterology and Hepatology, National University Health System, Singapore
| | - Mark Muthiah
- Department of Medicine, National University Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Gastroenterology and Hepatology, National University Health System, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Rohit Loomba
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
27
|
Park SH, Park J, Kwon SY, Lee YB, Kim G, Hur KY, Koh J, Jee JH, Kim JH, Kang M, Jin SM. Increased risk of incident diabetes in patients with MAFLD not meeting the criteria for NAFLD. Sci Rep 2023; 13:10677. [PMID: 37393407 PMCID: PMC10314928 DOI: 10.1038/s41598-023-37858-8] [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: 12/29/2022] [Accepted: 06/28/2023] [Indexed: 07/03/2023] Open
Abstract
We aimed to compare the risk of incident diabetes according to fatty liver disease (FLD) definition, focusing on the comparison between those who met criteria for either metabolic dysfunction-associated fatty liver disease (MAFLD) or nonalcoholic fatty liver disease (NAFLD) but not the other. This was a 5.0-year (interquartile range, 2.4-8.2) retrospective longitudinal cohort study of 21,178 adults who underwent at least two serial health checkup examinations. The presence of hepatic steatosis was determined by abdominal ultrasonography at the first health examination. Cox proportional hazard analyses were used to compare the risk of incident diabetes among five groups. Incident diabetes cases occurred in 1296 participants (6.1%). When non-FLD without metabolic dysfunction (MD) group was set as a reference, the risk of incident diabetes increased in the order of NAFLD-only, non-FLD with MD, both FLD, and MAFLD-only groups. The presence of excessive alcohol consumption and/or hepatitis B virus (HBV)/hepatitis C virus (HCV) infection, FLD, and MD synergistically increased the risk of incident diabetes. MAFLD-only group showed a greater increase in incidence of diabetes than non-FLD with MD and NAFLD-only groups. The interaction among excessive alcohol consumption, HBV/HCV infection, MD, and hepatic steatosis on the development of diabetes should not be overlooked.
Collapse
Affiliation(s)
- So Hee Park
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Jiyun Park
- Division of Endocrine and Metabolism, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam, Gyeonggi-do, 14396, Republic of Korea
| | - So Yoon Kwon
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - You-Bin Lee
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Gyuri Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Kyu Yeon Hur
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Janghyun Koh
- Department of Health Promotion Center, Center for Health Promotion, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Jae Hwan Jee
- Department of Health Promotion Center, Center for Health Promotion, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Jae Hyeon Kim
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea
| | - Mira Kang
- Department of Health Promotion Center, Center for Health Promotion, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
- Department of Digital Health, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| | - Sang-Man Jin
- Division of Endocrinology and Metabolism, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, Republic of Korea.
| |
Collapse
|
28
|
Kongmalai T, Srinonprasert V, Anothaisintawee T, Kongmalai P, McKay G, Attia J, Thakkinstian A. New anti-diabetic agents for the treatment of non-alcoholic fatty liver disease: a systematic review and network meta-analysis of randomized controlled trials. Front Endocrinol (Lausanne) 2023; 14:1182037. [PMID: 37441498 PMCID: PMC10335801 DOI: 10.3389/fendo.2023.1182037] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/14/2023] [Indexed: 07/15/2023] Open
Abstract
Objectives This network meta-analysis aims to compare the efficacy and safety of new anti-diabetic medications for the treatment of non-alcoholic fatty liver disease (NAFLD). Materials and methods PubMed and Scopus were searched from inception to 27th March 2022 to identify all randomized controlled trials (RCTs) in NAFLD patients. Outcomes included reductions in intrahepatic steatosis (IHS) and liver enzyme levels. The efficacy and safety of DPP-4 inhibitors, GLP-1 agonists, SGLT-2 inhibitors, and other therapies were indirectly compared using a NMA approach. Unstandardized mean difference (USMD) with 95% confidence intervals (CI) were calculated. Results 2,252 patients from 31 RCTs were included. "Add-on" GLP-1 agonists with standard of care (SoC) treatment showed significantly reduced IHS compared to SoC alone [USMD (95%CI) -3.93% (-6.54%, -1.33%)]. Surface under the cumulative ranking curve (SUCRA) identified GLP-1 receptor agonists with the highest probability to reduce IHS (SUCRA 88.5%), followed by DPP-4 inhibitors (SUCRA 69.6%) and pioglitazone (SUCRA 62.2%). "Add-on" GLP-1 receptor agonists were also the most effective treatment for reducing liver enzyme levels; AST [USMD of -5.04 (-8.46, -1.62)], ALT [USMD of -9.84 (-16.84, -2.85)] and GGT [USMD of -15.53 (-22.09, -8.97)] compared to SoC alone. However, GLP-1 agonists were most likely to be associated with an adverse event compared to other interventions. Conclusion GLP-1 agonists may represent the most promising anti-diabetic treatment to reduce hepatic steatosis and liver enzyme activity in T2DM and NAFLD patients. Nevertheless, longer-term studies are required to determine whether this delays progression of liver cirrhosis in patients with NAFLD and T2DM. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42021259336.1.
Collapse
Affiliation(s)
- Tanawan Kongmalai
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Varalak Srinonprasert
- Siriraj Health Policy Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Geriatric Medicine, Department of Medicine, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Mahidol University Health Technology Assessment Graduate Program, Mahidol University, Bangkok, Thailand
| | - Thunyarat Anothaisintawee
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pinkawas Kongmalai
- Department of Orthopaedics, Faculty of Medicine, Srinakharinwirot University, Ongkharak, Nakhon Nayok, Thailand
| | - Gareth McKay
- Centre for Public Health, School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University, Belfast, Ireland
| | - John Attia
- School of Medicine and Public Health, University of Newcastle, Callaghan, NSW, Australia
| | - Ammarin Thakkinstian
- Mahidol University Health Technology Assessment Graduate Program, Mahidol University, Bangkok, Thailand
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
29
|
Likitnukul S, Tepaarmorndech S, Kaewamatawong T, Yangchum A, Duangtha C, Jongjang P, Mangmool S, Pinthong D, Isaka M. Pyridylnidulin exerts anti-diabetic properties and improves non-alcoholic fatty liver disease in diet-induced obesity mice. Front Mol Biosci 2023; 10:1208215. [PMID: 37426418 PMCID: PMC10324605 DOI: 10.3389/fmolb.2023.1208215] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/15/2023] [Indexed: 07/11/2023] Open
Abstract
Introduction: Non-alcoholic fatty liver disease (NAFLD) is one of the metabolic disorders related to the pathophysiology of type 2 diabetes mellitus (T2DM). Therapeutic strategies are focused on the improvement of energy balance and lifestyle modification. Additionally, the derivative of the bioactive fungal metabolite is of interest to provide health benefits, especially in obese and pre-diabetic conditions. In our screening of anti-diabetic compounds from fungal metabolites and semisynthetic derivatives, a depsidone derivative, namely pyridylnidulin (PN), showed potent glucose uptake-inducing activity. The present study aimed to investigate the liver lipid metabolism and anti-diabetic properties of PN in diet-induced obesity mice. Methods: Male C57BL/6 mice were induced obesity and pre-diabetic conditions by dietary intervention with a high-fat diet (HFD) for 6 weeks. These obese mice were orally administered with PN (40 or 120 mg/kg), metformin (150 mg/kg), or vehicle for 4 weeks. Glucose tolerance, plasma adipocytokines, hepatic gene and protein expressions were assessed after treatment. Results: Improved glucose tolerance and reduced fasting blood glucose levels were found in the PN and metformin-treated mice. Additionally, hepatic triglyceride levels were consistent with the histopathological steatosis score regarding hepatocellular hypertrophy in the PN and metformin groups. The levels of plasma adipocytokines such as tumor necrosis factor-α (TNF-α) and monocyte chemoattractant protein-1 (MCP-1) were reduced in the PN (120 mg/kg) and metformin-treated mice. In addition, hepatic gene expression involved in lipid metabolism, including lipogenic enzymes was significantly reduced in the PN (120 mg/kg) and metformin-treated mice. The increased protein expression levels of phosphorylated AMP-activated protein kinase (p-AMPK) was also found in PN and metformin-treated mice. Discussion: Considering the increased p-AMPK protein expression levels in PN and metformin-treated mice were revealed as the underlying mechanisms to improve metabolic parameters. These results suggested that PN provided the health benefit to slow the progression of NAFLD and T2DM in obese and pre-diabetic conditions.
