1
|
Attanasi ML, Gregoski MJ, Rockey DC. Racial Differences in Liver Fibrosis Burden. Dig Dis Sci 2025:10.1007/s10620-025-08936-w. [PMID: 40102343 DOI: 10.1007/s10620-025-08936-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/15/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND & AIMS Liver histology is the classic method for staging the severity of liver fibrosis, which in turn is an important predictor of clinical outcome. Here, we have hypothesized that the susceptibility to develop fibrosis varies among racial groups. METHODS We examined the histology of all patients over 18 years of age who underwent liver biopsy at the Medical University of South Carolina from 1/1/2013 to 7/1/2021. Patients with malignancy, liver metastases, or missing data were excluded. Fibrosis was quantified using the Batts-Ludwig system (F0 = no fibrosis to F4 = histological cirrhosis). Racial groups were propensity matched based on age, gender, diabetes, alcohol consumption, and CDC/ATSDR Social Vulnerability Index Themes to mitigate the risk of selection bias. RESULTS We identified 1101 patients with liver biopsy histological fibrosis scores. The cohort included 23% Black patients. After propensity matching, Black patients were significantly more likely to have Hepatitis C (73/228 (32%) vs 45/228 (20%), p < 0.001) and autoimmune hepatitis (34/228 (15%) vs 6/228 (3%), p < 0.001) than White patients, while White patients were significantly more likely to have metabolic dysfunction associated steatotic liver disease (71/228 (31%) vs 18/228 (8%), p < 0.001). White patients were significantly more likely to have cirrhosis than Black patients (White - 89/228 (39%) vs Black - 68/228 (30%), p < 0.05). CONCLUSION White patients had a greater overall burden of advanced fibrosis (F4/cirrhosis) than Black patients, independent of etiology. The data suggest that fibrosis risk and/or progression may be worse in White than Black patients.
Collapse
Affiliation(s)
- Michael L Attanasi
- Department of Internal Medicine, North Shore University Hospital, Manhasset, NY, USA
| | - Mathew J Gregoski
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, SC, USA
| | - Don C Rockey
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA.
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, SC, USA.
- Department of Internal Medicine, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 803, Charleston, SC, 29425, USA.
| |
Collapse
|
2
|
Wang JJ, Chen XY, Zhang YR, Shen Y, Zhu ML, Zhang J, Zhang JJ. Role of genetic variants and DNA methylation of lipid metabolism-related genes in metabolic dysfunction-associated steatotic liver disease. Front Physiol 2025; 16:1562848. [PMID: 40166716 PMCID: PMC11955510 DOI: 10.3389/fphys.2025.1562848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), is one of the most common chronic liver diseases, which encompasses a spectrum of diseases, from metabolic dysfunction-associated steatotic liver (MASL) to metabolic dysfunction-associated steatohepatitis (MASH), and may ultimately progress to MASH-related cirrhosis and hepatocellular carcinoma (HCC). MASLD is a complex disease that is influenced by genetic and environmental factors. Dysregulation of hepatic lipid metabolism plays a crucial role in the development and progression of MASLD. Therefore, the focus of this review is to discuss the links between the genetic variants and DNA methylation of lipid metabolism-related genes and MASLD pathogenesis. We first summarize the interplay between MASLD and the disturbance of hepatic lipid metabolism. Next, we focus on reviewing the role of hepatic lipid related gene loci in the onset and progression of MASLD. We summarize the existing literature around the single nucleotide polymorphisms (SNPs) associated with MASLD identified by genome-wide association studies (GWAS) and candidate gene analyses. Moreover, based on recent evidence from human and animal studies, we further discussed the regulatory function and associated mechanisms of changes in DNA methylation levels in the occurrence and progression of MASLD, with a particular emphasis on its regulatory role of lipid metabolism-related genes in MASLD and MASH. Furthermore, we review the alterations of hepatic DNA and blood DNA methylation levels associated with lipid metabolism-related genes in MASLD and MASH patients. Finally, we introduce potential value of the genetic variants and DNA methylation profiles of lipid metabolism-related genes in developing novel prognostic biomarkers and therapeutic targets for MASLD, intending to provide references for the future studies of MASLD.
Collapse
Affiliation(s)
- Jun-Jie Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Xiao-Yuan Chen
- Department of Publication Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Yi-Rong Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yan Shen
- Department of Publication Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Meng-Lin Zhu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jun Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jun-Jie Zhang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| |
Collapse
|
3
|
Forouzesh P, Kheirouri S, Alizadeh M. Predicting hepatic steatosis degree in metabolic dysfunction-associated steatotic liver disease using obesity and lipid-related indices. Sci Rep 2025; 15:8612. [PMID: 40074727 PMCID: PMC11904216 DOI: 10.1038/s41598-024-73132-1] [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: 07/10/2024] [Accepted: 09/13/2024] [Indexed: 03/14/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as nonalcoholic fatty liver disease, represents a prevalent condition ranging from simple steatosis to advanced stages associated with liver cancer. Asymptomatic presentation in the majority of cases underscores the need for non-invasive, cost-effective methods to stratify degree of hepatic steatosis. This cross-sectional study aimed to assess the association between obesity and lipid-related indices with the degree of hepatic steatosis in MASLD patients. 150 individuals recently diagnosed with metabolic dysfunction-associated steatotic liver disease were recruited. Anthropometric measurements, including weight, height, and waist circumference (WC), were taken, alongside biochemical parameters such as alanine aminotransferase, aspartate aminotransferase, total cholesterol, triglycerides (TG), high-density lipoprotein, low-density lipoprotein, and fasting plasma glucose, following a 12-h fasting period. Various indicators of obesity and lipid metabolism, including body mass index, waist-to-height ratio (WHtR), a body shape index, lipid accumulation product (LAP), triglyceride-glucose index (TyG), visceral adiposity index, and hepatic steatosis index (HSI), were calculated. The diagnosis of MASLD and degree of hepatic steatosis were established through abdominal ultrasound examination. Data analysis was performed utilizing SPSS version 22. All the investigated indices displayed an area under the curve (AUC) surpassing 0.5, implying a correlation with the degree of hepatic steatosis. Notably, TyG-WC, TyG-WHtR, LAP, and a cardiometabolic obesity index showed the highest AUC values (> 0.7), indicating a relatively strong association with degree of hepatic steatosis. Specifically, in females, TyG-WC (AUC = 0.797, 95% CI 0.712-0.882, threshold = 865.991), while in males, LAP (AUC = 0.746, 95% CI 0.593-0.899, threshold = 74.290), demonstrated the highest AUC values. TyG-WHtR, TyG-WC, and LAP exhibited significant correlations with the degree of hepatic steatosis. Given their non-invasive nature and easy measurement, they hold promise for potential clinical utility, pending validation in additional studies.
Collapse
Affiliation(s)
- Paniz Forouzesh
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Attar Nishabouri St., Tabriz, 5166614711, Iran.
| | - Sorayya Kheirouri
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Attar Nishabouri St., Tabriz, 5166614711, Iran
| | - Mohammad Alizadeh
- Department of Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Attar Nishabouri St., Tabriz, 5166614711, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Speliotes EK, Schneider CV. PNPLA3 I148M Interacts With Environmental Triggers to Cause Human Disease. Liver Int 2025; 45:e16106. [PMID: 39559944 DOI: 10.1111/liv.16106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) affects up to 30% of Western populations. While obesity is a recognized risk factor, MASLD does not develop in all obese individuals, highlighting the need to understand genetic and environmental interactions. The PNPLA3 I148M variant has been identified as a key genetic risk factor, significantly increasing the likelihood of MASLD development and progression. METHODS We reviewed current literature on the role of PNPLA3 I148M in MASLD, focusing on gene-environment interactions involving diet, physical activity, obesity, and insulin resistance. We included studies analysing ethnic differences in PNPLA3 I148M prevalence and its association with MASLD. Additionally, we reviewed data on how PNPLA3 I148M influences the response to therapies, including lipid-lowering medications and GLP-1 agonists. RESULTS The PNPLA3 I148M variant markedly heightens MASLD risk, particularly in Hispanic populations, where a higher prevalence of MASLD is observed. Lifestyle factors such as high sugar intake, alcohol consumption, and physical inactivity exacerbate MASLD risk among I148M carriers. Evidence shows that insulin resistance amplifies MASLD risk associated with the I148M variant, especially in non-diabetic individuals. Moreover, the PNPLA3 I148M variant interacts with other genetic loci, further modifying MASLD risk and disease course. The variant also influences treatment response, with variability observed in effectiveness of lipid-lowering therapies and GLP-1 agonists among carriers. CONCLUSION The interplay between PNPLA3 I148M and environmental factors underscores the need for personalized MASLD prevention and treatment strategies. Targeting both genetic and lifestyle contributors may enhance MASLD management, offering a tailored approach to reducing disease burden.
Collapse
Affiliation(s)
- Elizabeth K Speliotes
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Carolin Victoria Schneider
- Department of Gastroenterology, Endocrinology and Intensive Care, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
5
|
Zhyzhneuskaya SV, Al‐Mrabeh AH, Peters C, Barnes AC, Hollingsworth KG, Welsh P, Sattar N, Lean MEJ, Taylor R. Clinical utility of liver function tests for resolution of metabolic dysfunction-associated steatotic liver disease after weight loss in the Diabetes Remission Clinical Trial. Diabet Med 2025; 42:e15462. [PMID: 39645664 PMCID: PMC11823348 DOI: 10.1111/dme.15462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 12/10/2024]
Abstract
AIMS Ectopic fat is reduced by effective weight management, but difficult to assess clinically. METHODS We evaluated paired data on 42 participants in the intervention group of the Diabetes Remission Clinical Trial (DiRECT) at baseline, 12 and 24 months after weight loss as indicators of liver fat content measured by 3-point Dixon MRI. RESULTS Baseline liver fat was elevated at 13.0 [7.8-23.3]% with fasting plasma glucose 7.9 [7.1-10.1] mmol/L. Prevalence of baseline MASLD was 86.4%. After weight loss of 11.9 ± 1.2 kg (0-37 kg) at 12 months, remission of MASLD occurred in 74% and liver fat normalised for many (1.8 [1.2-5.2]%; p < 0.0001) as did fasting glucose (5.9 [5.5-7.2] mmol/L; p < 0.0001). Alanine aminotransferase (ALT) and gamma glutamyl transferase (GGT) decreased at 12 months by 38 [19-60]% (p < 0·0001) and 38 [16-53]% (p < 0.0001) respectively. The positive predictive value for decrease in liver fat, with baseline values of >40 IU/L, was 100% for ALT and 87.5% for GGT. As expected, change in liver fat correlated with change in ALT (r = 0.64; p < 0.0001), GGT (r = 0.38; p = 0.013), AST (r = 0.36; p = 0.018), fatty liver index (r = 0.50; p < 0.0001) and hepatic steatosis index (r = 0.44; p < 0.0001). CONCLUSION Metabolic dysfunction-associated steatotic liver disease, an important marker of ill-health is improved by intentional weight loss. If enzyme levels are raised at baseline, following weight loss, changes in ALT and GGT usefully reflect change in liver fat content, with high positive predictive value. Monitoring liver enzymes can provide a simple way to assess change in liver fat following weight loss in day-to-day clinical practice.
Collapse
Affiliation(s)
- S. V. Zhyzhneuskaya
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
- University Hospital of North Durham, County Durham and Darlington NHS Foundation TrustDurhamUK
| | - A. H. Al‐Mrabeh
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of EdinburghEdinburghUK
| | - C. Peters
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
| | - A. C. Barnes
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle UniversityNewcastle upon TyneUK
| | - K. G. Hollingsworth
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
| | - P. Welsh
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - N. Sattar
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - M. E. J. Lean
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - R. Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
6
|
Du T, Huang Y, Lv Y, Yuan G. Liver fibrotic burden across the spectrum of hypothyroidism. J Gastroenterol 2025; 60:315-327. [PMID: 39601802 PMCID: PMC11880098 DOI: 10.1007/s00535-024-02184-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Data regarding the prevalence of hepatic fibrotic burden across the spectrum of hypothyroidism are scarce. Hence, we aimed to evaluate the prevalence of liver fibrotic burden across the spectrum of hypothyroidism. METHODS 30,091 individuals who attended a Health Management Centre between 2019 and 2021 were cross-sectionally analyzed. Participants were categorized as having strict-normal thyroid function, low-normal thyroid function, subclinical hypothyroidism, and overt hypothyroidism. Hepatic fibrosis was assessed by vibration-controlled transient elastography (VCTE). Significant and advanced fibrosis were defined as liver stiffness measurement in VCTE of 8.1-9.6 and 9.7-13.5 kPa, respectively. RESULTS Among both men and women, low-normal thyroid function group, subclinical hypothyroidism group, and overt hypothyroidism group all have more liver fibrosis present, including mild fibrosis, significant fibrosis, advanced fibrosis, and cirrhosis, than the strict-normal thyroid function group. The low-normal thyroid function group have the similar liver fibrotic burden to the subclinical hypothyroidism group. The highest liver fibrotic burden was noted in the overt hypothyroidism group. Both significant and advanced liver fibrosis were significantly associated with low-normal thyroid function, subclinical hypothyroidism, and overt hypothyroidism in both men and women. CONCLUSIONS Liver fibrotic burden are highly prevalent in subjects with overt hypothyroidism. Moreover, fibrotic burden increased across the spectrum of hypothyroidism even within the low normal thyroid function. These results suggested that screening for liver fibrosis in patients with hypothyroidism is necessary.
Collapse
Affiliation(s)
- Tingting Du
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China
| | - Yuchai Huang
- Department of Health Management Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yongman Lv
- Department of Health Management Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Gang Yuan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, China.
| |
Collapse
|
7
|
Kozlitina J, Sookoian S. Global Epidemiological Impact of PNPLA3 I148M on Liver Disease. Liver Int 2025; 45:e16123. [PMID: 39373119 DOI: 10.1111/liv.16123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has increased exponentially over the past three decades, in parallel with the global rise in obesity and type 2 diabetes. It is currently the most common cause of liver-related morbidity and mortality. Although obesity has been identified as a key factor in the increased prevalence of MASLD, individual differences in susceptibility are significantly influenced by genetic factors. PNPLA3 I148M (rs738409 C>G) is the variant with the greatest impact on the risk of developing progressive MASLD and likely other forms of steatotic liver disease. This variant is prevalent across the globe, with the risk allele (G) frequency exhibiting considerable variation. Here, we review the contribution of PNPLA3 I148M to global burden and regional differences in MASLD prevalence, focusing on recent evidence emerging from population-based sequencing studies and prevalence assessments. We calculated the population attributable fraction (PAF) as a means of quantifying the impact of the variant on MASLD. Furthermore, we employ quantitative trait locus (QTL) analysis to ascertain the associations between rs738409 and a range of phenotypic traits. This analysis suggests that these QTLs may underpin pleiotropic effects on extrahepatic traits. Finally, we outline potential avenues for further research and identify key areas for investigation in future studies.