Collapse
Affiliation(s)
- Sutharinee Likitnukul
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Theerayuth Kaewamatawong
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Arunrat Yangchum
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Chanathip Duangtha
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Pimrapat Jongjang
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Darawan Pinthong
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Masahiko Isaka
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| |
Collapse
|
30
|
Wiafe YA, Anyitey-Kokor IC, Nmai RA, Afihene M, Roberts LR. Diagnostic Performance of Greyscale Ultrasound in Detecting Fatty Liver Disease in a Type 2 Diabetes Population Using FibroScan as the Reference Standard. Cureus 2023; 15:e40756. [PMID: 37350981 PMCID: PMC10284594 DOI: 10.7759/cureus.40756] [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] [Accepted: 06/21/2023] [Indexed: 06/24/2023] Open
Abstract
Introduction Brightness mode ultrasound (B-mode US) and FibroScan (Echosens, Paris, France) are the two ultrasound methods often recommended for screening non-alcoholic fatty liver disease (NAFLD) in persons with type 2 diabetes mellitus (T2DM). This study assessed the diagnostic performance of B-mode US using FibroScan as the reference standard. Methods Persons with a known history of T2DM were invited to screen for NAFLD using B-mode US and FibroScan on separate days within a one-month period. Assessors of B-mode US and FibroScan were blinded to each other's findings. Both B-mode US and FibroScan independently assessed and graded each participant for the presence of NAFLD. Using the diagnostic test findings of FibroScan as a reference standard, the sensitivity and specificity of B-mode US were analyzed. The area under the receiver operating characteristic curve (AUROC) was analyzed using Jamovi (version 2.3.21). A multinomial logistic regression of the B-mode US and FibroScan in predicting NAFLD grade was also analyzed. Results A total of 171 participants were assessed. B-mode US detected NAFLD in T2DM patients with 63.6% sensitivity, 65.6% specificity, and 0.646 AUROC. Sensitivity and specificity in overweight and obese participants were 36-43% and 76-85%, respectively. Multinomial logistic regression demonstrated an insignificant statistical relationship between FibroScan and B-mode US in predicting grade 1 steatosis (p-value = 0.397), which was significantly affected by a higher BMI (p-value = 0.034) rather than a higher liver fibrosis level (p-value = 0.941). The logistic regression further showed a significant relationship between B-mode US and FibroScan in predicting steatosis grade 2 (p-value = 0.045) and grade 3 (p-value < 0.001), which was not significantly affected by BMI (p-value = 0.091). Conclusion B-mode US can replace FibroScan for severe steatosis; however, it cannot be used to screen for NAFLD in T2DM patients due to lower sensitivity for early detection in the overweight.
Collapse
Affiliation(s)
- Yaw A Wiafe
- Department of Medical Diagnostics, Kwame Nkrumah University of Science and Technology, Kumasi, GHA
| | - Ijeoma C Anyitey-Kokor
- Department of Medical Diagnostics, Kwame Nkrumah University of Science and Technology, Kumasi, GHA
| | - Richmond A Nmai
- Department of Medical Diagnostics, Kwame Nkrumah University of Science and Technology, Kumasi, GHA
| | - Mary Afihene
- School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, GHA
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, USA
| |
Collapse
|
31
|
Wiafe YA, Afihene MY, Anto EO, Nmai RA, Amoah-Kumi L, Frimpong J, Dickson FD, Antwi SO, Roberts LR. Non-Alcoholic Fatty Liver Disease and Liver Fibrosis in Persons with Type 2 Diabetes Mellitus in Ghana: A Study of Prevalence, Severity, and Contributing Factors Using Transient Elastography. J Clin Med 2023; 12:3741. [PMID: 37297935 PMCID: PMC10253760 DOI: 10.3390/jcm12113741] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder characterized by hyperglycemia, insulin resistance, and pancreatic islet cell dysfunction. T2DM is associated with non-alcoholic fatty liver disease (NAFLD) because of impaired glucose metabolism in both conditions. However, it is widely assumed that people with T2DM in sub-Saharan Africa (SSA) have a lower prevalence of NAFLD than in other parts of the world. With our recent access to transient elastography, we aimed to investigate the prevalence of, severity of, and contributing factors to NAFLD in persons with T2DM in Ghana. We performed a cross-sectional study recruiting 218 individuals with T2DM at the Kwadaso Seventh-Day Adventist and Mount Sinai Hospitals in the Ashanti region of Ghana using a simple randomized sampling technique. A structured questionnaire was used to obtain socio-demographic information, clinical history, exercise and other lifestyle factors, and anthropometric measurements. Transient elastography was performed using a FibroScan® machine to obtain the Controlled Attenuation Parameter (CAP) score and liver fibrosis score. The prevalence of NAFLD among Ghanaian T2DM participants was 51.4% (112/218), of whom 11.6% had significant liver fibrosis. An evaluation of the NAFLD group (n = 112) versus the non-NAFLD group (n = 106) revealed a higher BMI (28.7 vs. 25.2 kg/m2, p = 0.001), waist circumference (106.0 vs. 98.0 cm, p = 0.001), hip circumference (107.0 vs. 100.5 cm, p = 0.003), and waist-to-height ratio (0.66 vs. 0.62, p = 0.001) in T2DM patients with NAFLD compared to those without NAFLD. Being obese was an independent predictor of NAFLD in persons with T2DM than known history of hypertension and dyslipidaemia.
Collapse
Affiliation(s)
- Yaw Amo Wiafe
- Department of Medical Diagnostics, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana; (E.O.A.); (R.A.N.); (L.A.-K.); (J.F.)
| | - Mary Yeboah Afihene
- Department of Medicine, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana;
| | - Enoch Odame Anto
- Department of Medical Diagnostics, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana; (E.O.A.); (R.A.N.); (L.A.-K.); (J.F.)
| | - Richmond Ashitey Nmai
- Department of Medical Diagnostics, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana; (E.O.A.); (R.A.N.); (L.A.-K.); (J.F.)
| | - Lois Amoah-Kumi
- Department of Medical Diagnostics, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana; (E.O.A.); (R.A.N.); (L.A.-K.); (J.F.)
| | - Joseph Frimpong
- Department of Medical Diagnostics, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana; (E.O.A.); (R.A.N.); (L.A.-K.); (J.F.)
| | | | - Samuel O. Antwi
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic College of Medicine and Science, Jacksonville, FL 32224, USA;
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA;
| |
Collapse
|
32
|
Chen C, Zhang Y, Fan Y, Ying Z, Su Q, Li X, Qin L. The change of non-alcoholic fatty liver disease is associated with risk of incident diabetes. Front Endocrinol (Lausanne) 2023; 14:1108442. [PMID: 37214244 PMCID: PMC10194027 DOI: 10.3389/fendo.2023.1108442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 04/05/2023] [Indexed: 05/24/2023] Open
Abstract
Background & aims The effect of change in non-alcoholic fatty liver disease (NAFLD) status on incident diabetes has not been well studied. We aimed to investigate the association of NAFLD development and remission with the risk of incident diabetes during a median of 3.5-year follow-up. Methods A total of 2690 participants without diabetes were recruited in 2011-2012 and assessed for incident diabetes in 2014. Abdominal ultrasonography was used to determine the change of NAFLD. 75 g oral glucose tolerance test (OGTT) was performed to determine diabetes. NAFLD severity was assessed using Gholam's model. The odds ratios (ORs) for incident diabetes were estimated by logistic regression models. Results NAFLD was developed in 580 (33.2%) participants and NAFLD remission occurred in 150 (15.9%) participants during a median of 3.5-year follow-up. A total of 484 participants developed diabetes during follow-up, including 170 (14.6%) in consistent non-NAFLD group, 111 (19.1%) in NAFLD developed group, 19 (12.7%) in NAFLD remission group, and 184 (23.2%) in sustained NAFLD group. The development of NAFLD increased the risk of incident diabetes by 43% (OR, 1.43; 95%CI, 1.10-1.86) after adjustment for multiple confounders. Compared with sustained NAFLD group, remission of NAFLD reduced the risk of incident diabetes by 52% (OR, 0.48; 95%CI, 0.29-0.80). The effect of NAFLD alteration on incident diabetes was not changed after adjustment for body mass index or waist circumference, change of body mass index or waist circumference. In NAFLD remission group, participants with non-alcoholic steatohepatitis (NASH) at baseline were more likely to develop diabetes (OR, 3.03; 95%CI, 1.01-9.12). Conclusions NAFLD development increases the risk of incident diabetes, whereas NAFLD remission reduces the risk of incident diabetes. Moreover, presence of NASH at baseline could attenuate the protective effect of NAFLD remission on incident diabetes. Our study suggests that early intervention of NAFLD and maintenance of non-NAFLD are important for prevention of diabetes.
Collapse
Affiliation(s)
- Congling Chen
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuecheng Zhang
- General Practice Department, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, China
| | - Yujuan Fan
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhen Ying
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Su
- Department of Endocrinology and Metabolism, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoying Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Qin
- Department of Endocrinology and Metabolism, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Endocrinology, Chongming Hospital Affiliated to Shanghai University of Health & Medicine Sciences, Shanghai, China
| |
Collapse
|
33
|
Ganz MJ, Bose K, Herzog C, Bender S, Mertens PR, Scurt FG. Pathomechanismen der chronischen Nierenschädigung bei Diabetes und anderen Begleiterkrankungen. DIE DIABETOLOGIE 2023; 19:251-261. [DOI: 10.1007/s11428-023-01020-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/15/2023] [Indexed: 01/03/2025]
|
34
|
Lee DS, An TH, Kim H, Jung E, Kim G, Oh SY, Kim JS, Chun HJ, Jung J, Lee EW, Han BS, Han DH, Lee YH, Han TS, Hur K, Lee CH, Kim DS, Kim WK, Park JW, Koo SH, Seong JK, Lee SC, Kim H, Bae KH, Oh KJ. Tcf7l2 in hepatocytes regulates de novo lipogenesis in diet-induced non-alcoholic fatty liver disease in mice. Diabetologia 2023; 66:931-954. [PMID: 36759348 PMCID: PMC10036287 DOI: 10.1007/s00125-023-05878-8] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/28/2022] [Indexed: 02/11/2023]
Abstract
AIMS/HYPOTHESIS Non-alcoholic fatty liver disease (NAFLD) associated with type 2 diabetes may more easily progress towards severe forms of non-alcoholic steatohepatitis (NASH) and cirrhosis. Although the Wnt effector transcription factor 7-like 2 (TCF7L2) is closely associated with type 2 diabetes risk, the role of TCF7L2 in NAFLD development remains unclear. Here, we investigated how changes in TCF7L2 expression in the liver affects hepatic lipid metabolism based on the major risk factors of NAFLD development. METHODS Tcf7l2 was selectively ablated in the liver of C57BL/6N mice by inducing the albumin (Alb) promoter to recombine Tcf7l2 alleles floxed at exon 5 (liver-specific Tcf7l2-knockout [KO] mice: Alb-Cre;Tcf7l2f/f). Alb-Cre;Tcf7l2f/f and their wild-type (Tcf7l2f/f) littermates were fed a high-fat diet (HFD) or a high-carbohydrate diet (HCD) for 22 weeks to reproduce NAFLD/NASH. Mice were refed a standard chow diet or an HCD to stimulate de novo lipogenesis (DNL) or fed an HFD to provide exogenous fatty acids. We analysed glucose and insulin sensitivity, metabolic respiration, mRNA expression profiles, hepatic triglyceride (TG), hepatic DNL, selected hepatic metabolites, selected plasma metabolites and liver histology. RESULTS Alb-Cre;Tcf7l2f/f essentially exhibited increased lipogenic genes, but there were no changes in hepatic lipid content in mice fed a normal chow diet. However, following 22 weeks of diet-induced NAFLD/NASH conditions, liver steatosis was exacerbated owing to preferential metabolism of carbohydrate over fat. Indeed, hepatic Tcf7l2 deficiency enhanced liver lipid content in a manner that was dependent on the duration and amount of exposure to carbohydrates, owing to cell-autonomous increases in hepatic DNL. Mechanistically, TCF7L2 regulated the transcriptional activity of Mlxipl (also known as ChREBP) by modulating O-GlcNAcylation and protein content of carbohydrate response element binding protein (ChREBP), and targeted Srebf1 (also called SREBP1) via miRNA (miR)-33-5p in hepatocytes. Eventually, restoring TCF7L2 expression at the physiological level in the liver of Alb-Cre;Tcf7l2f/f mice alleviated liver steatosis without altering body composition under both acute and chronic HCD conditions. CONCLUSIONS/INTERPRETATION In mice, loss of hepatic Tcf7l2 contributes to liver steatosis by inducing preferential metabolism of carbohydrates via DNL activation. Therefore, TCF7L2 could be a promising regulator of the NAFLD associated with high-carbohydrate diets and diabetes since TCF7L2 deficiency may lead to development of NAFLD by promoting utilisation of excess glucose pools through activating DNL. DATA AVAILABILITY RNA-sequencing data have been deposited into the NCBI GEO under the accession number GSE162449 ( www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE162449 ).
Collapse
Affiliation(s)
- Da Som Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Tae Hyeon An
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hyunmi Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Eunsun Jung
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Gyeonghun Kim
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Seung Yeon Oh
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, Republic of Korea
| | - Jun Seok Kim
- Division of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Hye Jin Chun
- Department of Systems Biology, Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
| | - Jaeeun Jung
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Baek-Soo Han
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Dai Hoon Han
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong-Ho Lee
- Department of Systems Biology, Glycosylation Network Research Center, Yonsei University, Seoul, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Su Han
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Keun Hur
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chul-Ho Lee
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Dae-Soo Kim
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jun Won Park
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, ChunCheon-si, Gangwon-do, Republic of Korea
| | - Seung-Hoi Koo
- Division of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Je Kyung Seong
- College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, Republic of Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hail Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea.
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon, Republic of Korea.
| |
Collapse
|
35
|
Barton JC, Barton JC, Acton RT. Non-alcoholic fatty liver disease in hemochromatosis probands with iron overload and HFE p.C282Y/p.C282Y. BMC Gastroenterol 2023; 23:137. [PMID: 37118679 PMCID: PMC10148383 DOI: 10.1186/s12876-023-02763-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/11/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND The aim of this study was to identify characteristics of non-alcoholic fatty liver disease (NAFLD) in adults with HFE p.C282Y/p.C282Y. METHODS We retrospectively studied non-Hispanic white hemochromatosis probands with iron overload (serum ferritin (SF) > 300 µg/L (M), > 200 µg/L (F)) and p.C282Y/p.C282Y at non-screening diagnosis who did not report alcohol consumption > 14 g/d, have cirrhosis or other non-NAFLD liver disorders, use steatogenic medication, or have diagnoses of heritable disorders that increase NAFLD risk. We identified NAFLD-associated characteristics using univariate and multivariable analyses. RESULTS There were 66 probands (31 men, 35 women), mean age 49 ± 14 (SD) y, of whom 16 (24.2%) had NAFLD. The following characteristics were higher in probands with NAFLD: median SF (1118 µg/L (range 259, 2663) vs. 567 µg/L (247, 2385); p = 0.0192); prevalence of elevated ALT/AST (alanine/aspartate aminotransferase) (43.8% vs. 10.0%; p = 0.0056); and prevalence of type 2 diabetes (T2DM) (31.3% vs. 10.0%; p = 0.0427). Mean age, sex, and prevalences of human leukocyte antigen-A*03 positivity, body mass index ≥ 30.0 kg/m2, hyperlipidemia, hypertension, and metabolic syndrome in probands with/without NAFLD did not differ significantly. Logistic regression on NAFLD using variables SF, elevated ALT/AST, and T2DM revealed: SF (p = 0.0318; odds ratio 1.0-1.0) and T2DM (p = 0.0342; 1.1-22.3). Median iron removed to achieve iron depletion (QFe) in probands with/without NAFLD did not differ significantly (3.6 g (1.4-7.2 g) vs. 2.8 g (0.7-11.0 g), respectively; p = 0.6862). CONCLUSIONS NAFLD in hemochromatosis probands with p.C282Y/p.C282Y is associated with higher median SF and greater T2DM prevalence, after adjustment for other factors. NAFLD does not influence QFe significantly.