Collapse
Affiliation(s)
- Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Silvia Sookoian
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Clinical and Molecular Hepatology, Translational Health Research Center (CENITRES), Maimónides University, Buenos Aires, Argentina
| |
Collapse
|
8
|
Nakatsuka T, Tateishi R, Sato M, Hashizume N, Kamada A, Nakano H, Kabeya Y, Yonezawa S, Irie R, Tsujikawa H, Sumida Y, Yoneda M, Akuta N, Kawaguchi T, Takahashi H, Eguchi Y, Seko Y, Itoh Y, Murakami E, Chayama K, Taniai M, Tokushige K, Okanoue T, Sakamoto M, Fujishiro M, Koike K. Deep learning and digital pathology powers prediction of HCC development in steatotic liver disease. Hepatology 2025; 81:976-989. [PMID: 38768142 PMCID: PMC11825480 DOI: 10.1097/hep.0000000000000904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/05/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND AND AIMS Identifying patients with steatotic liver disease who are at a high risk of developing HCC remains challenging. We present a deep learning (DL) model to predict HCC development using hematoxylin and eosin-stained whole-slide images of biopsy-proven steatotic liver disease. APPROACH AND RESULTS We included 639 patients who did not develop HCC for ≥7 years after biopsy (non-HCC class) and 46 patients who developed HCC <7 years after biopsy (HCC class). Paired cases of the HCC and non-HCC classes matched by biopsy date and institution were used for training, and the remaining nonpaired cases were used for validation. The DL model was trained using deep convolutional neural networks with 28,000 image tiles cropped from whole-slide images of the paired cases, with an accuracy of 81.0% and an AUC of 0.80 for predicting HCC development. Validation using the nonpaired cases also demonstrated a good accuracy of 82.3% and an AUC of 0.84. These results were comparable to the predictive ability of logistic regression model using fibrosis stage. Notably, the DL model also detected the cases of HCC development in patients with mild fibrosis. The saliency maps generated by the DL model highlighted various pathological features associated with HCC development, including nuclear atypia, hepatocytes with a high nuclear-cytoplasmic ratio, immune cell infiltration, fibrosis, and a lack of large fat droplets. CONCLUSIONS The ability of the DL model to capture subtle pathological features beyond fibrosis suggests its potential for identifying early signs of hepatocarcinogenesis in patients with steatotic liver disease.
Collapse
Affiliation(s)
- Takuma Nakatsuka
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ryosuke Tateishi
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaya Sato
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Laboratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Natsuka Hashizume
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Ami Kamada
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Hiroki Nakano
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Yoshinori Kabeya
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Sho Yonezawa
- RWD Analytics, Healthcare & Life Science, IBM Japan Ltd., Tokyo, Japan
| | - Rie Irie
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Hanako Tsujikawa
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshio Sumida
- Department of Internal Medicine, Division of Hepatology and Pancreatology, Aichi Medical University, Aichi, Japan
| | - Masashi Yoneda
- Department of Internal Medicine, Division of Hepatology and Pancreatology, Aichi Medical University, Aichi, Japan
| | - Norio Akuta
- Department of Hepatology, Toranomon Hospital and Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - Takumi Kawaguchi
- Department of Medicine, Division of Gastroenterology, Kurume University School of Medicine, Fukuoka, Japan
| | | | - Yuichiro Eguchi
- Liver Center, Saga University Hospital, Saga, Japan
- Loco Medical General Institute, Saga, Japan
| | - Yuya Seko
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Eisuke Murakami
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazuaki Chayama
- Collaborative Research Laboratory of Medical Innovation, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Hiroshima Institute of Life Sciences, Hiroshima, Japan
| | - Makiko Taniai
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Katsutoshi Tokushige
- Department of Internal Medicine, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology, Saiseikai Suita Hospital, Suita, Osaka, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Mitsuhiro Fujishiro
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Hepatobiliary and Pancreatic Medicine, Kanto Central Hospital, Tokyo, Japan
| |
Collapse
|
9
|
Bashir A, Völzke H, Henck V, Schipf S, Dörr M, Nauck M, Schmidt CO, Aghdassi A, Khattak MNK, Markus MRP, Ittermann T. Prevalence trends of type 2 diabetes treatment, dyslipidemia and hepatic steatosis in Northeast Germany. J Public Health (Oxf) 2025; 47:24-33. [PMID: 39611572 DOI: 10.1093/pubmed/fdae302] [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: 06/12/2024] [Revised: 09/27/2024] [Accepted: 11/13/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The investigation of prevalence trends of metabolic cardiovascular risk factors is important for appropriate planning of future health programs aiming to prevent cardiovascular morbidity and mortality. In a previous study, we demonstrated an increase in the prevalence of type 2 diabetes (T2D) between 2000 and 2010 in Northeast Germany. The purpose of this study is to investigate prevalence trends of T2D treatment, dyslipidemia and hepatic steatosis in Northeast Germany. METHODS The baseline examinations of the first Study of Health in Pomerania (SHIP) project were carried out from 1997 to 2001 (SHIP-START-0, 4308 subjects). A second, independent random sample of the same region was enrolled between 2008 and 2012 (SHIP-TREND-0, 4420 subjects). All data were standardized with post-stratification weighting derived from the adult population of the German federal state of Mecklenburg-West Pomerania. RESULTS The prevalence of metformin intake increased from 2.1% to 4.1% and insulin use from 2.0% to 2.8%. While the prevalence of statin intake increased from 6.8% to 12.2%, the prevalence of dyslipidemia decreased slightly from 49.0% in SHIP-START-0 to 45.5% in SHIP-TREND-0. The prevalence of hepatic steatosis increased from 29.7% to 37.3%. This increase was most prominently observed in women and younger age groups. CONCLUSIONS T2D, dyslipidemia and hepatic steatosis are common and increasing health problems among adults in Northeast Germany. Reassuring healthy diet and controlling obesity may result in prevention of above-mentioned health problems.
Collapse
Affiliation(s)
- Aqsa Bashir
- Institute for Community Medicine-SHIP clinical-epidemiological research, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine-SHIP clinical-epidemiological research, University Medicine Greifswald, Greifswald, Germany
| | - Vivien Henck
- Institute for Community Medicine-SHIP clinical-epidemiological research, University Medicine Greifswald, Greifswald, Germany
| | - Sabine Schipf
- Institute for Community Medicine-SHIP clinical-epidemiological research, University Medicine Greifswald, Greifswald, Germany
- German Center for Diabetes Research (DZD), partner site Greifswald, Greifswald, Germany
| | - Marcus Dörr
- Department of Internal Medicine B-Cardiology, Intensive Care, Pulmonary Medicine and Infectious Diseases, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Matthias Nauck
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Carsten Oliver Schmidt
- Institute for Community Medicine-SHIP clinical-epidemiological research, University Medicine Greifswald, Greifswald, Germany
| | - Ali Aghdassi
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Muhammad N K Khattak
- Institute for Community Medicine-SHIP clinical-epidemiological research, University Medicine Greifswald, Greifswald, Germany
| | - Marcello R P Markus
- Department of Internal Medicine B-Cardiology, Intensive Care, Pulmonary Medicine and Infectious Diseases, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
- German Center for Diabetes Research (DZD), partner site Greifswald, Greifswald, Germany
| | - Till Ittermann
- Institute for Community Medicine-SHIP clinical-epidemiological research, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
10
|
Ebeid A, Mokhtar F, Martinez-Lebron V, Park S, Degann S, Payano J, Vahora Z, Gray S, Johnson L, El-Maouche D, Abutaleb A. Use of noninvasive fibrosis calculators in an urban diabetes center suggests a large burden of undetected advanced liver disease. BMC Endocr Disord 2025; 25:53. [PMID: 40011894 DOI: 10.1186/s12902-025-01881-9] [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: 08/19/2024] [Accepted: 02/13/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Metabolic dysfunction associated steatotic liver disease (MASLD) is prevalent in up to 60% of patients with type 2 diabetes mellitus (T2DM). T2DM accelerates the risk of hepatic fibrosis and hepatocellular carcinoma in patients with MASLD. Our goal in this study was to identify patients with suspected MASLD and hepatic fibrosis in a large T2DM clinic by using noninvasive fibrosis scoring systems. METHODS We conducted a retrospective study of patients with T2DM seen by our endocrinologists at the Medical Faculty Associates (MFA) Diabetes Center in Washington, DC, from November 1, 2021, until November 1, 2022. We included all subjects who were over 18 years old with a hemoglobin A1c (HbA1c) of 6.5 or higher. Patients with a history of significant alcohol consumption, decompensated cirrhosis, previous bariatric surgery, or prior chronic liver disease were excluded from the study. We identified patients at risk for hepatic fibrosis by using the Fibrosis-4 (FIB-4) Index, NAFLD Fibrosis Score (NFS) and AST to Platelet Ratio Index (APRI) when lab values were available. RESULTS A total of 1,411 patients were evaluated for T2DM by an endocrinology provider during the one-year period. Out of these, 336 patients met one or more of the exclusion criteria, leaving a total of 1075 patients included in the analysis. The majority were African American (n = 582, 54%), 261 were Caucasian (24.3%), and 85 were Hispanic (7.9%). Most patients were females (n = 675, 62.7%). The mean HbA1c was 8.1 ± 2.3. 643 patients (59.8%) were insulin dependent. Based on FIB-4 scores, we found that 35 (3.9%) patients had a score of > 2.67 associated with advanced fibrosis and 257 (29%) patients with scores of 1.3-2.67 had moderate fibrosis. Using the NFS calculator, there were 281 (28%) patients with values of > 0.675 consistent with F3-F4 disease. 715 (71.8%) patients with values of < 0.675 consistent with F0-F2 fibrosis. A total of 6(< 1%) patients met criteria for advanced fibrosis by APRI scoring. CONCLUSION In our urban Diabetes Center, utilizing the NFS calculator may detect many patients with advanced liver disease. Further research is needed to ensure the internal validity of the non-invasive tests in predicting liver fibrosis and to correlate these findings with transient elastography and other imaging evidence of fatty liver disease. CLINICAL TRIAL NUMBER Non-applicable.
Collapse
Affiliation(s)
- Ahmed Ebeid
- The George Washington University, Washington, DC, USA
| | - Fatma Mokhtar
- The George Washington University, Washington, DC, USA
| | | | - Susie Park
- Department of Medicine, The George Washington University Hospital, Washington, DC, USA
| | - Seta Degann
- Department of Medicine, The George Washington University Hospital, Washington, DC, USA
| | - Jeremy Payano
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA
| | - Zahid Vahora
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA
| | - Stephen Gray
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA
| | - Lynt Johnson
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA
| | - Diala El-Maouche
- Department of Endocrinology, The George Washington University Medical Faculty Associates, Washington, DC, USA
| | - Ameer Abutaleb
- Department of Surgery, The George Washington Transplant Institute, The George Washington University Hospital, Washington, DC, USA.
| |
Collapse
|
11
|
Talari NK, Mattam U, Rahman AP, Hemmelgarn BK, Wyder MA, Sylvestre PB, Greis KD, Chella Krishnan K. Functional compartmentalization of hepatic mitochondrial subpopulations during MASH progression. Commun Biol 2025; 8:258. [PMID: 39966593 PMCID: PMC11836293 DOI: 10.1038/s42003-025-07713-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
The role of peridroplet mitochondria (PDM) in diseased liver, such as during the progression of metabolic dysfunction-associated steatohepatitis (MASH), remains unknown. We isolated hepatic cytoplasmic mitochondria (CM) and PDM from a mouse model of diet-induced MASLD/MASH to characterize their functions from simple steatosis to advanced MASH, using chow-fed mice as controls. Our findings show an inverse relationship between hepatic CM and PDM levels from healthy to steatosis to advanced MASH. Proteomics analysis revealed these two mitochondrial populations are compositionally and functionally distinct. We found that hepatic PDM are more bioenergetically active than CM, with higher pyruvate oxidation capacity in both healthy and diseased liver. Higher respiration capacity of PDM was associated with elevated OXPHOS protein complexes and increased TCA cycle flux. In contrast, CM showed higher fatty acid oxidation capacity with MASH progression. Transmission electron microscopy revealed larger and elongated mitochondria during healthy and early steatosis, which appeared small and fragmented during MASH progression. These changes coincided with higher MFN2 protein levels in hepatic PDM and higher DRP1 protein levels in hepatic CM. These findings highlight the distinct roles of hepatic CM and PDM in MASLD progression towards MASH.
Collapse
Affiliation(s)
- Noble Kumar Talari
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ushodaya Mattam
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Afra P Rahman
- Medical Sciences Baccalaureate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brook K Hemmelgarn
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael A Wyder
- Department of Cancer Biology, Proteomics Laboratory, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Pamela B Sylvestre
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kenneth D Greis
- Department of Cancer Biology, Proteomics Laboratory, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Karthickeyan Chella Krishnan
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
12
|
Podszun MC, Alawad AS, Lingala S, Morris N, Huang WCA, Rotman Y. Development of Subtle Iron Deficiency During Vitamin E Treatment For Metabolic Dysfunction-Associated Steatotic Liver Disease. J Diet Suppl 2025; 22:284-299. [PMID: 39960325 PMCID: PMC11851352 DOI: 10.1080/19390211.2025.2465414] [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] [Indexed: 02/25/2025]
Abstract
Vitamin E is an effective treatment for metabolic dysfunction-associated steatohepatitis (MASH) but associated with hemorrhagic complications when used for other indications. We aimed to determine the risk of developing iron deficiency during treatment of metabolic dysfunction-associated steatotic liver disease (MASLD) with vitamin E. Iron status was monitored prospectively in 20 people with MASLD treated with 200 - 800 IU/d vitamin E (https://clinicaltrials.gov/study/NCT01792115). To gain mechanistic insights liver histology, hepatic gene expression, hepatic 4-hydroxynonenal, haptoglobin genotype and plasma vitamin E levels were assessed. We found iron deficiency to occur in 11/20 subjects (55%) after a median 11 weeks (range 4-13) of vitamin E treatment, and anemia to occur in 6 of the 11 (30% of study population) after 23 weeks (16-36). Ferritin (84.5 ± 85.2 to 47.8 ± 54.9μg/L, p < 0.001) and mean corpuscular volume (MCV, 86.2 ± 4.9 to 84.3±4.3fL, p = 0.003) significantly decreased, with a concomitant rise in red-cell distribution width (RDW, 13.4 ± 1.3 to 14.4 ± 1.9%, p = 0.003). A gastrointestinal bleeding source was found in 75% of subjects with complete work-up. Iron deficiency occurred in all diabetics vs. 47% of non-diabetics (p 0.007). Iron deficiency risk was not associated with cirrhosis, platelet count, prothrombin time, haptoglobin genotype, or plasma vitamin E level. Changes in hepatic gene expression and oxidative stress were suggestive of an extrahepatic effect. Iron deficiency resolved with appropriate care even with continued vitamin E treatment. We conclude that occult gastrointestinal bleeding and iron deficiency were frequently observed during vitamin E treatment, possibly reflecting an effect on platelet function. Close monitoring is warranted during the first months of treatment, especially in diabetics and subjects with risk factors for gastrointestinal bleeding.
Collapse
Affiliation(s)
- Maren C. Podszun
- Liver & Energy Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ahmad Samer Alawad
- Liver & Energy Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shilpa Lingala
- Liver & Energy Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nevitt Morris
- Liver & Energy Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Wen-Chun A. Huang
- Liver & Energy Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yaron Rotman
- Liver & Energy Metabolism Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
13
|
Bourganou MV, Chondrogianni ME, Kyrou I, Flessa CM, Chatzigeorgiou A, Oikonomou E, Lambadiari V, Randeva HS, Kassi E. Unraveling Metabolic Dysfunction-Associated Steatotic Liver Disease Through the Use of Omics Technologies. Int J Mol Sci 2025; 26:1589. [PMID: 40004054 PMCID: PMC11855544 DOI: 10.3390/ijms26041589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), now referred to as metabolic dysfunction-associated steatotic liver disease (MASLD), is the most prevalent liver disorder globally, linked to obesity, type 2 diabetes, and cardiovascular risk. Understanding its potential progression from simple steatosis to cirrhosis and hepatocellular carcinoma (HCC) is crucial for patient management and treatment strategies. The disease's complexity requires innovative approaches for early detection and personalized care. Omics technologies-such as genomics, transcriptomics, proteomics, metabolomics, and exposomics-are revolutionizing the study of MASLD. These high-throughput techniques allow for a deeper exploration of the molecular mechanisms driving disease progression. Genomics can identify genetic predispositions, whilst transcriptomics and proteomics reveal changes in gene expression and protein profiles during disease evolution. Metabolomics offers insights into the metabolic alterations associated with MASLD, while exposomics links environmental exposures to MASLD progression and pathology. By integrating data from various omics platforms, researchers can map out the intricate biochemical pathways involved in liver disease progression. This review discusses the roles of omics technologies in enhancing the understanding of disease progression and highlights potential diagnostic and therapeutic targets within the MASLD spectrum, emphasizing the need for non-invasive tools in disease staging and treatment development.
Collapse
Affiliation(s)
- Maria V. Bourganou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Maria Eleni Chondrogianni
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioannis Kyrou
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
- College of Health, Psychology and Social Care, University of Derby, Derby DE22 IGB, UK
| | - Christina-Maria Flessa
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Thoracic Diseases Hospital of Athens, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Vaia Lambadiari
- 2nd Department of Internal-Medicine, Diabetes Centre, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Institute for Cardiometabolic Medicine, University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Centre for Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (M.V.B.); (M.E.C.); (C.-M.F.)
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
14
|
Bekheit M, Kamera B, Colacino L, Dropmann A, Delibegovic M, Almadhoob F, Hanafy N, Bermano G, Hammad S. Mechanisms underpinning the effect of exercise on the non-alcoholic fatty liver disease: review. EXCLI JOURNAL 2025; 24:238-266. [PMID: 40071029 PMCID: PMC11895063 DOI: 10.17179/excli2024-7718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/27/2025] [Indexed: 03/14/2025]
Abstract
Non-alcoholic Fatty Liver Disease (NAFLD) - whose terminology was recently replaced by metabolic liver disease (MAFLD) - is an accumulation of triglycerides in the liver of >5 % of its weight. Epidemiological studies indicated an association between NAFLD and reduced physical activity. In addition, exercise has been shown to improve NAFLD independently of weight loss. In this paper, we aim to systematically review molecular changes in sedentary experimental NAFLD models vs. those subjected to exercise. We utilized the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist and standard review techniques. Studies were considered for inclusion if they addressed the primary question: the mechanisms by which exercise influenced NAFLD. This review summarized experimental evidence of improvements in NAFLD with exercise in the absence of weight loss. The pathways involved appeared to have AMPK as a common denominator. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Mohamed Bekheit
- Department of Surgery, NHS Grampian, Foresterhill Health Campus, Ashgrove Road, AB252ZN Aberdeen, UK
- Institute of Medical Sciences, Medical School, Foresterhill Health Campus, Ashgrove Road, AB252ZN Aberdeen, UK
| | - Blessed Kamera
- Department of Surgery, NHS Grampian, Foresterhill Health Campus, Ashgrove Road, AB252ZN Aberdeen, UK
- Institute of Medical Sciences, Medical School, Foresterhill Health Campus, Ashgrove Road, AB252ZN Aberdeen, UK
| | - Laura Colacino
- Department of Surgery, NHS Grampian, Foresterhill Health Campus, Ashgrove Road, AB252ZN Aberdeen, UK
- Institute of Medical Sciences, Medical School, Foresterhill Health Campus, Ashgrove Road, AB252ZN Aberdeen, UK
| | - Anne Dropmann
- Molecular Hepatology Section, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Mirela Delibegovic
- Department of Surgery, NHS Grampian, Foresterhill Health Campus, Ashgrove Road, AB252ZN Aberdeen, UK
- Institute of Medical Sciences, Medical School, Foresterhill Health Campus, Ashgrove Road, AB252ZN Aberdeen, UK
| | - Fatema Almadhoob
- St. Helens and Knowsley Teaching Hospitals NHS Trust, Prescot, Prescot, UK
| | - Nemany Hanafy
- Group of Bionanotechnology and Molecular Cell Biology, Nanomedicine Department, Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, 33516 Kafrelsheikh, Egypt
| | - Giovanna Bermano
- Centre for Obesity Research and Education (CORE), School of Pharmacy and Life Sciences, Robert Gordon University, Sir Ian Wood Building, Garthdee Road, Aberdeen AB10 7GJ, UK
| | - Seddik Hammad
- Molecular Hepatology Section, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
- Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, 83523 Qena, Egypt
| |
Collapse
|
15
|
Karady J, Mayrhofer T, Foldyna B, Lu MT, Meyersohn N, Hoffmann U, Balogon O, Pagidipati N, Shah S, Douglas PS, Ferencik M, Corey K. Coronary Artery Disease and Major Adverse Cardiovascular Events in People With Hepatic Steatosis at Low Atherosclerotic Cardiovascular Disease Risk. Aliment Pharmacol Ther 2025; 61:558-569. [PMID: 39610294 DOI: 10.1111/apt.18415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 06/28/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Hepatic steatosis (HS) and 10-year atherosclerotic cardiovascular disease (ASCVD) risk ≥ 7.5% are associated with increased risk for cardiovascular events. AIM To assess underlying coronary artery disease (CAD) and major adverse cardiovascular event (MACE) among those with and without HS at different ASCVD risk. METHODS We evaluated stable chest pain patients receiving coronary computed tomography (CT) in the PROMISE trial. HS and CAD endpoints were defined on coronary CT. MACE was defined as unstable angina, non-fatal myocardial infarction, and all-cause death. Multivariable Cox regression, adjusting for CAD characteristics, assessed the association of HS with MACE for ASCVD < 7.5%. RESULTS One thousand two hundred and four of 3702 (32.5%) patients were at ASCVD < 7.5% and 20.3% (244/1204) of them had HS. Individuals with HS were younger (54.3 ± 5.2 vs. 55.8 ± 5.2; p < 0.001), more often males (40.2% [98/244] vs. 27.1% [260/960]; p < 0.001), had more risk factors/person (2.06 ± 0.89 vs. 1.93 ± 0.91; p = 0.047). CAD characteristics were similar between HS vs. non-HS patients at ASCVD < 7.5% and ASCVD ≥ 7.5% (all p > 0.05). Patients with HS had greater MACE rate compared to non-HS patients (ASCVD < 7.5%: 3.75%[9/244] vs. 1.5% [14/960]; p = 0.027 and ASCVD ≥ 7.5%: 4.7% [33/696] vs. 3.1% [56/1802]; p = 0.043). In patients without HS, MACE rate was higher in the ASCVD ≥ 7.5% vs. < 7.5% (3.1% [56/1802] vs. 1.5% [14/960]; p = 0.011). In patients with HS, MACE rates were not significantly different between ASCVD ≥ 7.5% vs. < 7.5% (4.7% [33/696] vs. 3.7% [9/244]; p = 0.484). In ASCVD < 7.5%, HS predicted MACE (aHR:2.34, 95%CI:1.01-5.43; p = 0.048), independent of CAD characteristics. CONCLUSIONS Individuals with HS at ASCVD < 7.5% risk had similar CAD characteristics as patients without HS at < 7.5% ASCVD risk, yet experienced comparable MACE rates as those at ASCVD ≥ 7.5%.
Collapse
Affiliation(s)
- Julia Karady
- Cardiovascular Imaging Research Center, Harvard Medical School-Massachusetts General Hospital, Boston, Massachusetts, USA
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Harvard Medical School-Massachusetts General Hospital, Boston, Massachusetts, USA
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Harvard Medical School-Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Harvard Medical School-Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nandini Meyersohn
- Cardiovascular Imaging Research Center, Harvard Medical School-Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center, Harvard Medical School-Massachusetts General Hospital, Boston, Massachusetts, USA
- Cleerly Inc., Denver, Colorado, USA
| | - Oluwafemi Balogon
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Neha Pagidipati
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Svati Shah
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Maros Ferencik
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Kathleen Corey
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
16
|
Al Ashi S, Rizvi AA, Rizzo M. Altered kidney function in fatty liver disease: confronting the "MAFLD-renal syndrome". FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2025; 5:1539117. [PMID: 39845775 PMCID: PMC11751235 DOI: 10.3389/fcdhc.2024.1539117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025]
Affiliation(s)
- Suleiman Al Ashi
- Endocrinology Fellow, UCF COM HCA Healthcare GME – Endocrinology, Diabetes, and Metabolism Fellowship Orlando VA Healthcare System, Orlando, FL, United States
| | - Ali A. Rizvi
- Department of Medicine, Division of Endocrinology, Orlando VA Medical Center and University of Central Florida College of Medicine, Orlando, FL, United States
| | - Manfredi Rizzo
- School of Medicine, Promise Department, University of Palermo, Palermo, Italy
- Department of Medicine, Ras Al Khaimah (RAK) Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| |
Collapse
|
17
|
Obradović F, Vitello DJ, Hasjim BJ, Obayemi J, Polineni P, Gmeiner M, Koep E, Jain A, Crippa F, Duarte-Rojo A, Rohan VS, Kulik L, Doll JM, Banea T, McNatt GE, Zhao L, VanWagner LB, Manski CF, Ladner DP. Comparing the cost of cirrhosis to other common chronic diseases: A longitudinal study in a large national insurance database. Hepatology 2025:01515467-990000000-01133. [PMID: 39773884 DOI: 10.1097/hep.0000000000001206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND AND AIMS Cirrhosis prevalence is increasing, yet costs associated with its chronic, complex care are poorly understood. The aim was to characterize the costs of care for patients with cirrhosis and compare them to other chronic diseases such as heart failure (HF) and chronic obstructive pulmonary disease (COPD), for which the public health burden is better recognized. APPROACH AND RESULTS Patients enrolled in Medicare Advantage plans from a large national insurer between 2011 and 2020 with cirrhosis, HF, and COPD were identified by ICD-9/-10 codes. Costs (USD) of care were calculated per patient-month and included inpatient medical, emergency medical, pharmacy, and other costs. In all, 93,308 patients with cirrhosis, 355,520 patients with HF, and 318,949 patients with COPD were analyzed. Patients with cirrhosis, HF, and COPD had a mean (SD) age of 69.6 (9.5), 75.9 (9.7), and 72.9 (9.8) years, respectively. The most frequent etiologies were metabolic dysfunction-associated steatohepatitis (37.7%) and alcohol-associated cirrhosis (22.1%). The total monthly cost of care for patients with cirrhosis, HF, and COPD was $3032.00, $2491.60, and $1955.60 respectively. The cost for patients with cirrhosis exceeded that for HF by $540.40 (21.7% higher) and COPD by $1076.30 (55.0% higher). The monthly cost of care for decompensated cirrhosis was $3969.30, which was 59.3% ($1477.70) higher than for HF and 103.0% ($1,955.60) higher than for COPD. CONCLUSIONS The cost of care for cirrhosis is high, significantly higher than HF and COPD. Interventions directed at optimizing care to prevent progression to cirrhosis and decompensation are likely to alleviate this public health burden.
Collapse
Affiliation(s)
- Filip Obradović
- Department of Economics, Northwestern University, Evanston, Illinois, USA
| | - Dominic J Vitello
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Bima J Hasjim
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Joy Obayemi
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Praneet Polineni
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michael Gmeiner
- Department of Economics, London School of Economics, London, UK
| | - Eleena Koep
- Center for Health Care Research, UnitedHealth Group, Eden Prairie, Minnesota, USA
| | - Aditya Jain
- Department of Economics, Northwestern University, Evanston, Illinois, USA
| | - Federico Crippa
- Department of Economics, Northwestern University, Evanston, Illinois, USA
| | - Andrés Duarte-Rojo
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Northwestern Medicine, Chicago, Illinois, USA
| | - Vinayak S Rohan
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Surgery, Division of Transplantation, Northwestern Medicine, Chicago, Illinois, USA
| | - Laura Kulik
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Northwestern Medicine, Chicago, Illinois, USA
| | - Julianna M Doll
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Therese Banea
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Gwen E McNatt
- Organ Transplant Center, University of Iowa Healthcare, Iowa City, Iowa, USA
| | - Lihui Zhao
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Lisa B VanWagner
- Department of Medicine, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Charles F Manski
- Department of Economics, Northwestern University, Evanston, Illinois, USA
- Institute for Policy Research, Northwestern University, Evanston, Illinois, USA
| | - Daniela P Ladner
- Northwestern University Transplant Outcomes Research Collaborative (NUTORC), Comprehensive Transplant Center (CTC), Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Surgery, Division of Transplantation, Northwestern Medicine, Chicago, Illinois, USA
| |
Collapse
|
18
|
Schenker RB, Ramirez CB, Jang C, Allayee H, Zhao X, Setchell KDR, Kohli R, Goran MI. Dihydroxyacetone phosphate is a novel predictor of hepatic fibrosis in Latino adolescents with obesity. J Pediatr Gastroenterol Nutr 2025; 80:174-181. [PMID: 39582475 DOI: 10.1002/jpn3.12420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/25/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVES Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common pediatric liver disease and can progress to liver fibrosis. Latino adolescents have increased MASLD and fibrosis risk. While fibrosis is diagnosed by biopsy or imaging, more accessible, noninvasive, and economical screening methods are needed. We aimed to use plasma metabolomics/lipidomics to identify potential fibrosis biomarkers in Latino adolescents with obesity. METHODS Liver stiffness (LS) was measured in 93 Latino adolescents with obesity using magnetic resonance elastography. Metabolites and lipids were extracted from plasma and identified on Compound Discoverer. Associations between metabolites/lipids and fibrosis (LS > 2.73 kPa) were determined using linear regression models after covariate adjustment. False discovery rate (FDR) adjusted Pearson's correlations were performed. Analytes yielding significant FDR-adjusted correlations were examined further by receiver operator curve analysis. RESULTS Mean (±standard deviation) alanine transaminase (ALT) was 45.7(±65.2) IU/L, hepatic fat fraction was 12.7(±9.1)%, and LS was 2.4(±0.3) kPa. We identified 795 metabolites and 413 lipids in plasma, but only one single metabolite, dihydroxyacetone phosphate (DHAP), a marker of triglyceride synthesis, was significantly associated with fibrosis after FDR adjustment (p < 0.05). In terms of predicting fibrosis, ALT had an area under the curve (AUC) of 0.79, and DHAP had an AUC of 0.79. When combined, ALT + DHAP had an AUC of 0.89. CONCLUSIONS The combination of ALT + DHAP may have the potential as an accurate, noninvasive test for liver fibrosis. Our data are limited to Latino children with obesity, and a larger cohort should be examined to further validate this novel biomarker.