Collapse
Affiliation(s)
- James C Barton
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
- Southern Iron Disorders Center, Birmingham, AL, USA.
| | | | - Ronald T Acton
- Southern Iron Disorders Center, Birmingham, AL, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
36
|
Cazac GD, Lăcătușu CM, Ștefănescu G, Mihai C, Grigorescu ED, Onofriescu A, Mihai BM. Glucagon-like Peptide-1 Receptor Agonists in Patients with Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease-Current Background, Hopes, and Perspectives. Metabolites 2023; 13:metabo13050581. [PMID: 37233622 DOI: 10.3390/metabo13050581] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents the most common chronic liver disease worldwide, reaching one of the highest prevalences in patients with type 2 diabetes mellitus (T2DM). For now, no specific pharmacologic therapies are approved to prevent or treat NAFLD. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are currently evaluated as potential candidates for NAFLD treatment in patients with T2DM. Some representatives of this class of antihyperglycemic agents emerged as potentially beneficial in patients with NAFLD after several research studies suggested they reduce hepatic steatosis, ameliorate lesions of nonalcoholic steatohepatitis (NASH), or delay the progression of fibrosis in this population. The aim of this review is to summarize the body of evidence supporting the effectiveness of GLP-1RA therapy in the management of T2DM complicated with NAFLD, describing the studies that evaluated the effects of these glucose-lowering agents in fatty liver disease and fibrosis, their possible mechanistic justification, current evidence-based recommendations, and the next steps to be developed in the field of pharmacological innovation.
Collapse
Affiliation(s)
- Georgiana-Diana Cazac
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Cristina-Mihaela Lăcătușu
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Gabriela Ștefănescu
- Unit of Medical Semiology and Gastroenterology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Gastroenterology and Hepatology, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Cătălina Mihai
- Unit of Medical Semiology and Gastroenterology, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Gastroenterology and Hepatology, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Elena-Daniela Grigorescu
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alina Onofriescu
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Bogdan-Mircea Mihai
- Unit of Diabetes, Nutrition, and Metabolic Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, "Sf. Spiridon" County Clinical Emergency Hospital, 700111 Iași, Romania
| |
Collapse
|
37
|
Bays HE, Bindlish S, Clayton TL. Obesity, diabetes mellitus, and cardiometabolic risk: An Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) 2023. OBESITY PILLARS 2023; 5:100056. [PMID: 37990743 PMCID: PMC10661981 DOI: 10.1016/j.obpill.2023.100056] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 11/23/2023]
Abstract
Background This Obesity Medicine Association (OMA) Clinical Practice Statement (CPS) is intended to provide clinicians an overview of type 2 diabetes mellitus (T2DM), an obesity-related cardiometabolic risk factor. Methods The scientific support for this CPS is based upon published citations and clinical perspectives of OMA authors. Results Topics include T2DM and obesity as cardiometabolic risk factors, definitions of obesity and adiposopathy, and mechanisms for how obesity causes insulin resistance and beta cell dysfunction. Adipose tissue is an active immune and endocrine organ, whose adiposopathic obesity-mediated dysfunction contributes to metabolic abnormalities often encountered in clinical practice, including hyperglycemia (e.g., pre-diabetes mellitus and T2DM). The determination as to whether adiposopathy ultimately leads to clinical metabolic disease depends on crosstalk interactions and biometabolic responses of non-adipose tissue organs such as liver, muscle, pancreas, kidney, and brain. Conclusions This review is intended to assist clinicians in the care of patients with the disease of obesity and T2DM. This CPS provides a simplified overview of how obesity may cause insulin resistance, pre-diabetes, and T2DM. It also provides an algorithmic approach towards treatment of a patient with obesity and T2DM, with "treat obesity first" as a priority. Finally, treatment of obesity and T2DM might best focus upon therapies that not only improve the weight of patients, but also improve the health outcomes of patients (e.g., cardiovascular disease and cancer).
Collapse
Affiliation(s)
- Harold Edward Bays
- Louisville Metabolic and Atherosclerosis Research Center, University of Louisville School of Medicine, 3288 Illinois Avenue, Louisville, KY, 40213, USA
| | - Shagun Bindlish
- Diabetology, One Medical, Adjunct Faculty Touro University, CA, USA
| | | |
Collapse
|
38
|
Shaheen M, Schrode KM, Tedlos M, Pan D, Najjar SM, Friedman TC. Racial/ethnic and gender disparity in the severity of NAFLD among people with diabetes or prediabetes. Front Physiol 2023; 14:1076730. [PMID: 36891143 PMCID: PMC9986441 DOI: 10.3389/fphys.2023.1076730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/03/2023] [Indexed: 02/22/2023] Open
Abstract
Aim: Non-alcoholic fatty liver disease (NAFLD) exhibits a racial disparity. We examined the prevalence and the association between race, gender, and NAFLD among prediabetes and diabetes populations among adults in the United States. Methods: We analyzed data for 3,190 individuals ≥18 years old from the National Health and Nutrition Examination Survey (NHANES) 2017-2018. NAFLD was diagnosed by FibroScan® using controlled attenuation parameter (CAP) values: S0 (none) < 238, S1 (mild) = 238-259, S2 (moderate) = 260-290, S3 (severe) > 290. Data were analyzed using Chi-square test and multinomial logistic regression, adjusting for confounding variables and considering the design and sample weights. Results: Of the 3,190 subjects, the prevalence of NAFLD was 82.6%, 56.4%, and 30.5% (p < 0.0001) among diabetes, prediabetes and normoglycemia populations respectively. Mexican American males with prediabetes or diabetes had the highest prevalence of severe NAFLD relative to other racial/ethnic groups (p < 0.05). In the adjusted model, among the total, prediabetes, and diabetes populations, a one unit increase in HbA1c was associated with higher odds of severe NAFLD [adjusted odds ratio (AOR) = 1.8, 95% confidence level (CI) = 1.4-2.3, p < 0.0001; AOR = 2.2, 95% CI = 1.1-4.4, p = 0.033; and AOR = 1.5, 95% CI = 1.1-1.9, p = 0.003 respectively]. Conclusion: We found that prediabetes and diabetes populations had a high prevalence and higher odds of NAFLD relative to the normoglycemic population and HbA1c is an independent predictor of NAFLD severity in prediabetes and diabetes populations. Healthcare providers should screen prediabetes and diabetes populations for early detection of NAFLD and initiate treatments including lifestyle modification to prevent the progression to non-alcoholic steatohepatitis or liver cancer.
Collapse
Affiliation(s)
- Magda Shaheen
- Charles R. Drew University, Los Angeles, CA, United States
| | | | | | - Deyu Pan
- Charles R. Drew University, Los Angeles, CA, United States
| | - Sonia M. Najjar
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | | |
Collapse
|
39
|
Lopez-Pentecost M, Hallmark B, Thomson CA, Chilton F, Garcia DO. Association between Dietary Fatty Acid Intake and Liver Steatosis and Fibrosis in a Sample of Mexican-Origin Hispanic Adults with Overweight or Obesity. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3103. [PMID: 36833798 PMCID: PMC9960945 DOI: 10.3390/ijerph20043103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 06/18/2023]
Abstract
Rates of non-alcoholic fatty liver disease (NAFLD) vary dramatically among Hispanic subpopulations, with Mexican-origin (MO) Hispanics experiencing a disproportionate burden. This study examined dietary fatty acid (FA) intake among overweight and obese MO Hispanic adults in the United States (US) and evaluated its association with liver steatosis and fibrosis. Participants (N = 285, MO Hispanic adults) completed 24-h dietary recalls to assess dietary FA exposure. Liver steatosis and fibrosis were estimated using transient elastography (FibroScan®). Multiple regression analysis tested relationships between FA intakes and liver steatosis or fibrosis, adjusting for age, sex, body mass index (BMI) and total energy. A total of 51% (n = 145) of participants were suspected to have NAFLD and 20% self-reported a type 2 diabetes diagnosis. No significant association was observed between Linoleic Acid and α-Linolenic Acid (LA:ALA) ratio, or omega-6 to omega-3 (n-6:n-3) ratio and liver steatosis. However, a one-point increase in the LA:ALA ratio resulted in a 1.01% increase in the liver fibrosis scores (95% CI: [1.00, 1.03]; p = 0.03), and a one-point increase in the n-6:n-3 ratio resulted in a 1.02% increase in liver fibrosis score (95% CI: [1.01, 1.03]; p = 0.01). Further research is needed to determine if modulation of FA intake could reduce NAFLD risk in this high-risk population.