Collapse
Affiliation(s)
- Rachel B Schenker
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Cuauhtemoc B Ramirez
- Department of Biologic Chemistry, University of California Irvine School of Medicine, Irvine, California, USA
| | - Cholsoon Jang
- Department of Biologic Chemistry, University of California Irvine School of Medicine, Irvine, California, USA
| | - Hooman Allayee
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, University of Southern California, Los Angeles, California, USA
| | - Xueheng Zhao
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Cincinnati Children's Hospital Medical Center, Division of Pathology and Laboratory Medicine, Cincinnati, Ohio, USA
| | - Kenneth D R Setchell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Cincinnati Children's Hospital Medical Center, Division of Pathology and Laboratory Medicine, Cincinnati, Ohio, USA
| | - Rohit Kohli
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Michael I Goran
- Department of Pediatrics, Division of Endocrinology, Children's Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
19
|
Miller KC, Geyer B, Alexopoulos AS, Moylan CA, Pagidipati N. Disparities in Metabolic Dysfunction-Associated Steatotic Liver Disease Prevalence, Diagnosis, Treatment, and Outcomes: A Narrative Review. Dig Dis Sci 2025; 70:154-167. [PMID: 39560808 DOI: 10.1007/s10620-024-08722-0] [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: 12/19/2023] [Accepted: 10/26/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), is a leading cause of morbidity and mortality, and health disparities have been shown to influence disease burden. AIM In this review, we aim to characterize disparities in prevalence, diagnosis, treatment, and outcomes of MASLD, and to make recommendations for next steps to minimize these disparities. METHODS Literature search on PubMed and Scopus databases was conducted to identify relevant articles published before September 2, 2024. RESULTS Relative to women and White populations, MASLD is more common in men and Hispanic populations and less common in Black populations. It is also more prevalent among those with lower SES. Noninvasive clinical scores may perform differently across groups, and screening practices vary both for initial disease and for progression to metabolic dysfunctionassociated steatohepatitis (MASH), formerly called non-alcoholic steatohepatitis (NASH). Women and Black and Hispanic patients suffer worse outcomes including rates of progression to MASH and mortality. CONCLUSIONS Health disparities related to race, ethnicity, gender, and socioeconomic factors impact multiple stages of care for patients with MASLD.
Collapse
|
20
|
Nso N, Bookani KR, Trimingham M, Orji R, Njei B, Balasubramanian SS, Pursnani A. Liver Fibrosis and Cardiovascular Events. South Med J 2025; 118:19-25. [PMID: 39753232 DOI: 10.14423/smj.0000000000001769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
OBJECTIVES Liver fibrosis represents a common sequela of nonalcoholic fatty liver disease (NAFLD) and other chronic liver diseases. Noninvasive liver fibrosis scores (LFSs) aim to evaluate the severity of liver fibrosis. Whether LFSs can predict the risk of future cardiovascular events (CVEs) remains unclear. This systematic review aimed to clarify the association between liver fibrosis and CVEs by studying the value of LFSs, namely the Fibrosis-4 (FIB-4) Index for Liver Fibrosis score and the NAFLD Fibrosis Score (NFS), for predicting CVEs. METHODS PubMed, Scopus, Web of Science, and Cochrane Library were searched for relevant prospective studies. Retrieved articles were screened to confirm their eligibility for the systematic review. We evaluated the quality of the included studies using the National Institutes of Health tool. RESULTS Twelve studies of high to fair quality were included in this systematic review. Of note, 10/12 studies reported an independent association between high LFSs and the risk of CVEs, cardiovascular mortality, and all-cause mortality (all P < 0.05). In addition, an advanced histological grade of liver fibrosis (grade 3 or 4) was suggestive of CVE occurrence. NAFLD also appeared to be associated with a higher risk of CVEs at any severity of fibrosis (all P < 0.05). CONCLUSIONS The findings of this review suggest that liver fibrosis in patients with NAFLD is an independent predictor of future adverse CVEs, cardiovascular mortality, and all-cause mortality. Noninvasive and easy-to-perform LFSs, including FIB-4 score and the NFS, appear useful in predicting such events in patients with a spectrum of cardiovascular diseases and the general population without known cardiovascular disease.
Collapse
Affiliation(s)
- Nso Nso
- From the Department of Medicine, Division of Cardiovascular Disease, University of Chicago-Northshore Program, Evanston, Illinois
| | - Kaveh Rezaei Bookani
- From the Department of Medicine, Division of Cardiovascular Disease, University of Chicago-Northshore Program, Evanston, Illinois
| | - Mia Trimingham
- American University of Antigua, College of Medicine, St Johns, Antigua and Barbuda
| | - Richard Orji
- Department of Internal Medicine, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois
| | - Basile Njei
- Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Senthil S Balasubramanian
- From the Department of Medicine, Division of Cardiovascular Disease, University of Chicago-Northshore Program, Evanston, Illinois
| | - Amit Pursnani
- From the Department of Medicine, Division of Cardiovascular Disease, University of Chicago-Northshore Program, Evanston, Illinois
| |
Collapse
|
21
|
Le TN, Bright R, Truong V, Li J, Juneja R, Vasilev K. Key biomarkers in type 2 diabetes patients: A systematic review. Diabetes Obes Metab 2025; 27:7-22. [PMID: 39355932 PMCID: PMC11618249 DOI: 10.1111/dom.15991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/10/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is not just a local health issue but a significant global health burden, affecting patient outcomes and clinical management worldwide. Despite the wealth of studies reporting T2DM biomarkers, there is an urgent need for a comparative review. This review aims to provide a comprehensive analysis based on the reported T2DM biomarkers and how these are linked with other conditions, such as inflammation and wound healing. A comparative review was conducted on 24 001 study participants, including 10 024 T2DM patients and 13 977 controls (CTL; age 30-90 years). Four main profiles were extracted and analysed from the clinical reports over the past 11 years: haematological (1084 cases vs. 1458 CTL), protein (6753 cases vs. 9613 CTL), cytokine (975 cases vs. 1350 CTL) and lipid (1212 cases vs. 1556 CTL). This review provides a detailed analysis of the haematological profile in T2DM patients, highlighting fundamental changes such as increased white blood cells and platelet counts, accompanied by decreases in red blood cell counts and iron absorption. In the serum protein profile, a reduction in albumin and anti-inflammatory cytokines was noted along with an increase in globulin levels and pro-inflammatory cytokines. Furthermore, changes in lipid profiles were discussed, specifically the decreases in high-density lipoprotein (HDL) and the increases in low-density lipoprotein (LDL) and triglycerides. Understanding the changes in these four biomarker profiles is essential for developing innovative strategies to create diagnostic and prognostic tools for diabetes management.
Collapse
Affiliation(s)
- Thien Ngoc Le
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Richard Bright
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Vi‐Khanh Truong
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| | - Jordan Li
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
- Department of Renal Medicine, Flinders Medical CentreBedford ParkSouth AustraliaAustralia
| | - Rajiv Juneja
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
- Department of Renal Medicine, Flinders Medical CentreBedford ParkSouth AustraliaAustralia
| | - Krasimir Vasilev
- College of Medicine and Public HealthFlinders UniversityAdelaideSouth AustraliaAustralia
| |
Collapse
|
22
|
Perry AS, Hadad N, Chatterjee E, Jimenez-Ramos M, Farber-Eger E, Roshani R, Stolze LK, Betti MJ, Zhao S, Huang S, Martens L, Kendall TJ, Thone T, Amancherla K, Bailin S, Gabriel CL, Koethe J, Carr JJ, Terry JG, Vaitinadin NS, Freedman JE, Tanriverdi K, Alsop E, Van Keuren-Jensen K, Sauld JFK, Mahajan G, Khan SS, Colangelo L, Nayor M, Fisher-Hoch S, McCormick JB, North KE, Below JE, Wells QS, Abel ED, Kalhan R, Scott C, Guilliams M, Gamazon ER, Fallowfield JA, Banovich NE, Das S, Shah R. A prognostic molecular signature of hepatic steatosis is spatially heterogeneous and dynamic in human liver. Cell Rep Med 2024; 5:101871. [PMID: 39657669 PMCID: PMC11722105 DOI: 10.1016/j.xcrm.2024.101871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/06/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
Hepatic steatosis is a central phenotype in multi-system metabolic dysfunction and is increasing in parallel with the obesity pandemic. We use a translational approach integrating clinical phenotyping and outcomes, circulating proteomics, and tissue transcriptomics to identify dynamic, functional biomarkers of hepatic steatosis. Using multi-modality imaging and broad proteomic profiling, we identify proteins implicated in the progression of hepatic steatosis that are largely encoded by genes enriched at the transcriptional level in the human liver. These transcripts are differentially expressed across areas of steatosis in spatial transcriptomics, and several are dynamic during stages of steatosis. Circulating multi-protein signatures of steatosis strongly associate with fatty liver disease and multi-system metabolic outcomes. Using a humanized "liver-on-a-chip" model, we induce hepatic steatosis, confirming cell-specific expression of prioritized targets. These results underscore the utility of this approach to identify a prognostic, functional, dynamic "liquid biopsy" of human liver, relevant to biomarker discovery and mechanistic research applications.
Collapse
Affiliation(s)
- Andrew S Perry
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Niran Hadad
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Maria Jimenez-Ramos
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | | | - Rashedeh Roshani
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Michael J Betti
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shilin Zhao
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shi Huang
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Liesbet Martens
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Timothy J Kendall
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK; Edinburgh Pathology, University of Edinburgh, Edinburgh, UK
| | - Tinne Thone
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Samuel Bailin
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Curtis L Gabriel
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - J Jeffrey Carr
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | - Jane E Freedman
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Eric Alsop
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | | | | | - Sadiya S Khan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Laura Colangelo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Susan Fisher-Hoch
- School of Public Health, The University of Texas Health Science Center at Houston, Brownsville, TX, USA
| | - Joseph B McCormick
- School of Public Health, The University of Texas Health Science Center at Houston, Brownsville, TX, USA
| | - Kari E North
- CVD Genetic Epidemiology Computational Laboratory, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quinn S Wells
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, USA
| | - Ravi Kalhan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Charlotte Scott
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Eric R Gamazon
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Ravi Shah
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
23
|
Ha S, Wong VWS, Zhang X, Yu J. Interplay between gut microbiome, host genetic and epigenetic modifications in MASLD and MASLD-related hepatocellular carcinoma. Gut 2024; 74:141-152. [PMID: 38950910 PMCID: PMC11671994 DOI: 10.1136/gutjnl-2024-332398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/08/2024] [Indexed: 07/03/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses a wide spectrum of liver injuries, ranging from hepatic steatosis, metabolic dysfunction-associated steatohepatitis (MASH), fibrosis, cirrhosis to MASLD-associated hepatocellular carcinoma (MASLD-HCC). Recent studies have highlighted the bidirectional impacts between host genetics/epigenetics and the gut microbial community. Host genetics influence the composition of gut microbiome, while the gut microbiota and their derived metabolites can induce host epigenetic modifications to affect the development of MASLD. The exploration of the intricate relationship between the gut microbiome and the genetic/epigenetic makeup of the host is anticipated to yield promising avenues for therapeutic interventions targeting MASLD and its associated conditions. In this review, we summarise the effects of gut microbiome, host genetics and epigenetic alterations in MASLD and MASLD-HCC. We further discuss research findings demonstrating the bidirectional impacts between gut microbiome and host genetics/epigenetics, emphasising the significance of this interconnection in MASLD prevention and treatment.
Collapse
Affiliation(s)
- Suki Ha
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vincent Wai-Sun Wong
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiang Zhang
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- 1Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
24
|
Kim K, Lee Y, Lee JS, Kim MN, Kim BK, Kim SU, Park JY, Kim DY, Ahn SH, Jung I, Lee HW. Incidence of metabolic dysfunction-associated steatotic liver disease and advanced fibrosis and impact of overweight/obesity in elderly population: a nationwide cohort study. J Gastroenterol Hepatol 2024; 39:2845-2852. [PMID: 39343427 DOI: 10.1111/jgh.16755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 09/02/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND AND AIM The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing worldwide, coinciding with aging population. However, limited studies have evaluated its incidence and progression to advanced fibrosis in the elderly population. Therefore, our study aimed to investigate the incidence of MASLD and advanced fibrosis in this age group. METHODS We included 878 686 individuals aged ≥60 years from the Korea National Health Insurance Service-Senior cohort. After excluding participants with preexisting MASLD, 329 388 individuals were finally analyzed. Participants were categorized into four groups based on the presence of overweight/obesity and additional risk factors (aRF) included in the cardiometabolic diagnostic criteria of MASLD. RESULTS The overall incidence of MASLD was 1.94 per 100 person-years, and the incidence of advanced fibrosis in MASLD patients was 1.78 per 100 person-years. MASLD development was significantly higher in overweight/obese patients (2.65 per 100 person-years) compared to lean patients (1.09 per 100 person-years), and this trend persisted after stratification by the presence of aRF. Similarly, the incidence of advanced fibrosis among MASLD patients was higher in overweight/obese individuals (2.06 per 100 person-years) compared to lean counterparts (0.87 per 100 person-years), irrespective of aRF. CONCLUSIONS The lower incidence of MASLD in the elderly population compared to the general population underscores the importance of identifying age-specific risk factors. Overweight/obesity emerged as a robust predictor of MASLD development and advanced fibrosis. Additionally, the presence of additional cardiometabolic risk factors further increased the risk of incident MASLD and advanced fibrosis among the elderly.
Collapse
Affiliation(s)
- Kunhee Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yaeji Lee
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Seung Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Yonsei Liver Center, Severance Hospital, Seoul, South Korea
| | - Mi Na Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Yonsei Liver Center, Severance Hospital, Seoul, South Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Yonsei Liver Center, Severance Hospital, Seoul, South Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Yonsei Liver Center, Severance Hospital, Seoul, South Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Yonsei Liver Center, Severance Hospital, Seoul, South Korea
| | - Do Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Yonsei Liver Center, Severance Hospital, Seoul, South Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Yonsei Liver Center, Severance Hospital, Seoul, South Korea
| | - Inkyung Jung
- Division of Biostatistics, Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye Won Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, South Korea
- Yonsei Liver Center, Severance Hospital, Seoul, South Korea
| |
Collapse
|
25
|
Kaylan KB, Paul S. NAFLD No More: A Review of Current Guidelines in the Diagnosis and Evaluation of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Curr Diab Rep 2024; 25:5. [PMID: 39535566 DOI: 10.1007/s11892-024-01558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW Provide a concise update on metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), as well as a practical approach to screening and initial evaluation. RECENT FINDINGS Nomenclature changes have placed a greater focus on cardiometabolic risk factors in the definition of MASLD. Screening for MASLD is by stepwise noninvasive serum and imaging tests which can identify patients at risk for advanced fibrosis and liver-related complications. MASLD has been increasing in prevalence and disease burden but is underrecognized in primary care and endocrinology clinics. Multiple society guidelines, synthesized here, provide a framework for the initial approach in the diagnosis and evaluation of MASLD. Recent advances in pharmacologic treatment underline the importance of screening for patients who are at risk for advanced fibrosis as they are most likely to benefit from new drug classes, such as the liver-directed thyroid receptor agonist resmiterom.
Collapse
Affiliation(s)
- Kerim B Kaylan
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago Medicine, Chicago, IL, USA
| | - Sonali Paul
- Section of Gastroenterology, Hepatology, and Nutrition, Center for Liver Diseases, The University of Chicago Medicine, Chicago, IL, USA.
| |
Collapse
|
26
|
Yang Y, Li S, An Z, Li S. The correlation between non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) with non-alcoholic fatty liver disease: an analysis of the population-based NHANES (2017-2018). Front Med (Lausanne) 2024; 11:1477820. [PMID: 39582979 PMCID: PMC11581862 DOI: 10.3389/fmed.2024.1477820] [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: 08/08/2024] [Accepted: 10/29/2024] [Indexed: 11/26/2024] Open
Abstract
Background/objective Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of liver disorders, from benign steatosis to more severe conditions like non-alcoholic steatohepatitis, with risks of progressing to fibrosis, cirrhosis, and hepatocellular carcinoma. The non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol ratio (NHHR) indicates lipid metabolic dysregulation and is associated with increased risks of various diseases. This study examines the relationship between NHHR and NAFLD to evaluate NHHR as a potential predictive biomarker for NAFLD. Methods Data from the 2017-2018 National Health and Nutrition Examination Survey (NHANES) were used for cross-sectional analysis. After excluding individuals with incomplete data, hepatitis infections, heavy alcohol use, and those under 18, the study included 2,757 adults. The relationship between NHHR and NAFLD was analyzed using multivariable logistic regression, including subgroup analysis and interaction testing. Results Among the 2,757 participants (mean age 49.91 years), 44.9% had NAFLD. NHHR showed a significant positive association with NAFLD, with an unadjusted odds ratio (OR) of 1.71 and a fully adjusted OR of 1.45. Quartile analysis revealed a 228% higher prevalence of NAFLD in the highest NHHR quartile, with an OR of 3.28. This positive association was consistent across various subgroups. Conclusion Our findings suggest that elevated NHHR is positively correlated with the prevalence of NAFLD and possesses predictive value. We recommend that future research validate the clinical utility of NHHR, particularly for early detection of high-risk individuals and guiding personalized interventions.
Collapse
Affiliation(s)
- Yuhao Yang
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shengxi Li
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Zhenmei An
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
| | - Shuangqing Li
- General Practice Ward/International Medical Center Ward, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
de Abreu JDMF, Azulay RS, Rodrigues V, de Abreu SLL, da Glória Tavares M, Pinheiro FCM, de Oliveira Neto CP, Andrade C, Facundo A, Sá AG, Azevedo PR, de Almeida AGP, Costa DCDA, Castro RS, Magalhães M, Nascimento GC, Faria MDS, Ferreira ADSP. Predictors of Hepatic Fibrosis in Type 2 Diabetes Patients with Metabolic-Dysfunction-Associated Steatotic Liver Disease. Biomedicines 2024; 12:2542. [PMID: 39595107 PMCID: PMC11592232 DOI: 10.3390/biomedicines12112542] [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: 10/15/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Approximately 25% of the world's population and more than 60% of patients with type 2 diabetes (T2D) have metabolic-dysfunction-associated steatotic liver disease (MASLD). The association between these pathologies is an important cause of morbidity and mortality in Brazil and worldwide due to the high frequency of advanced fibrosis and cirrhosis. The objective of this study was to determine the epidemiologic and clinical-laboratory profile of patients with T2D and MASLD treated at an endocrinology reference service in a state in northeastern Brazil, and to investigate the association of liver fibrosis with anthropometric and laboratory measurements. METHODS A cross-sectional study was performed in a specialized outpatient clinic with 240 patients evaluated from July 2022 to February 2024, using a questionnaire, physical examination, laboratory tests, and liver elastography with FibroScan®. RESULTS Estimates showed that women (adjusted OR = 2.69, 95% CI = 1.35-5.35, p = 0.005), obesity (adjusted OR = 2.23, 95% CI = 1.22-4.07, p = 0.009), high GGT (adjusted OR = 3.78, 95% CI = 2.01-7.14, p < 0. 001), high AST (adjusted OR = 6.07, 95% CI = 2.27-16.2, p < 0.001), and high ALT (adjusted OR = 3.83, 95% CI = 1.80-8.11, p < 0.001) were associated with the risk of liver fibrosis even after adjusted analysis. CONCLUSIONS The study findings suggested that female sex and BMI were associated with an increased risk of liver fibrosis, highlighting the importance of comprehensive evaluation of these patients. In addition, FIB-4 and MAF-5 provided a good estimate of liver fibrosis in our population and may serve as a useful tool in a public health setting with limited resources.
Collapse
Affiliation(s)
- Joana D’Arc Matos França de Abreu
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Rossana Sousa Azulay
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Vandilson Rodrigues
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Sterffeson Lamare Lucena de Abreu
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Maria da Glória Tavares
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Flávia Coelho Mohana Pinheiro
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Clariano Pires de Oliveira Neto
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Caio Andrade
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis 65020-070, Brazil
| | - Alexandre Facundo
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Adriana Guimarães Sá
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Patrícia Ribeiro Azevedo
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Ana Gregória Pereira de Almeida
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Debora Camelo de Abreu Costa
- Service of Hepatology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (D.C.d.A.C.); (R.S.C.); (A.d.S.P.F.)
| | - Rogério Soares Castro
- Service of Hepatology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (D.C.d.A.C.); (R.S.C.); (A.d.S.P.F.)
| | - Marcelo Magalhães
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Gilvan Cortês Nascimento
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Manuel dos Santos Faria
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis 65020-070, Brazil
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil
| | - Adalgisa de Souza Paiva Ferreira
- Service of Hepatology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (D.C.d.A.C.); (R.S.C.); (A.d.S.P.F.)
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil
| |
Collapse
|
28
|
Whelehan G, Bello O, Hakim O, Ladwa M, Umpleby AM, Amiel SA, Bodicoat DH, Goff LM. Ethnic differences in the relationship between ectopic fat deposition and insulin sensitivity in Black African and White European men across a spectrum of glucose tolerance. Diabetes Obes Metab 2024; 26:5211-5221. [PMID: 39149769 DOI: 10.1111/dom.15867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 08/17/2024]
Abstract
AIM To examine the hypothesis that there would be ethnic differences in the relationship between ectopic fat and tissue-specific insulin resistance (IR) across a spectrum of glucose tolerance in Black African (BA) and White European (WE) men. MATERIALS AND METHODS Fifty-three WE men (23/10/20 normal glucose tolerance [NGT]/impaired glucose tolerance [IGT]/type 2 diabetes [T2D]) and 48 BA men (20/10/18, respectively) underwent a two-step hyperinsulinaemic-euglycaemic clamp with infusion of D-[6,6-2H2]-glucose and [2H5]-glycerol to assess hepatic, peripheral and adipose tissue IR. Magnetic resonance imaging was used to measure subcutaneous adipose tissue, visceral adipose tissue (VAT) and intrahepatic lipid (IHL). Associations between ectopic fat and IR were assessed using linear regression models. RESULTS There were no differences in tissue-specific IR between ethnic groups at any stage of glucose tolerance. VAT level was consistently lower in the BA population; NGT (p = 0.013), IGT (p = 0.006) and T2D (p = 0.015). IHL was also lower in the BA compared with the WE men (p = 0.013). VAT and IHL levels were significantly associated with hepatic IR in the BA population (p = 0.001) and with peripheral IR in the WE population (p = 0.027). CONCLUSIONS The present study suggests that BA and WE men exhibit the same degree of IR across a glucose tolerance continuum, but with lower VAT and IHL levels in the BA population, suggesting that IR may be driven by a mechanism other than increased ectopic fat accumulation in BA men.
Collapse
Affiliation(s)
- Gráinne Whelehan
- Diabetes Research Centre, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, UK
| | - Oluwatoyosi Bello
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Olah Hakim
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Meera Ladwa
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - A Margot Umpleby
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Stephanie A Amiel
- Department of Diabetes, School of Life Course Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | | | - Louise M Goff
- Diabetes Research Centre, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, UK
| |
Collapse
|
29
|
Haghshomar M, Antonacci D, Smith AD, Thaker S, Miller FH, Borhani AA. Diagnostic Accuracy of CT for the Detection of Hepatic Steatosis: A Systematic Review and Meta-Analysis. Radiology 2024; 313:e241171. [PMID: 39499183 DOI: 10.1148/radiol.241171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Background CT plays an important role in the opportunistic identification of hepatic steatosis. CT performance for steatosis detection has been inconsistent across various studies, and no clear guidelines on optimum thresholds have been established. Purpose To conduct a systematic review and meta-analysis to assess CT diagnostic accuracy in hepatic steatosis detection and to determine reliable cutoffs for the commonly mentioned measures in the literature. Materials and Methods A systematic search of the PubMed, Embase, and Scopus databases (English-language studies published from September 1977 to January 2024) was performed. Studies evaluating the diagnostic accuracy of noncontrast CT (NCCT), contrast-enhanced (CECT), and dual-energy CT (DECT) for hepatic steatosis detection were included. Reference standards included biopsy, MRI proton density fat fraction (PDFF), or NCCT. In several CECT and DECT studies, NCCT was used as the reference standard, necessitating subgroup analysis. Statistical analysis included a random-effects meta-analysis, assessment of heterogeneity with use of the I2 statistic, and meta-regression to explore potential sources of heterogeneity. When available, mean liver attenuation, liver-spleen attenuation difference, liver to spleen attenuation ratio, and the DECT-derived fat fraction for hepatic steatosis diagnosis were assessed. Results Forty-two studies (14 186 participants) were included. NCCT had a sensitivity and specificity of 72% and 88%, respectively, for steatosis (>5% fat at biopsy) detection and 82% and 94% for at least moderate steatosis (over 20%-33% fat at biopsy) detection. CECT had a sensitivity and specificity of 66% and 90% for steatosis detection and 68% and 93% for at least moderate steatosis detection. DECT had a sensitivity and specificity of 85% and 88% for steatosis detection. In the subgroup analysis, the sensitivity and specificity for detecting steatosis were 80% and 99% for CECT and 84% and 93% for DECT. There was heterogeneity among studies focusing on CECT and DECT. Liver attenuation less than 40-45 HU, liver-spleen attenuation difference less than -5 to 0 HU, and liver to spleen attenuation ratio less than 0.9-1 achieved high specificity for detection of at least moderate steatosis. Conclusion NCCT showed high performance for detection of at least moderate steatosis. © RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Maryam Haghshomar
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Dominic Antonacci
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Andrew D Smith
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Sarang Thaker
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Frank H Miller
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Amir A Borhani
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| |
Collapse
|
30
|
Zheng H, Guo T, Zhao X, Wang K, Shan S, Xie S, Xu Y, Liu C, Lu W. Helicobacter pylori Infection Is Not Associated with Nonalcoholic Fatty Liver Disease: A Two-Year Cohort Study. Dig Dis 2024; 43:75-83. [PMID: 39496224 DOI: 10.1159/000542180] [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: 02/02/2024] [Accepted: 10/06/2024] [Indexed: 11/06/2024]
Abstract
INTRODUCTION Previous studies reported inconsistent results of the association between Helicobacter pylori infection and nonalcoholic fatty liver disease (NAFLD). METHODS A cohort study of 2,063 adults without NAFLD at baseline, who participated in a repeated health checkup including a 13C-urea breath test and abdominal ultrasonography, was conducted to evaluate the link between H. pylori infection and NAFLD development. RESULTS During a mean follow-up period of 1.7 years, we did not find a significant association between H. pylori infection and NAFLD (hazard ratio = 1.10 (0.86, 1.40), p = 0.4689). We also found that higher age, body mass index (BMI), systolic blood pressure (systolic BP), diastolic blood pressure (diastolic BP), fasting blood glucose, triglycerides, total cholesterol, low-density lipoprotein cholesterol (LDL-C), alanine aminotransferase (ALT), and aspartate aminotransferase (AST) were risk factors, and high-density lipoprotein cholesterol (HDL-C) was a protective factor for NAFLD development. CONCLUSION H. pylori infection might not be positively related to NAFLD development.