Collapse
Affiliation(s)
- Melissa Lopez-Pentecost
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Brian Hallmark
- The BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
| | - Cynthia A. Thomson
- Department of Health Promotion Sciences Tucson, University of Arizona, Tucson, AZ 85721, USA
| | - Floyd Chilton
- The BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - David O. Garcia
- Department of Health Promotion Sciences Tucson, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
40
|
Non-alcoholic Fatty Liver Disease (NAFLD), Type 2 Diabetes, and Non-viral Hepatocarcinoma: Pathophysiological Mechanisms and New Therapeutic Strategies. Biomedicines 2023; 11:biomedicines11020468. [PMID: 36831004 PMCID: PMC9953066 DOI: 10.3390/biomedicines11020468] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
In recent years, the incidence of non-viral hepatocellular carcinoma (HCC) has increased dramatically, which is probably related to the increased prevalence of metabolic syndrome, together with obesity and type 2 diabetes mellitus (T2DM). Several epidemiological studies have established the association between T2DM and the incidence of HCC and have demonstrated the role of diabetes mellitus as an independent risk factor for the development of HCC. The pathophysiological mechanisms underlying the development of Non-alcoholic fatty liver disease (NAFLD) and its progression to Non-alcoholic steatohepatitis (NASH) and cirrhosis are various and involve pro-inflammatory agents, oxidative stress, apoptosis, adipokines, JNK-1 activation, increased IGF-1 activity, immunomodulation, and alteration of the gut microbiota. Moreover, these mechanisms are thought to play a significant role in the development of NAFLD-related hepatocellular carcinoma. Early diagnosis and the timely correction of risk factors are essential to prevent the onset of liver fibrosis and HCC. The purpose of this review is to summarize the current evidence on the association among obesity, NASH/NAFLD, T2DM, and HCC, with an emphasis on clinical impact. In addition, we will examine the main mechanisms underlying this complex relationship, and the promising strategies that have recently emerged for these diseases' treatments.
Collapse
|
41
|
Thymiakou E, Tzardi M, Kardassis D. Impaired hepatic glucose metabolism and liver-α-cell axis in mice with liver-specific ablation of the Hepatocyte Nuclear Factor 4α (Hnf4a) gene. Metabolism 2023; 139:155371. [PMID: 36464036 DOI: 10.1016/j.metabol.2022.155371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Hnf4a gene ablation in mouse liver causes hepatic steatosis, perturbs HDL structure and function and affects many pathways and genes related to glucose metabolism. Our aim here was to investigate the role of liver HNF4A in glucose homeostasis. METHODS Serum and tissue samples were obtained from Alb-Cre;Hnf4afl/fl (H4LivKO) mice and their littermate Hnf4afl/fl controls. Fasting glucose and insulin, glucose tolerance, insulin tolerance and glucagon challenge tests were performed by standard procedures. Binding of HNF4A to DNA was assessed by chromatin immunoprecipitation assays. Gene expression analysis was performed by quantitative reverse transcription PCR. RESULTS H4LivKO mice presented lower blood levels of fasting glucose, improved glucose tolerance, increased serum lactate levels and reduced response to glucagon challenge compared to their control littermates. Insulin signaling in the liver was reduced despite the increase in serum insulin levels. H4LivKO mice showed altered expression of genes involved in glycolysis, gluconeogenesis and glycogen metabolism in the liver. The expression of the gene encoding the glucagon receptor (Gcgr) was markedly reduced in H4LivKO liver and chromatin immunoprecipitation assays revealed specific and strong binding of HNF4A to the Gcgr promoter. H4LivKO mice presented increased amino acid concentration in the serum, α-cell hyperplasia and a dramatic increase in glucagon levels suggesting an impairment of the liver-α-cell axis. Glucose administration in the drinking water of H4LivKO mice resulted in an impressive extension of survival. The expression of several genes related to non-alcoholic fatty liver disease progression to more severe liver pathologies, including Mcp1, Gdf15, Igfbp-1 and Hmox1, was increased in H4LivKO mice as early as 6 weeks of age and this increased expression was sustained until the endpoint of the study. CONCLUSIONS Our results reveal a novel role of liver HNF4A in controlling blood glucose levels via regulation of glucagon signaling. In combination with the steatotic phenotype, our results suggest that H4LivKO mice could serve as a valuable model for studying glucose homeostasis in the context of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Efstathia Thymiakou
- Laboratory of Biochemistry, University of Crete Medical School, Heraklion 71003, Greece; Gene Regulation and Epigenetics group, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion 71003, Greece
| | - Maria Tzardi
- Department of Pathology, University of Crete Medical School, Heraklion, Crete, Greece
| | - Dimitris Kardassis
- Laboratory of Biochemistry, University of Crete Medical School, Heraklion 71003, Greece; Gene Regulation and Epigenetics group, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology of Hellas, Heraklion 71003, Greece.
| |
Collapse
|
42
|
Zhu W, Shi P, Fu J, Liang A, Zheng T, Wu X, Yuan S. Development and application of a novel model to predict the risk of non-alcoholic fatty liver disease among lean pre-diabetics with normal blood lipid levels. Lipids Health Dis 2022; 21:149. [PMID: 36585668 PMCID: PMC9804963 DOI: 10.1186/s12944-022-01752-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 12/07/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has been associated with type 2 diabetes, but its relationship with pre-diabetes is still unknown. This study aims to determine whether pre-diabetes is associated with NAFLD, followed by establishing a NAFLD predictive nomogram for lean Chinese pre-diabetics with normal blood lipids. METHODS Datasets from 3 previous studies, 1 (2774 pre-diabetics with normal blood lipids for training, 925 for validation), 2 (546 for longitudinal internal validation, post-5-year follow-up), and 3 (501 from another institution for external validation), were used. Kaplan-Meier determined cumulative NAFLD hazard, and least absolute shrinkage and selection operator regression analysis uncovered its risk factors. Multivariate logistic regression analysis constructed the nomogram, followed by validation with receiver operating characteristic curve, calibration plot, and decision curve analyses. RESULTS NAFLD incidence increased with diabetes progression, and pre-diabetics had higher cumulative risk versus non-diabetics, even for lean individuals with normal blood lipids. Six risk factors were identified: body mass index, total cholesterol, alanine aminotransferase:aspartate aminotransferase, triglyceride:high density lipoprotein cholesterol, fasting blood glucose and γ-glutamyl-transferase. The nomogram yielded areas under the curve of 0.808, 0.785, 0.796 and 0.832, for respectively, training, validation, longitudinal internal validation, and external validation, which, along with calibration curve values of p = 0.794, 0.875, 0.854 and 0.810 for those 4 datasets and decision curve analyses, validated its clinical utility. CONCLUSIONS Lean pre-diabetic Chinese with normal blood lipids have higher NAFLD risk versus non-diabetics. The nomogram is able to predict NAFLD among such individuals, with high discrimination, enabling its use for early detection and intervention.