Collapse
Affiliation(s)
- Huabo Zheng
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Tangmeng Guo
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofang Zhao
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shengshuai Shan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia, USA
| | - Songpu Xie
- Laboratory of Experimental Cardiology, Department Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Yichen Xu
- Department of Histology and Embryology, Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Chengyun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weilin Lu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Malik A, Javaid S, Malik MI, Qureshi S. Relationship between sarcopenia and metabolic dysfunction-associated steatotic liver disease (MASLD): A systematic review and meta-analysis. Ann Hepatol 2024; 29:101544. [PMID: 39214253 DOI: 10.1016/j.aohep.2024.101544] [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: 10/25/2023] [Revised: 05/13/2024] [Accepted: 06/17/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION AND OBJECTIVES Metabolic dysfunction-associated steatotic liver disease (MASLD) formerly known as Nonalcoholic fatty liver disease (NAFLD) is a common chronic disease. Identifying MASLD risk factors could help early intervention and reduce the burden of the disease. Previous studies investigated the association between sarcopenia and NAFLD. Several trials were published after the last meta-analysis with indecisive results. This is an updated meta-analysis which aims to assess the association between sarcopenia, MASLD, and MASLD-related fibrosis. MATERIALS AND METHODS Relevant trials published on PubMed, Web of Science, Scopus, and Cochrane Library databases until October 2022 were included. We included studies in which skeletal mass index (SMI) or sarcopenia was compared between patients with and without NAFLD now MASLD. Also, studies comparing fibrosis between MASLD patients with and without sarcopenia were included. Data were pooled as odds ratios (ORs) and 95 % confidence intervals (CIs) using Review Manager Software. RESULTS A total of 25 studies were included. The incidence of sarcopenia was significantly higher in MASLD than controls (OR, 1.25; 95 % CI, 1.08-1.44; P = 0.003). SMI odds showed no significant difference between MASLD patients and controls (OR, 1.02; 95 % CI, 0.91-1.15; P = 0.7). MASLD patients with sarcopenia had higher odds of fibrosis than MASLD patients without sarcopenia (OR, 1.49; 95 % CI, 1.03-2.14; P = 0.03). CONCLUSIONS Sarcopenia increased MASLD's probability and was associated with a higher probability of liver fibrosis in MASLD patients. However, SMI had no predictive value of MASLD occurrence.
Collapse
Affiliation(s)
- Adnan Malik
- Mountain Vista Medical Center, Mesa Arizona, USA.
| | | | | | | |
Collapse
|
32
|
Hermanson JB, Tolba SA, Chrisler EA, Leone VA. Gut microbes, diet, and genetics as drivers of metabolic liver disease: a narrative review outlining implications for precision medicine. J Nutr Biochem 2024; 133:109704. [PMID: 39029595 PMCID: PMC11480923 DOI: 10.1016/j.jnutbio.2024.109704] [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: 04/05/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly increasing in prevalence, impacting over a third of the global population. The advanced form of MASLD, Metabolic dysfunction-associated steatohepatitis (MASH), is on track to become the number one indication for liver transplant. FDA-approved pharmacological agents are limited for MASH, despite over 400 ongoing clinical trials, with only a single drug (resmetirom) currently on the market. This is likely due to the heterogeneous nature of disease pathophysiology, which involves interactions between highly individualized genetic and environmental factors. To apply precision medicine approaches that overcome interpersonal variability, in-depth insights into interactions between genetics, nutrition, and the gut microbiome are needed, given that each have emerged as dynamic contributors to MASLD and MASH pathogenesis. Here, we discuss the associations and molecular underpinnings of several of these factors individually and outline their interactions in the context of both patient-based studies and preclinical animal model systems. Finally, we highlight gaps in knowledge that will require further investigation to aid in successfully implementing precision medicine to prevent and alleviate MASLD and MASH.
Collapse
Affiliation(s)
- Jake B Hermanson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Samar A Tolba
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Evan A Chrisler
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Vanessa A Leone
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
33
|
Atoum MF, Padma KR, Don KR. Curcumin is a potential therapeutic agent that ameliorates diabetes among non-alcoholic fatty liver disease coexist with type 2 diabetes. NUTRITION AND HEALTHY AGING 2024; 9:77-90. [DOI: 10.3233/nha-231504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Type 2 diabetes mellitus (T2DM) and non-alcoholic fatty liver disease (NAFLD) harmonize and act synergistically in clinical practices. About 70–80% of diabetic patients develop NAFLD. At the same time, NAFLD existence increases T2DM development. Meanwhile, the presence of T2DM increases the progression to liver disease such as NAFLD, and to non-alcoholic steatohepatitis (NASH). The most prevalent chronic liver disease worldwide is a NAFLD. NAFLD and (T2DM) have a two-way pathophysiologic relationship, with the latter driving the development of the former into NASH. Nonetheless, NASH enhances the threat of cirrhosis as well as hepatocellular carcinoma (HCC), both cases in turn need transplantation of the liver. The only treatment for NAFLD is still lifestyle management because there are no FDA-approved drugs for the condition. In the current study, we review how curcumin (a naturally occurring phytopolyphenol pigment) treats NAFLD. Also we showed broad insights on curcumin-based therapy, by severe reduction of hepatic inflammation. Thus, our review showed that curcumin ingestion considerably decreased glycemic parameters (fasting blood glucose, glycosylated hemoglobin, insulin resistance index (HOMA-IR), and free fatty acids) and adipocyte-fatty acid binding protein (A-FABP), and adipokine released from adipocytes. Clinical trials are needed to evaluate the effects of curcumin and its specific dosage on liver enzymes, glycemic consequences, among NAFLD coexist with T2DM patients.
Collapse
Affiliation(s)
- Manar Fayiz Atoum
- Department of Medical Laboratory Sciences, Faculty of Applied Health Sciences, The Hashemite University, Zarqa, Jordan
| | - Kanchi Ravi Padma
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam (Women’s) University, Tirupati, AP, India
| | - Kanchi Ravi Don
- Department of Oral Pathology and Microbiology, Sree Balaji Dental College and Hospital, Bharath Institute of Higher Education and Research (BIHER) Bharath University, Chennai, Tamil Nadu, India
| |
Collapse
|
34
|
Tang H, Xie L, Liu L, Shen Y, Yang P, Wu J, Zhao X, Li Y, Wang Z, Mao Y. Renal fat deposition measured on dixon-based MRI is significantly associated with early kidney damage in obesity. Abdom Radiol (NY) 2024; 49:3476-3484. [PMID: 38839650 DOI: 10.1007/s00261-024-04391-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/07/2024]
Abstract
PURPOSE To investigate the renal fat deposition on Dixon-based magnetic resonance imaging (MRI) and to explore the predictive value of renal fat biomarkers of magnetic resonance (MR-RFBs) for early kidney damage in obesity. METHODS This prospective study included 56 obese volunteers and 47 non-obese healthy volunteers. All volunteers underwent renal magnetic resonance examinations. The differences in MR-RFBs [including renal proton density fat fraction (PDFF), renal sinus fat volume (RSFV), and perirenal fat thickness (PRFT)] measured on Dixon-based MRI between the obese and non-obese volunteers were analyzed using a general linear model, taking sex, age, diabetes, and hypertension as covariates. The relationship between estimated glomerular filtration rate (eGFR) and demographic, laboratory, and imaging parameters in obese volunteers was examined by correlation analysis. RESULTS Obese volunteers had higher MR-RFBs than non-obese volunteers after controlling for confounders (all p < 0.001). Renal PDFF (r = - 0.383; p = 0.004), RSFV (r = - 0.368; p = 0.005), and PRFT (r = - 0.451; p < 0.001) were significantly negatively correlated with eGFR in obesity. After adjusting for age, sex, body mass index, diabetes, hypertension, visceral adipose tissue, subcutaneous adipose tissue, renal PDFF, and RSFV, PRFT remained independently negatively associated with eGFR (β = - 0.587; p = 0.003). CONCLUSIONS All MR-RFBs are negatively correlated with eGFR in obesity. The MR-RFBs, especially PRFT, may have predictive value for early kidney damage in obesity.
Collapse
Affiliation(s)
- Huali Tang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, China
| | - Lianghua Xie
- Department of Radiology, Affiliated Hospital of Guilin Medical University, No.15 Lequn Road, Guilin Guangxi, China
| | - Liu Liu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, China
| | - Yan Shen
- Chongqing Three Gorges Medical College, Chongqing, China
| | - Ping Yang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, China
| | - Jiamei Wu
- Department of Radiology, Chongqing Dongnan Hospital, No.98 Tongjiang Avenue, Chayuan New District, Nan'an District, Chongqing, China
| | - Xiaofang Zhao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, China
| | - Yi Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, China
| | - Zhihong Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, China
| | - Yun Mao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No.1 Youyi Road, Yuzhong District, Chongqing, China.
| |
Collapse
|
35
|
Srnic N, Dearlove D, Johnson E, MacLeod C, Krupa A, McGonnell A, Frazer-Morris C, O'Rourke P, Parry S, Hodson L. Greater oxidation of dietary linoleate compared to palmitate in humans following an acute high-carbohydrate diet. Clin Nutr 2024; 43:2305-2315. [PMID: 39226718 DOI: 10.1016/j.clnu.2024.08.028] [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: 04/30/2024] [Revised: 08/14/2024] [Accepted: 08/23/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND We have previously demonstrated that dietary saturated fatty acids (SFA), when compared to polyunsaturated fatty acids (PUFA), are preferentially partitioned into oxidation pathways. However, it remains unclear if this preferential handling is maintained when hepatocellular metabolism is shifted toward fatty acid (FA) esterification and away from oxidation, such as when hepatic de novo lipogenesis (DNL) is upregulated. AIM To investigate whether an acute upregulation of hepatic DNL influences dietary FA partitioning into oxidation pathways. METHODS 20 healthy volunteers (11 females) underwent a fasting baseline visit followed by two study days, 2-weeks apart. Prior to each study day, participants consumed an isocaloric high-carbohydrate diet (to upregulate hepatic DNL) for 3-days. On the two study days, participants consumed an identical standardised test meal that contained either [U13C]palmitate or [U13C]linoleate, in random order, to trace the fate of dietary FA. Blood and breath samples were collected over a 6h postprandial period and 13C enrichment in breath CO2 and plasma lipid fractions were measured using gas-chromatography-combustion-isotope ratio mass spectrometry. RESULTS Compared to the baseline visit, fasting plasma triglyceride concentrations and markers of hepatic DNL, the lipogenic and stearyl-CoA desaturase indices, were significantly (p < 0.05) increased after consumption of the high-carbohydrate diet. Appearance of 13C in expired CO2 and tracer recovery were significantly (p < 0.05) higher after consumption of the meal containing [U13C]linoleate compared to [U13C]palmitate (5.1 ± 0.5% vs. 3.7 ± 0.4%), respectively. Incorporation of 13C into the plasma triglyceride and non-esterified fatty acid pool was significantly (p < 0.001) greater for [U13C]palmitate compared to [U13C]linoleate. CONCLUSION Dietary PUFA compared to SFA appear to be preferentially partitioned into oxidation pathways during an acute upregulation of hepatic DNL, thus consumption of a PUFA-enriched diet may help mitigate intrahepatic triglyceride accumulation in individuals at risk of cardiometabolic disease.
Collapse
Affiliation(s)
- Nikola Srnic
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - David Dearlove
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Elspeth Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Cameron MacLeod
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Antoni Krupa
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Alice McGonnell
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Charlotte Frazer-Morris
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Paige O'Rourke
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Sion Parry
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK; Aston Medical School, Aston University, Birmingham B4 7ET, UK
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK; Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
36
|
Gawrieh S, Vilar-Gomez E, Wilson LA, Pike F, Kleiner DE, Neuschwander-Tetri BA, Diehl AM, Dasarathy S, Kowdley KV, Hameed B, Tonascia J, Loomba R, Sanyal AJ, Chalasani N. Increases and decreases in liver stiffness measurement are independently associated with the risk of liver-related events in NAFLD. J Hepatol 2024; 81:600-608. [PMID: 38762169 PMCID: PMC11410523 DOI: 10.1016/j.jhep.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 04/29/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND & AIMS The clinical significance of change in liver stiffness measurement (LSM) by vibration-controlled transient elastography (VCTE) in patients with non-alcoholic fatty liver disease (NAFLD) is not well-understood. We prospectively defined rates of progression to and regression from LSM-defined compensated advanced chronic liver disease (cACLD) and their associations with liver-related events (LREs). METHODS Participants in the NASH Clinical Research Network-led NAFLD Database 2 and 3 studies were included. Progression to cACLD was defined as reaching LSM ≥10 kPa in participants with LSM <10 kPa on initial VCTE; regression from cACLD was defined as reaching LSM <10 kPa in participants with baseline LSM ≥10 kPa. LREs were defined as liver-related death, liver transplant, hepatocellular carcinoma, MELD >15, development of varices, or hepatic decompensation. Univariate and multivariable interval-censored Cox regression analyses were used to compare the cumulative LRE probability by LSM progression and regression status. RESULTS In 1,403 participants, 89 LREs developed over a mean follow-up of 4.4 years, with an annual incidence rate for LREs of 1.5 (95% CI 1.2-1.8). In participants at risk, progression to LSM ≥10 or ≥15 kPa occurred in 29% and 17%, respectively, whereas regression to LSM <10 or <15 kPa occurred in 44% and 49%, respectively. Progressors to cACLD (≥10 kPa) experienced a higher cumulative LRE rate vs. non-progressors (16% vs. 4%, adjusted hazard ratio 4.0; 95% (1.8-8.9); p <0.01). Regressors from cACLD (to LSM <10 kPa) experienced a lower LRE rate than non-regressors (7% vs. 32%, adjusted hazard ratio 0.25; 95% CI 0.10-0.61; p <0.01). CONCLUSIONS Change in LSM over time is independently and bi-directionally associated with risk of LRE and is a non-invasive surrogate for clinical outcomes in patients with NAFLD. IMPACT AND IMPLICATIONS The prognostic value of change in LSM in patients with NAFLD is not well understood. In this large prospective study of patients with NAFLD and serial vibration-controlled transient elastography exams, baseline and dynamic changes in LSM were associated with the risk of developing liver-related events. LSM is a useful non-invasive surrogate of clinical outcomes in patients with NAFLD.
Collapse
Affiliation(s)
- Samer Gawrieh
- Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, IN, United States
| | - Eduardo Vilar-Gomez
- Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, IN, United States
| | - Laura A Wilson
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, United States
| | - Francis Pike
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, IN, United States
| | - David E Kleiner
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, United States
| | | | - Anna Mae Diehl
- Division of Gastroenterology and Hepatology, Duke University, Durham, NC, United States
| | - Srinivasan Dasarathy
- Division of Gastroenterology and Hepatology, Cleveland Clinic Foundation, Cleveland, OH, United States
| | | | - Bilal Hameed
- Division of Gastroenterology and Hepatology, University of California, San Francisco, CA, United States
| | - James Tonascia
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, United States
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, University of California, San Diego, CA, United States
| | - Arun J Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University, Richmond, VA, United States
| | - Naga Chalasani
- Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, IN, United States.
| |
Collapse
|
37
|
Okuma H, Tsuchiya K. Tissue-specific activation of insulin signaling as a potential target for obesity-related metabolic disorders. Pharmacol Ther 2024; 262:108699. [PMID: 39111411 DOI: 10.1016/j.pharmthera.2024.108699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/31/2024] [Indexed: 09/14/2024]
Abstract
The incidence of obesity is rapidly increasing worldwide. Obesity-associated insulin resistance has long been established as a significant risk factor for obesity-related disorders such as type 2 diabetes and atherosclerosis. Insulin plays a key role in systemic glucose metabolism, with the liver, skeletal muscle, and adipose tissue as the major acting tissues. Insulin receptors and the downstream insulin signaling-related molecules are expressed in various tissues, including vascular endothelial cells, vascular smooth muscle cells, and monocytes/macrophages. In obesity, decreased insulin action is considered a driver for associated disorders. However, whether insulin action has a positive or negative effect on obesity-related disorders depends on the tissue in which it acts. While an enhancement of insulin signaling in the liver increases hepatic fat accumulation and exacerbates dyslipidemia, enhancement of insulin signaling in adipose tissue protects against obesity-related dysfunction of various organs by increasing the capacity for fat accumulation in the adipose tissue and inhibiting ectopic fat accumulation. Thus, this "healthy adipose tissue expansion" by enhancing insulin sensitivity in adipose tissue, but not in the liver, may be an effective therapeutic strategy for obesity-related disorders. To effectively address obesity-related metabolic disorders, the mechanisms of insulin resistance in various tissues of obese patients must be understood and drugs that enhance insulin action must be developed. In this article, we review the potential of interventions that enhance insulin signaling as a therapeutic strategy for obesity-related disorders, focusing on the molecular mechanisms of insulin action in each tissue.