Collapse
Affiliation(s)
- Wentao Zhu
- grid.412604.50000 0004 1758 4073Department of Infectious Diseases, the First Affiliated Hospital of Nanchang University, No. 17 Yongwai Street, Donghu District, Nanchang, China
| | - Pei Shi
- grid.412604.50000 0004 1758 4073Department of Infectious Diseases, the First Affiliated Hospital of Nanchang University, No. 17 Yongwai Street, Donghu District, Nanchang, China
| | - Jiwei Fu
- grid.412604.50000 0004 1758 4073Department of Infectious Diseases, the First Affiliated Hospital of Nanchang University, No. 17 Yongwai Street, Donghu District, Nanchang, China
| | - An Liang
- grid.412604.50000 0004 1758 4073Department of Infectious Diseases, the First Affiliated Hospital of Nanchang University, No. 17 Yongwai Street, Donghu District, Nanchang, China
| | - Ting Zheng
- grid.412604.50000 0004 1758 4073Department of Infectious Diseases, the First Affiliated Hospital of Nanchang University, No. 17 Yongwai Street, Donghu District, Nanchang, China
| | - Xiaoping Wu
- grid.412604.50000 0004 1758 4073Department of Infectious Diseases, the First Affiliated Hospital of Nanchang University, No. 17 Yongwai Street, Donghu District, Nanchang, China
| | - Songsong Yuan
- grid.412604.50000 0004 1758 4073Department of Infectious Diseases, the First Affiliated Hospital of Nanchang University, No. 17 Yongwai Street, Donghu District, Nanchang, China
| |
Collapse
|
43
|
Alfadda NA, Aljuraiban GS, Awwad HM, Khaleel MS, Almaghamsi AM, Sherbeeni SM, Alqutub AN, Aldosary AS, Alfadda AA. Higher carbohydrate intake in relation to non-alcoholic fatty liver disease in patients with type 2 diabetes. Front Nutr 2022; 9:996004. [PMID: 36570126 PMCID: PMC9773196 DOI: 10.3389/fnut.2022.996004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) is an overlooked complication of type 2 diabetes (T2D). Current recommendations for the management of NAFLD are mainly focused on weight reduction, overlooking the role of macronutrient composition. Although dietary carbohydrates play a major role in intrahepatic fat synthesis, their association with the progression of liver steatosis has not been fully investigated in patients with T2D. Aim To investigate the association between higher carbohydrate intake and the presence of liver steatosis in patients with T2D. Methods This cross-sectional study included men and women aged 18-60 years diagnosed with T2D. Anthropometric measurements, hepatic steatosis assessment using the controlled attenuation parameter (CAP), blood samples, and dietary data were analyzed. Participants were divided into two groups: NAFLD and NAFLD-free. A two-sample t-test was used to evaluate the differences between the two groups. Stepwise multiple linear regression models adjusted for potential confounders were used to determine the association between CAP values and higher carbohydrate intake. Results In total, 358 participants were included. NAFLD was present in 79.3% of the participants. Body mass index, waist circumference, ALT, HbA1c, and triglycerides showed direct, while HDL-Cholesterol revealed inverse associations with CAP values. No significant relationship was found between carbohydrate intake and steatosis in the total study sample; however, multiple linear regression analysis revealed a significant relationship between carbohydrate intake and CAP values in patients aged ≤50 years. Conclusion In patients with T2D, higher carbohydrate intake was associated with liver steatosis in those aged 50 years and below. Further studies are required to confirm the causality between carbohydrate intake and liver steatosis.
Collapse
Affiliation(s)
- Nora A. Alfadda
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ghadeer S. Aljuraiban
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Hadeel M. Awwad
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad S. Khaleel
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | | | - Adel N. Alqutub
- Department of Gastroenterology and Hepatology, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Abdullah S. Aldosary
- Department of Medical Imaging Administration, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Assim A. Alfadda
- Obesity Research Center, College of Medicine, King Saud University, Riyadh, Saudi Arabia,Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia,*Correspondence: Assim A. Alfadda,
| |
Collapse
|
44
|
Liu Q, Zhao G, Li Q, Wu W, Zhang Y, Bian H. A comparison of NAFLD and MAFLD diagnostic criteria in contemporary urban healthy adults in China: a cross-sectional study. BMC Gastroenterol 2022; 22:471. [PMID: 36402947 PMCID: PMC9675196 DOI: 10.1186/s12876-022-02576-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022] Open
Abstract
Background A recently proposed diagnostic criteria of metabolic dysfunction-associated fatty liver disease (MAFLD) is more available for various clinical situations than nonalcoholic fatty liver disease (NAFLD), but understanding about differences between NAFLD and MAFLD in clinical practice remains limited in the general adult urban population in China. Methods A total of 795 subjects were recruited from Wu Song Branch of Zhongshan Hospital who participated in the general health assessment. Examination results was obtained through analysis of blood samples and abdominal ultrasonography. Participants were divided into four subgroups according to whether they had NAFLD or MAFLD (NAFLD- MAFLD-, NAFLD + MAFLD-, NAFLD- MAFLD + and NAFLD + MAFLD+). Results Among the urban healthy adults investigated, 345 people (43.4%) were diagnosed with NAFLD and 356 people (44.8%) with MAFLD. No significant differences in the prevalence, age, fasting blood glucose, glycosylated hemoglobin, liver enzyme examination, percentage of overweight, hypertension or dyslipidaemia were found between NAFLD and MAFLD patients. Patients with MAFLD had worse metabolic disorders than NAFLD + MAFLD- patients. The NAFLD fibrosis score (NFS) of the NAFLD- MAFLD + group was higher than that of the NAFLD + MAFLD- group. Higher proportion of patients in the NAFLD- MAFLD + group have NFS ≥-1.455. Conclusion MAFLD criteria have similar prevalence and patient characteristics compared with previous NAFLD but help to identify a group of patients with high risks of metabolic disorders and liver fibrosis who have been missed with NAFLD, and has superior utility.
Collapse
|
45
|
Shedding light on non-alcoholic fatty liver disease: Pathogenesis, molecular mechanisms, models, and emerging therapeutics. Life Sci 2022; 312:121185. [PMID: 36375569 DOI: 10.1016/j.lfs.2022.121185] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disorder globally impacting an estimated 25% of the population associated with severe consequences such as cirrhosis, hepatocellular carcinoma (HCC), and overall mortality. Fatty liver disease is triggered through multiple pathways, but the most prominent cause is either diabetes or obesity, or a combination of both. Therefore, hepatic glucose, insulin and fatty acid signaling becomes a dire need to understand which is well elaborated in this review. This review summarizes the popular two-hit pathogenesis of NAFLD, the molecular mechanisms underlying hepatic insulin resistance. As fatty liver disease gets advanced, it requires in-vitro as well as in-vivo models closer to disease progression in humans for better understanding the pathological state and identifying a novel therapeutic target. This review summarizes in-vitro (2D cell-culture/co-culture, 3D spheroid/organoid/liver-on-a-chip) models as well as in-vivo (genetically/dietary/chemically induced fatty liver disease) research models. Fatty liver disease research has gathered lots of attention recently since there is no FDA approved therapy available so far. However, there have been numerous promising targets to treat fatty liver disease including potential therapeutic targets under clinical trials are listed in this review.
Collapse
|
46
|
Indolepropionic Acid, a Gut Bacteria-Produced Tryptophan Metabolite and the Risk of Type 2 Diabetes and Non-Alcoholic Fatty Liver Disease. Nutrients 2022; 14:nu14214695. [PMID: 36364957 PMCID: PMC9653718 DOI: 10.3390/nu14214695] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/30/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
An intricate relationship between gut microbiota, diet, and the human body has recently been extensively investigated. Gut microbiota and gut-derived metabolites, especially, tryptophan derivatives, modulate metabolic and immune functions in health and disease. One of the tryptophan derivatives, indolepropionic acid (IPA), is increasingly being studied as a marker for the onset and development of metabolic disorders, including type 2 diabetes (T2D) and non-alcoholic fatty liver disease (NAFLD). The IPA levels heavily depend on the diet, particularly dietary fiber, and show huge variations among individuals. We suggest that these variations could partially be explained using genetic variants known to be associated with specific diseases such as T2D. In this narrative review, we elaborate on the beneficial effects of IPA in the mitigation of T2D and NAFLD, and further study the putative interactions between IPA and well-known genetic variants (TCF7L2, FTO, and PPARG), known to be associated with the risk of T2D. We have investigated the long-term preventive value of IPA in the development of T2D in the Finnish prediabetic population and the correlation of IPA with phytosterols in obese individuals from an ongoing Kuopio obesity surgery study. The diversity in IPA-linked mechanisms affecting glucose metabolism and liver fibrosis makes it a unique small metabolite and a promising candidate for the reversal or management of metabolic disorders, mainly T2D and NAFLD.
Collapse
|
47
|
Kimura M, Iguchi T, Iwasawa K, Dunn A, Thompson WL, Yoneyama Y, Chaturvedi P, Zorn AM, Wintzinger M, Quattrocelli M, Watanabe-Chailland M, Zhu G, Fujimoto M, Kumbaji M, Kodaka A, Gindin Y, Chung C, Myers RP, Subramanian GM, Hwa V, Takebe T. En masse organoid phenotyping informs metabolic-associated genetic susceptibility to NASH. Cell 2022; 185:4216-4232.e16. [PMID: 36240780 PMCID: PMC9617783 DOI: 10.1016/j.cell.2022.09.031] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/01/2022] [Accepted: 09/23/2022] [Indexed: 11/07/2022]
Abstract
Genotype-phenotype associations for common diseases are often compounded by pleiotropy and metabolic state. Here, we devised a pooled human organoid-panel of steatohepatitis to investigate the impact of metabolic status on genotype-phenotype association. En masse population-based phenotypic analysis under insulin insensitive conditions predicted key non-alcoholic steatohepatitis (NASH)-genetic factors including the glucokinase regulatory protein (GCKR)-rs1260326:C>T. Analysis of NASH clinical cohorts revealed that GCKR-rs1260326-T allele elevates disease severity only under diabetic state but protects from fibrosis under non-diabetic states. Transcriptomic, metabolomic, and pharmacological analyses indicate significant mitochondrial dysfunction incurred by GCKR-rs1260326, which was not reversed with metformin. Uncoupling oxidative mechanisms mitigated mitochondrial dysfunction and permitted adaptation to increased fatty acid supply while protecting against oxidant stress, forming a basis for future therapeutic approaches for diabetic NASH. Thus, "in-a-dish" genotype-phenotype association strategies disentangle the opposing roles of metabolic-associated gene variant functions and offer a rich mechanistic, diagnostic, and therapeutic inference toolbox toward precision hepatology. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Masaki Kimura
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Takuma Iguchi
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kentaro Iwasawa
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andrew Dunn
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Wendy L Thompson
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yosuke Yoneyama
- Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan
| | - Praneet Chaturvedi
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Aaron M Zorn
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michelle Wintzinger
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Miki Watanabe-Chailland
- NMR-Based Metabolomics Core Facility, Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Gaohui Zhu
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Masanobu Fujimoto
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Meenasri Kumbaji
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Asuka Kodaka
- Communication Design Center, Advanced Medical Research Center, Yokohama City University, Yokohama 236-0004, Japan
| | | | | | - Robert P Myers
- Gilead Sciences, Foster City, CA 94404, USA; The Liver Company, Inc., Palo Alto, CA 94303, USA
| | - G Mani Subramanian
- Gilead Sciences, Foster City, CA 94404, USA; The Liver Company, Inc., Palo Alto, CA 94303, USA
| | - Vivian Hwa
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Communication Design Center, Advanced Medical Research Center, Yokohama City University, Yokohama 236-0004, Japan.
| |
Collapse
|
48
|
Vyletelová V, Nováková M, Pašková Ľ. Alterations of HDL's to piHDL's Proteome in Patients with Chronic Inflammatory Diseases, and HDL-Targeted Therapies. Pharmaceuticals (Basel) 2022; 15:1278. [PMID: 36297390 PMCID: PMC9611871 DOI: 10.3390/ph15101278] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/03/2022] [Accepted: 10/14/2022] [Indexed: 09/10/2023] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, steatohepatitis, periodontitis, chronic kidney disease, and others are associated with an increased risk of atherosclerotic cardiovascular disease, which persists even after accounting for traditional cardiac risk factors. The common factor linking these diseases to accelerated atherosclerosis is chronic systemic low-grade inflammation triggering changes in lipoprotein structure and metabolism. HDL, an independent marker of cardiovascular risk, is a lipoprotein particle with numerous important anti-atherogenic properties. Besides the essential role in reverse cholesterol transport, HDL possesses antioxidative, anti-inflammatory, antiapoptotic, and antithrombotic properties. Inflammation and inflammation-associated pathologies can cause modifications in HDL's proteome and lipidome, transforming HDL from atheroprotective into a pro-atherosclerotic lipoprotein. Therefore, a simple increase in HDL concentration in patients with inflammatory diseases has not led to the desired anti-atherogenic outcome. In this review, the functions of individual protein components of HDL, rendering them either anti-inflammatory or pro-inflammatory are described in detail. Alterations of HDL proteome (such as replacing atheroprotective proteins by pro-inflammatory proteins, or posttranslational modifications) in patients with chronic inflammatory diseases and their impact on cardiovascular health are discussed. Finally, molecular, and clinical aspects of HDL-targeted therapies, including those used in therapeutical practice, drugs in clinical trials, and experimental drugs are comprehensively summarised.
Collapse
Affiliation(s)
| | | | - Ľudmila Pašková
- Department of Cell and Molecular Biology of Drugs, Faculty of Pharmacy, Comenius University, 83232 Bratislava, Slovakia
| |
Collapse
|
49
|
Hepatocyte phosphatase DUSP22 mitigates NASH-HCC progression by targeting FAK. Nat Commun 2022; 13:5945. [PMID: 36209205 PMCID: PMC9547917 DOI: 10.1038/s41467-022-33493-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/21/2022] [Indexed: 11/08/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH), a common clinical disease, is becoming a leading cause of hepatocellular carcinoma (HCC). Dual specificity phosphatase 22 (DUSP22, also known as JKAP or JSP-1) expressed in numerous tissues plays essential biological functions in immune responses and tumor growth. However, the effects of DUSP22 on NASH still remain unknown. Here, we find a significant decrease of DUSP22 expression in human and murine fatty liver, which is mediated by reactive oxygen species (ROS) generation. Hepatic-specific DUSP22 deletion particularly exacerbates lipid deposition, inflammatory response and fibrosis in liver, facilitating NASH and non-alcoholic fatty liver disease (NAFLD)-associated HCC progression. In contrast, transgenic over-expression, lentivirus or adeno-associated virus (AAV)-mediated DUSP22 gene therapy substantially inhibit NASH-related phenotypes and HCC development in mice. We provide mechanistic evidence that DUSP22 directly interacts with focal adhesion kinase (FAK) and restrains its phosphorylation at Tyr397 (Y397) and Y576 + Y577 residues, subsequently prohibiting downstream activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and nuclear factor-κB (NF-κB) cascades. The binding of DUSP22 to FAK and the dephosphorylation of FAK are indispensable for DUSP22-meliorated NASH progression. Collectively, our findings identify DUSP22 as a key suppressor of NASH-HCC, and underscore the DUSP22-FAK axis as a promising therapeutic target for treatment of the disease.
Collapse
|
50
|
Konieczna J, Fiol M, Colom A, Martínez-González MÁ, Salas-Salvadó J, Corella D, Soria-Florido MT, Martínez JA, Alonso-Gómez ÁM, Wärnberg J, Vioque J, López-Miranda J, Estruch R, Bernal-López MR, Lapetra J, Serra-Majem L, Bueno-Cavanillas A, Tur JA, Martín Sánchez V, Pintó X, Gaforio JJ, Matía-Martín P, Vidal J, Vázquez C, Daimiel L, Ros E, Bes-Rastrollo M, Pascual M, Sorlí JV, Goday A, Zulet MÁ, Moreno-Rodriguez A, Carmona González FJ, Valls-Enguix R, Janer JM, Garcia-Rios A, Casas R, Gomez-Perez AM, Santos-Lozano JM, Basterra-Gortari FJ, Martínez MÁ, Ortega-Azorin C, Bayó J, Abete I, Salaverria-Lete I, Ruiz-Canela M, Babio N, Carres L, Romaguera D. Does Consumption of Ultra-Processed Foods Matter for Liver Health? Prospective Analysis among Older Adults with Metabolic Syndrome. Nutrients 2022; 14:4142. [PMID: 36235794 PMCID: PMC9570694 DOI: 10.3390/nu14194142] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) includes a spectrum of liver alterations that can result in severe disease and even death. Consumption of ultra-processed foods (UPF) has been associated with obesity and related comorbidities. However, the link between UPF and NAFLD has not been sufficiently assessed. We aimed to investigate the prospective association between UPF consumption and liver health biomarkers. Methods: We followed for 1 year 5867 older participants with overweight/obesity and metabolic syndrome (MetS) from the PREDIMED-Plus trial. A validated 143-item semi-quantitative food frequency questionnaire was used to evaluate consumption of UPF at baseline, 6, and 12 months. The degree of processing for foods and beverages (g/day) was established according to the NOVA classification system. The non-invasive fatty liver index (FLI) and hepatic steatosis index (HSI) were used to evaluate liver health at three points in time. The associations between changes in UPF consumption (percentage of total daily dietary intake (g)) and liver biomarkers were assessed using mixed-effects linear models with repeated measurements. Results: In this cohort, UPF consumption at baseline was 8.19% (SD 6.95%) of total daily dietary intake in grams. In multivariable models, each 10% daily increment in UPF consumption in 1 year was associated with significantly greater FLI (β 1.60 points, 95% CI 1.24;1.96 points) and HSI (0.43, 0.29; 0.57) scores (all p-values < 0.001). These associations persisted statistically significant after adjusting for potential dietary confounders and NAFLD risk factors. Conclusions: A higher UPF consumption was associated with higher levels of NAFLD-related biomarkers in older adults with overweight/obesity and MetS.