Collapse
Affiliation(s)
- Hideyuki Okuma
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan
| | - Kyoichiro Tsuchiya
- Department of Diabetes and Endocrinology, Graduate School of Interdisciplinary Research, Faculty of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi 4093898, Japan.
| |
Collapse
|
38
|
Yang Y, Chen G, Li J, Li J, Zhang O, Zhang X, Li L, Hao J, Wang E, Heng PA. Enabling target-aware molecule generation to follow multi objectives with Pareto MCTS. Commun Biol 2024; 7:1074. [PMID: 39223327 PMCID: PMC11368924 DOI: 10.1038/s42003-024-06746-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Target-aware drug discovery has greatly accelerated the drug discovery process to design small-molecule ligands with high binding affinity to disease-related protein targets. Conditioned on targeted proteins, previous works utilize various kinds of deep generative models and have shown great potential in generating molecules with strong protein-ligand binding interactions. However, beyond binding affinity, effective drug molecules must manifest other essential properties such as high drug-likeness, which are not explicitly addressed by current target-aware generative methods. In this article, aiming to bridge the gap of multi-objective target-aware molecule generation in the field of deep learning-based drug discovery, we propose ParetoDrug, a Pareto Monte Carlo Tree Search (MCTS) generation algorithm. ParetoDrug searches molecules on the Pareto Front in chemical space using MCTS to enable synchronous optimization of multiple properties. Specifically, ParetoDrug utilizes pretrained atom-by-atom autoregressive generative models for the exploration guidance to desired molecules during MCTS searching. Besides, when selecting the next atom symbol, a scheme named ParetoPUCT is proposed to balance exploration and exploitation. Benchmark experiments and case studies demonstrate that ParetoDrug is highly effective in traversing the large and complex chemical space to discover novel compounds with satisfactory binding affinities and drug-like properties for various multi-objective target-aware drug discovery tasks.
Collapse
Affiliation(s)
- Yaodong Yang
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Jinpeng Li
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | - Jianye Hao
- Noah's Ark Lab, Huawei, Shenzhen, China.
| | | | - Pheng-Ann Heng
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
39
|
Bali AD, Rosenzveig A, Frishman WH, Aronow WS. Nonalcoholic Fatty Liver Disease and Cardiovascular Disease: Causation or Association. Cardiol Rev 2024; 32:453-462. [PMID: 36825899 DOI: 10.1097/crd.0000000000000537] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a disease process that is gaining increasing recognition. The global prevalence of NAFLD is increasing in parallel with growing rates of risk factors for NAFLD such as hypertension, obesity, diabetes, and metabolic syndrome. NAFLD has been referred to as a risk factor for cardiovascular disease (CVD). As CVD is the leading cause of morbidity and mortality worldwide, there are constant efforts to describe and alleviate its risk factors. Although there is conflicting data supporting NAFLD as a causative or associative factor for CVD, NAFLD has been shown to be associated with structural, electrical, and atherosclerotic disease processes of the heart. Shared risk factors and pathophysiologic mechanisms between NAFLD and CVD warrant further explication. Pathologic mechanisms such as endothelial dysfunction, oxidative stress, insulin resistance, genetic underpinnings, and gut microbiota dysregulation have been described in both CVD and NAFLD. The mainstay of treatment for NAFLD is lifestyle intervention including physical exercise and hypocaloric intake in addition to bariatric surgery. Investigations into various therapeutic targets to alleviate hepatic steatosis and fibrosis by way of maintaining the balance between lipid synthesis and breakdown. A major obstacle preventing the success of many pharmacologic approaches has been the effects of these medications on CVD risk. The future of pharmacologic treatment of NAFLD is promising as effective medications with limited CVD harm are being investigated.
Collapse
Affiliation(s)
- Atul D Bali
- From the Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | | | - William H Frishman
- From the Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Wilbert S Aronow
- From the Department of Cardiology, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
40
|
Amini-Salehi E, Letafatkar N, Norouzi N, Joukar F, Habibi A, Javid M, Sattari N, Khorasani M, Farahmand A, Tavakoli S, Masoumzadeh B, Abbaspour E, Karimzad S, Ghadiri A, Maddineni G, Khosousi MJ, Faraji N, Keivanlou MH, Mahapatro A, Gaskarei MAK, Okhovat P, Bahrampourian A, Aleali MS, Mirdamadi A, Eslami N, Javid M, Javaheri N, Pra SV, Bakhsi A, Shafipour M, Vakilpour A, Ansar MM, Kanagala SG, Hashemi M, Ghazalgoo A, Kheirandish M, Porteghali P, Heidarzad F, Zeinali T, Ghanaei FM, Hassanipour S, Ulrich MT, Melson JE, Patel D, Nayak SS. Global Prevalence of Nonalcoholic Fatty Liver Disease: An Updated Review Meta-Analysis comprising a Population of 78 million from 38 Countries. Arch Med Res 2024; 55:103043. [PMID: 39094335 DOI: 10.1016/j.arcmed.2024.103043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/09/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a global health challenge, with a rising rate in line with other metabolic diseases. We aimed to assess the global prevalence of NAFLD in adult and pediatric populations. METHODS PubMed, Scopus and Web of Science databases were systematically searched up to May 2023. Heterogeneity was assessed using Cochran's Q test and I2 statistics, and random-effects model was used for meta-analysis. Analyses were performed using STATA version 18. RESULTS A total of 479 studies with 78,001,755 participants from 38 countries were finally included. The global prevalence of NAFLD was estimated to be 30.2% (95% CI: 28.7-31.7%). Regionally, the prevalence of NAFLD was as follows: Asia 30.9% (95% CI: 29.2-32.6%), Australia 16.1% (95% CI: 9.0-24.8%), Europe 30.2% (95% CI: 25.6-35.0%), North America 29% (95% CI: 25.8-32.3%), and South America 34% (95% CI: 16.9-53.5%). Countries with a higher human development index (HDI) had significantly lower prevalence of NAFLD (coefficient = -0.523, p = 0.005). Globally, the prevalence of NAFLD in men and women was 36.6% (95% CI: 34.7-38.4%) and 25.5% (95% CI: 23.9-27.1%), respectively. The prevalence of NAFLD in adults, adults with obesity, children, and children with obesity was 30.2% (95% CI: 28.8-31.7%), 57.5% (95% CI: 43.6-70.9%), 14.3% (95% CI: 10.3-18.8%), and 38.0% (95% CI: 31.5-44.7%), respectively. CONCLUSION The prevalence of NAFLD is remarkably high, particularly in countries with lower HDI. This substantial prevalence in both adults and children underscores the need for disease management protocols to reduce the burden.
Collapse
Affiliation(s)
- Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Negin Letafatkar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Naeim Norouzi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Arman Habibi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mona Javid
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Nazila Sattari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mehrdad Khorasani
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Farahmand
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Shervin Tavakoli
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Behnaz Masoumzadeh
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Elaheh Abbaspour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Department of Radiology, Poursina Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Sahand Karimzad
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Ghadiri
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Gautam Maddineni
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Mohammad Javad Khosousi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Niloofar Faraji
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Abinash Mahapatro
- Department of Internal Medicine, Hi-Tech Medical College and Hospital, Rourkela, Odisha, India
| | | | - Paria Okhovat
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Bahrampourian
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Maryam Sadat Aleali
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Arian Mirdamadi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Narges Eslami
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohamadreza Javid
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Naz Javaheri
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Arash Bakhsi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Shafipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Azin Vakilpour
- Department of Internal Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Malek Moein Ansar
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Department of Biochemistry and Medical Physics, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Mohamad Hashemi
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Arezoo Ghazalgoo
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Masoumeh Kheirandish
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parham Porteghali
- Department of Internal Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Forough Heidarzad
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Taraneh Zeinali
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Fariborz Mansour Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Michael T Ulrich
- Department of Internal Medicine, Riverside University Health System Medical Center, Moreno Valley, CA, USA
| | - Joshua E Melson
- Division of Gastroenterology, Department of Medicine, University of Arizona Medical Center-Banner Health, Tucson, AZ, USA
| | - Dhruvan Patel
- Division of Gastroenterology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
41
|
Li J, Lu Y, Chen H, Zheng P, Zhang X, Zhang Z, Ding L, Wang D, Xu C, Ai X, Zhang Q, Xian J, Hong M. Effects of Dietary Fish Oil Supplementation on the Growth, Proximate Composition, and Liver Health of Chinese Stripe-Necked Turtle ( Mauremys sinensis). Animals (Basel) 2024; 14:2511. [PMID: 39272296 PMCID: PMC11394261 DOI: 10.3390/ani14172511] [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: 06/13/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Dietary lipids provide energy for animals and can also be converted into other nutrients (such as non-essential amino acids), which play a role in saving protein. The Chinese stripe-necked turtle is a protected and endangered species that has been bred in captivity; however, basic data on lipid requirements remain unavailable. In this study, 360 Mauremys sinensis (body weight of 65.32 ± 0.15 g) were randomly divided into six groups with three replicates per group; the turtles were fed experimental diets supplemented with various levels of fish oil (i.e., 1% (control group, CG), 3.5% (HF-1), 6% (HF-2), 8.5% (HF-3), 11% (HF-4), and 13.5% (HF-5)) for 10 weeks. The results showed that compared with CG, increasing the fish oil level promoted the growth performance of turtles, and the HF-3 group achieved the best effect. The HF-4 group showed the highest increases in the hepatosomatic index and viscerosomatic index. In addition, increased lipid levels also increased the crude lipid content and reduced the crude protein content in muscle tissue. Oil red O staining showed that the liver lipid content increased with the level of supplemented fish oil, which is consistent with the results of the hepatosomatic index. Compared with CG, triglyceride, total cholesterol, and low-density lipoprotein cholesterol increased significantly in both the liver and serum when fish oil levels exceeded 8.5% (p < 0.05), while high-density lipoprotein cholesterol decreased significantly. Aspartate transaminase and cerealthirdtransaminase levels in serum increased significantly when fish oil levels exceeded 8.5% (p < 0.05). Moreover, the activities of antioxidant enzymes (GSH-Px, SOD, T-AOC, and CAT) and MDA showed similar results, indicating that high fish oil levels (8.5-13.5%) caused liver tissue damage in M. sinensis. Increased fish oil levels significantly upregulated the expression levels of cytokines (IFN-γ, TNF-α, TGF-β1, IL-10, and IL-12) (p < 0.05), downregulated the expression levels of antioxidant enzyme-related genes (cat, mn-sod, and gsh-px), and increased apoptosis of liver cells. Supplementation of the diet with 3.5-6% fish oil improved the growth performance of M. sinensis, and the turtles maintained a beneficial immune status. The results provide a scientific basis for optimizing the commercial feed formula of M. sinensis.
Collapse
Affiliation(s)
- Juntao Li
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Yaopeng Lu
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Huiqin Chen
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Peihua Zheng
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Xiuxia Zhang
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Zelong Zhang
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Li Ding
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Dongmei Wang
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Chi Xu
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Xiaoqi Ai
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Qiongyu Zhang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| | - Jianan Xian
- Hainan Provincial Key Laboratory for Functional Components Research and Utilization of Marine Bio-Resources, Institute of Tropical Biosciences and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Meiling Hong
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou 571158, China
| |
Collapse
|
42
|
Hassan F, Farman M, Khan KA, Awais M, Akhtar S. Prevalence of nonalcoholic fatty liver disease in Pakistan: a systematic review and meta-analysis. Sci Rep 2024; 14:19573. [PMID: 39179792 PMCID: PMC11344154 DOI: 10.1038/s41598-024-70481-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver condition globally and the leading cause of liver-related death and morbidity. The goal of this study was to collect current data in order to calculate the pooled prevalence of NAFLD in Pakistan. We conducted a comprehensive literature search on four electronic databases until March 2024 to find studies on the prevalence of NAFLD in Pakistan. Pooled prevalence estimates of NAFLD were obtained using random-effects meta-analytic models. The chi-square test was used to account for study heterogeneity, whereas the I2 statistic was used to assess inconsistency. The data were stratified by the general population (average risk) and individuals with metabolic diseases (high risk). Two reviewers thoroughly and independently screened, reviewed, and assessed all studies. In total, 468 studies were reviewed, and 34 were included. The pooled NAFLD prevalence in the general population was 29.82% (95% CI 21.39-39.01%; prediction interval: 2.98-68.92%) based on 13 studies. In individuals with metabolic disorders, the prevalence of NAFLD in patients with diabetes, hypertension, and obesity, was 58.47% (95% CI 54.23-62.64%; prediction interval: 38.16-77.40%), 74.08% (95% CI 60.50-85.70%), and 47.43% (95% CI 30.49-64.66%), respectively. There was no evidence of publication bias, although a statistically significant level of heterogeneity was seen among the studies (I2 ranged from 57.5 to 98.69%). The findings of this study indicate a substantial prevalence of NAFLD in the population of Pakistan. The Pakistani government must formulate a comprehensive approach and plan aimed at augmenting awareness, control, prevention, and treatment of fatty liver disease.Prospero Registration no: CRD42022356607.