Collapse
Grants
- PI13/00673, PI13/00492, PI13/00272, PI13/01123, PI13/00462, PI13/00233, PI13/02184, PI13/00728, PI13/01090, PI13/01056, PI14/01722, PI14/00636, PI14/00618, PI14/00696, PI14/01206, PI14/01919, PI14/00853, PI14/01374, PI14/00972, PI14/00728, PI14/01471, PI1 Instituto de Salud Carlos III
- PI0458/2013, PS0358/2016, PI0137/2018 the Consejería de Salud de la Junta de Andalucía
- PROMETEO/2017/017 Generalitat Valenciana
- N/A SEMERGEN
- ICREA Academia programme Institució Catalana de Recerca i Estudis Avançats
- #340918 European Research Council
- N/A Especial Action Project
- 2013ACUP00194 Recercaixa
- IJC2019-042420-I Spanish Ministry of Economy, Industry and Competitiveness and European Social Funds
Collapse
Affiliation(s)
- Jadwiga Konieczna
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Miguel Fiol
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Antoni Colom
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Miguel Ángel Martínez-González
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, IDISNA, University of Navarra, 31008 Pamplona, Spain
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jordi Salas-Salvadó
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició, 43201 Reus, Spain
- University Hospital of Sant Joan de Reus, Nutrition Unit, 43204 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Dolores Corella
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine, University of Valencia,46010 Valencia, Spain
| | - María Trinidad Soria-Florido
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar de Investigaciones Médicas Municipal d’Investigació Médica (IMIM), 08003 Barcelona, Spain
| | - J. Alfredo Martínez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Nutrition, Food Sciences, and Physiology, University of Navarra, IDISNA, 31008 Pamplona, Spain
- Precision Nutrition and Cardiometabolic Health Program, IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain
| | - Ángel M. Alonso-Gómez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba Health Research Institute, Cardiovascular, Respiratory and Metabolic Area, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
| | - Julia Wärnberg
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Nursing, University of Málaga, Institute of Biomedical Research in Malaga (IBIMA), 29016 Málaga, Spain
| | - Jesús Vioque
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL-UMH), 03010 Alicante, Spain
| | - José López-Miranda
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
| | - Ramon Estruch
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08007 Barcelona, Spain
| | - M. Rosa Bernal-López
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Regional University Hospital of Malaga, Instituto de Investigación Biomédica de Málaga (IBIMA), University of Málaga, 29009 Málaga, Spain
| | - José Lapetra
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, 41013 Sevilla, Spain
| | - Lluís Serra-Majem
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Research Institute of Biomedical and Health Sciences (IUIBS), University of Las Palmas de Gran Canaria & Centro Hospitalario Universitario Insular Materno Infantil (CHUIMI), Canarian Health Service, 35001 Las Palmas de Gran Canaria, Spain
| | - Aurora Bueno-Cavanillas
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, 18011 Granada, Spain
| | - Josep A. Tur
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Research Group on Community Nutrition & Oxidative Stress, University of Balearic Islands, 07122 Palma de Mallorca, Spain
| | - Vicente Martín Sánchez
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Institute of Biomedicine (IBIOMED), University of León, 24071 León, Spain
| | - Xavier Pintó
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Lipids and Vascular Risk Unit, Internal Medicine, Hospital Universitario de Bellvitge Idibell-UB, Hospitalet de Llobregat, 08907 Barcelona, Spain
| | - José J. Gaforio
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Ciencias de la Salud, Centro de Estudios Avanzados en Olivar y Aceites de Oliva, Universidad de Jaén, 23071 Jaén, Spain
| | - Pilar Matía-Martín
- Department of Endocrinology and Nutrition, Instituto de Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Josep Vidal
- CIBER Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Department of Endocrinology, Institut d’ Investigacions Biomédiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08007 Barcelona, Spain
| | - Clotilde Vázquez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Endocrinology and Nutrition, Hospital Fundación Jimenez Díaz, Instituto de Investigaciones Biomédicas IISFJD, University Autonoma, 28040 Madrid, Spain
| | - Lidia Daimiel
- Nutritional Control of the Epigenome Group, Precision Nutrition and Obesity Program, IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain
| | - Emilio Ros
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Lipid Clinic, Department of Endocrinology and Nutrition, IDIBAPS, Hospital Clínic, 08036 Barcelona, Spain
| | - Maira Bes-Rastrollo
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, IDISNA, University of Navarra, 31008 Pamplona, Spain
| | - María Pascual
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició, 43201 Reus, Spain
- University Hospital of Sant Joan de Reus, Nutrition Unit, 43204 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Jose V. Sorlí
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine, University of Valencia,46010 Valencia, Spain
| | - Albert Goday
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Unit of Cardiovascular Risk and Nutrition, Institut Hospital del Mar de Investigaciones Médicas Municipal d’Investigació Médica (IMIM), 08003 Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - María Ángeles Zulet
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Nutrition, Food Sciences, and Physiology, University of Navarra, IDISNA, 31008 Pamplona, Spain
| | - Anai Moreno-Rodriguez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba Health Research Institute, Cardiovascular, Respiratory and Metabolic Area, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
| | - Francisco Jesús Carmona González
- Unidad de Gestión Clínica Torrequebrada, Distrito de Atención Primaria Costa del Sol, Servicio Andaluz de Salud, 29640 Benalmádena, Spain
| | | | - Juana M. Janer
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Salud Camp Redó (UBS Son Sardina) Gerència Atenció Primària de Mallorca, 07010 Palma de Mallorca, Spain
| | - Antonio Garcia-Rios
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, 14004 Cordoba, Spain
| | - Rosa Casas
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Internal Medicine, Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, 08007 Barcelona, Spain
| | - Ana M. Gomez-Perez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Virgen de la Victoria Hospital, Department of Endocrinology, Instituto de Investigación Biomédica de Málaga (IBIMA), University of Málaga, 29071 Málaga, Spain
| | - José Manuel Santos-Lozano
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Family Medicine, Research Unit, Distrito Sanitario Atención Primaria Sevilla, 41013 Sevilla, Spain
| | - F. Javier Basterra-Gortari
- Department of Preventive Medicine and Public Health, IDISNA, University of Navarra, 31008 Pamplona, Spain
- Servicio de Endocrinología. Complejo Hospitalario de Navarra, Servicio Navarro de Salud, 31003 Pamplona, Spain
| | - María Ángeles Martínez
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició, 43201 Reus, Spain
- University Hospital of Sant Joan de Reus, Nutrition Unit, 43204 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Carolina Ortega-Azorin
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine, University of Valencia,46010 Valencia, Spain
| | - Joan Bayó
- CAP El Clot, Institut Català de la Salut, 08018 Barcelona, Spain
| | - Itziar Abete
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Nutrition, Food Sciences, and Physiology, University of Navarra, IDISNA, 31008 Pamplona, Spain
| | - Itziar Salaverria-Lete
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Bioaraba Health Research Institute, Cardiovascular, Respiratory and Metabolic Area, Osakidetza Basque Health Service, Araba University Hospital, University of the Basque Country UPV/EHU, 01009 Vitoria-Gasteiz, Spain
| | - Miguel Ruiz-Canela
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, IDISNA, University of Navarra, 31008 Pamplona, Spain
| | - Nancy Babio
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
- Universitat Rovira i Virgili, Departament de Bioquímica i Biotecnologia, Unitat de Nutrició, 43201 Reus, Spain
- University Hospital of Sant Joan de Reus, Nutrition Unit, 43204 Reus, Spain
- Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
| | - Lourdes Carres
- Atención Primaria Sant Martí, Institut Català de la Salut, 08020 Barcelona, Spain
| | - Dora Romaguera
- Research Group on Nutritional Epidemiology & Cardiovascular Physiopathology (NUTRECOR), Health Research Institute of the Balearic Islands (IdISBa), University Hospital Son Espases (HUSE), 07120 Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, 28029 Madrid, Spain
| |
Collapse
|