Collapse
Affiliation(s)
- Fazal Hassan
- Department of Mathematics and Statistics, The University of Haripur, Haripur, KP, Pakistan
| | | | - Kauser Aftab Khan
- Department of Community Medicine Gujranwala, Medical College Gujranwala, Gujranwala, Pakistan
| | | | - Sohail Akhtar
- Department of Mathematics and Statistics, The University of Haripur, Haripur, KP, Pakistan.
| |
Collapse
|
43
|
Habib S. Team players in the pathogenesis of metabolic dysfunctions-associated steatotic liver disease: The basis of development of pharmacotherapy. World J Gastrointest Pathophysiol 2024; 15:93606. [PMID: 39220834 PMCID: PMC11362842 DOI: 10.4291/wjgp.v15.i4.93606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
Nutrient metabolism is regulated by several factors. Social determinants of health with or without genetics are the primary regulator of metabolism, and an unhealthy lifestyle affects all modulators and mediators, leading to the adaptation and finally to the exhaustion of cellular functions. Hepatic steatosis is defined by presence of fat in more than 5% of hepatocytes. In hepatocytes, fat is stored as triglycerides in lipid droplet. Hepatic steatosis results from a combination of multiple intracellular processes. In a healthy individual nutrient metabolism is regulated at several steps. It ranges from the selection of nutrients in a grocery store to the last step of consumption of ATP as an energy or as a building block of a cell as structural component. Several hormones, peptides, and genes have been described that participate in nutrient metabolism. Several enzymes participate in each nutrient metabolism as described above from ingestion to generation of ATP. As of now several publications have revealed very intricate regulation of nutrient metabolism, where most of the regulatory factors are tied to each other bidirectionally, making it difficult to comprehend chronological sequence of events. Insulin hormone is the primary regulator of all nutrients' metabolism both in prandial and fasting states. Insulin exerts its effects directly and indirectly on enzymes involved in the three main cellular function processes; metabolic, inflammation and repair, and cell growth and regeneration. Final regulators that control the enzymatic functions through stimulation or suppression of a cell are nuclear receptors in especially farnesoid X receptor and peroxisome proliferator-activated receptor/RXR ligands, adiponectin, leptin, and adiponutrin. Insulin hormone has direct effect on these final modulators. Whereas blood glucose level, serum lipids, incretin hormones, bile acids in conjunction with microbiota are intermediary modulators which are controlled by lifestyle. The purpose of this review is to overview the key players in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) that help us understand the disease natural course, risk stratification, role of lifestyle and pharmacotherapy in each individual patient with MASLD to achieve personalized care and target the practice of precision medicine. PubMed and Google Scholar databases were used to identify publication related to metabolism of carbohydrate and fat in states of health and disease states; MASLD, cardiovascular disease and cancer. More than 1000 publications including original research and review papers were reviewed.
Collapse
Affiliation(s)
- Shahid Habib
- Department of Hepatology, Liver Institute PLLC, Tucson, AZ 85712, United States
| |
Collapse
|
44
|
Han HS, Choi BH, Jang SY, Choi S, Hwang GS, Koo SH. Regulation of hepatic lipogenesis by asymmetric arginine methylation. Metabolism 2024; 157:155938. [PMID: 38795769 DOI: 10.1016/j.metabol.2024.155938] [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: 12/26/2023] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND AND AIMS Hepatic lipogenesis is elevated in nutrient abundant conditions to convert the excess carbohydrate into triacylglycerol (TAG). Fatty acyl moiety of TAG is eventually transported into adipose tissues by very low density lipoprotein, leading to the accumulation of TAG as a preferred storage form of excess energy. Disruption of the balance between TAG clearance and synthesis leads to the accumulation of lipids in the liver, leading to the progression of non-alcoholic fatty liver disease (NAFLD) including non-alcoholic steatohepatitis. Protein arginine methyltransferase (PRMT) 6 has been linked to the various metabolic processes including hepatic gluconeogenesis, muscle atrophy and lipodystrophy in mouse models. However, the role of PRMT6 in the control of hepatic lipogenesis has not been elucidated to date. METHODS We assessed the interaction between PRMT6 and LXR alpha by using co-immunoprecipitation assay. The specific arginine residue of LXR alpha that is methylated by PRMT6 was assessed by LC-MS/MS assay and the functional consequences of LXR alpha methylation was explored by mSREBP-1c luciferase assay. The effect of PRMT6 on hepatic lipogenesis was assessed by adenovirus-mediated ectopic expression of PRMT6 or knockdown of PRMT6 via shRNA in hepatocytes. Finally, the role of PRMT6 in hepatic lipid metabolism in vivo was explored by either ectopic expression of LXR alpha mutant that is defective in PRMT6-mediated arginine methylation or knockdown of PRMT6 in liver. RESULTS We found that promoter activity of sterol regulatory element binding protein (SREBP) 1c is robustly activated by PRMT6. Interestingly, we demonstrated that PRMT6 binds to LXR alpha, a transcription factor for SREBP-1c, via its LXXLL motif, leading to the asymmetric dimethylation of an arginine residue and activation of this protein. Indeed, ectopic expression of PRMT6 in hepatocytes led to the enhanced expression of LXR alpha target genes in the lipogenic pathway. Conversely, genetic or pharmacological inhibition of PRMT6 diminished expression of lipogenic genes and the lipid accumulation in primary hepatocytes. Mechanistically, we found that asymmetric dimethylation of LXR alpha led to the dissociation of small heterodimer partner (SHP), a transcriptional co-inhibitor of this factor, resulting in the activation of LXR alpha-mediated transcriptional process. Finally, we showed that disruption of asymmetric dimethylation of LXR alpha in the liver led to the diminished expression of genes in the lipogenesis, resulting in the reduced hepatic lipid accumulation in high fat diet-fed mice in vivo. CONCLUSIONS We showed that PRMT6 modulates LXR alpha activity by conferring asymmetric dimethylation of arginine 253, thus blocking SHP-mediated inhibition and promoting hepatic lipid accumulation. These results suggest that PRMT6 is critical in the control of lipid homeostasis by regulation of LXR alpha-mediated lipogenesis in the liver.
Collapse
Affiliation(s)
- Hye-Sook Han
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Byeong Hun Choi
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Seo Young Jang
- Integrated Metabolomics Research Group, Metropolitan Seoul Center, Korea Basic Science Institute, Seoul 03759, Republic of Korea
| | - Seri Choi
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Metropolitan Seoul Center, Korea Basic Science Institute, Seoul 03759, Republic of Korea; College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Seung-Hoi Koo
- Division of Life Sciences, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
45
|
Qiao X, Wang X, Guo L, Pan Q. Excessive Daytime Napping Increases the Risk of Non-Alcoholic Fatty Liver Disease: A Meta-Analysis and a Mendelian Randomization Study. Nat Sci Sleep 2024; 16:1067-1074. [PMID: 39071543 PMCID: PMC11283788 DOI: 10.2147/nss.s468444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024] Open
Abstract
Background Prior research based on observations has furnished evidence that supports a connection between daytime napping and the prevalence of non-alcoholic fatty liver disease (NAFLD). Nevertheless, the question of whether this correlation is indicative of a causal link has not been definitively answered. Methods We used meta-analysis and Mendelian randomization (MR) to synthesize genetic and observational data. A two-sample MR analysis was conducted, leveraging 105 single-nucleotide polymorphisms (SNPs) known to be associated with daytime napping patterns. Additionally, summary-level data pertaining to NAFLD outcomes were acquired from the comprehensive UK Biobank study. Network meta-analyses were employed to investigate the relationship between excessive daytime napping and NAFLD, while subgroup was also performed. Results Significant associations were observed between daytime napping and NAFLD. The systematic review/meta-analysis uncovered a heightened risk of NAFLD development among individuals who engaged in daytime naps exceeding 30 minutes, when compared to those who did not nap(odds ratio [OR] = 1.32, 95% confidence interval [CI]: 1.05 to 1.66). Furthermore, MR analysis indicated that a genetic propensity towards longer daytime napping was significantly linked to an increased likelihood of NAFLD (OR = 2.26, 95% CI: 1.38 to 3.73). Conclusion Daytime napping has been found to be causally related to a higher risk of NAFLD. Furthermore, across all participants, napping for an average duration over 30 minutes was linked to an elevated likelihood of NAFLD.
Collapse
Affiliation(s)
- Xiuqi Qiao
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, People’s Republic of China
| | - Xiaoxia Wang
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People’s Republic of China
| | - Lixin Guo
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, People’s Republic of China
| | - Qi Pan
- Department of Endocrinology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People’s Republic of China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, People’s Republic of China
| |
Collapse
|
46
|
de Oliveira LDRP, Ribeiro TCDR, Mourao CA, Barra MADL, Silva MHG, Shafee LP, Zacarias SM, Campos LDC, Valério HMG, Chebli JMF. Metabolic dysfunction-associated steatotic liver disease prevalence and risk factors in inflammatory bowel disease in tertiary center. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e20231321. [PMID: 39045949 PMCID: PMC11288268 DOI: 10.1590/1806-9282.20231321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/02/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVE The aim of this study was to evaluate the prevalence and risk factors related to metabolic dysfunction-associated steatotic liver disease in inflammatory bowel disease patients. METHODS This is a cross-sectional study conducted on adults with inflammatory bowel disease from 2019 to 2021. Metabolic dysfunction-associated steatotic liver disease encompasses patients with steatosis and at least one cardiometabolic risk factor. Patients with alcohol consumption ≥20 g/day, chronic liver diseases, or methotrexate use were excluded. RESULTS Almost 140 patients were included: 67.1% were female, with a mean age of 49.7±13.7 years, and 63.6% had Crohn's disease. The mean duration of inflammatory bowel disease was 9.7±7.9 years. Metabolic dysfunction-associated steatotic liver disease was observed in 44.3% and advanced liver fibrosis was excluded in 63.5% by Fibrosis-4. Patients with metabolic dysfunction-associated steatotic liver disease were older (p = 0.003) and had a higher number of metabolic syndrome components (2.9±1.1 versus 1.6±1.0; p<0.001), greater abdominal circumference (p<0.001), and body mass index (p<0.001). The only factor related to inflammatory bowel disease associated with metabolic dysfunction-associated steatotic liver disease was disease duration (11.6±9.5 versus 8.3±6.2; p = 0.017). A higher number of metabolic syndrome components and obesity increase by 2.2 times and an altered waist circumference by 2.6 times the occurrence of metabolic dysfunction-associated steatotic liver disease. CONCLUSION A high prevalence of metabolic dysfunction-associated steatotic liver disease was observed in patients with inflammatory bowel disease, with the main risk factors being associated with metabolic syndrome predicting it, but not with inflammatory bowel disease features and/or its treatment.
Collapse
Affiliation(s)
| | | | - Carlos Alberto Mourao
- Universidade Federal de Juiz de Fora, Department of Physiology – Juiz de Fora (MG), Brazil
| | | | | | - Luis Pordeus Shafee
- Universidade Federal de Juiz de Fora, Hospital Universitário – Juiz de Fora (MG), Brazil
| | - Sarah Mendes Zacarias
- Universidade Federal de Juiz de Fora, Hospital Universitário – Juiz de Fora (MG), Brazil
| | | | | | | |
Collapse
|
47
|
Arowolo O, Suvorov A. Underexplored Molecular Mechanisms of Toxicity. J Xenobiot 2024; 14:939-949. [PMID: 39051348 PMCID: PMC11270369 DOI: 10.3390/jox14030052] [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: 05/31/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
Social biases may concentrate the attention of researchers on a small number of well-known molecules/mechanisms leaving others underexplored. In accordance with this view, central to mechanistic toxicology is a narrow range of molecular pathways that are assumed to be involved in a significant part of the responses to toxicity. It is unclear, however, if there are other molecular mechanisms which play an important role in toxicity events but are overlooked by toxicology. To identify overlooked genes sensitive to chemical exposures, we used publicly available databases. First, we used data on the published chemical-gene interactions for 17,338 genes to estimate their sensitivity to chemical exposures. Next, we extracted data on publication numbers per gene for 19,243 human genes from the Find My Understudied Genes database. Thresholds were applied to both datasets using our algorithm to identify chemically sensitive and chemically insensitive genes and well-studied and underexplored genes. A total of 1110 underexplored genes highly sensitive to chemical exposures were used in GSEA and Shiny GO analyses to identify enriched biological categories. The metabolism of fatty acids, amino acids, and glucose were identified as underexplored molecular mechanisms sensitive to chemical exposures. These findings suggest that future effort is needed to uncover the role of xenobiotics in the current epidemics of metabolic diseases.
Collapse
Affiliation(s)
| | - Alexander Suvorov
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, 686 North Pleasant Street, Amherst, MA 01003, USA;
| |
Collapse
|
48
|
Xiao JL, Liu HY, Sun CC, Tang CF. Regulation of Keap1-Nrf2 signaling in health and diseases. Mol Biol Rep 2024; 51:809. [PMID: 39001962 DOI: 10.1007/s11033-024-09771-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/01/2024] [Indexed: 07/15/2024]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) functions as a central regulator in modulating the activities of diverse antioxidant enzymes, maintaining cellular redox balance, and responding to oxidative stress (OS). Kelch-like ECH-associated protein 1 (Keap1) serves as a principal negative modulator in controlling the expression of detoxification and antioxidant genes. It is widely accepted that OS plays a pivotal role in the pathogenesis of various diseases. When OS occurs, leading to inflammatory infiltration of neutrophils, increased secretion of proteases, and the generation of large quantities of reactive oxygen radicals (ROS). These ROS can oxidize or disrupt DNA, lipids, and proteins either directly or indirectly. They also cause gene mutations, lipid peroxidation, and protein denaturation, all of which can result in disease. The Keap1-Nrf2 signaling pathway regulates the balance between oxidants and antioxidants in vivo, maintains the stability of the intracellular environment, and promotes cell growth and repair. However, the antioxidant properties of the Keap1-Nrf2 signaling pathway are reduced in disease. This review overviews the mechanisms of OS generation, the biological properties of Keap1-Nrf2, and the regulatory role of its pathway in health and disease, to explore therapeutic strategies for the Keap1-Nrf2 signaling pathway in different diseases.
Collapse
Affiliation(s)
- Jiang-Ling Xiao
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan, 410012, China
| | - Heng-Yuan Liu
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan, 410012, China
| | - Chen-Chen Sun
- Institute of Physical Education, Hunan First Normal University, Changsha, Hunan, 410205, China.
| | - Chang-Fa Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of the Hunan Province, College of Physical Education, Hunan Normal University, Changsha, Hunan, 410012, China.
| |
Collapse
|
49
|
Zhang J, Li Y, Yang L, Ma N, Qian S, Chen Y, Duan Y, Xiang X, He Y. New advances in drug development for metabolic dysfunction-associated diseases and alcohol-associated liver disease. Cell Biosci 2024; 14:90. [PMID: 38971765 PMCID: PMC11227172 DOI: 10.1186/s13578-024-01267-9] [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: 03/14/2024] [Accepted: 06/19/2024] [Indexed: 07/08/2024] Open
Abstract
Metabolic disorders are currently threatening public health worldwide. Discovering new targets and developing promising drugs will reduce the global metabolic-related disease burden. Metabolic disorders primarily consist of lipid and glucose metabolic disorders. Specifically, metabolic dysfunction-associated steatosis liver disease (MASLD) and alcohol-associated liver disease (ALD) are two representative lipid metabolism disorders, while diabetes mellitus is a typical glucose metabolism disorder. In this review, we aimed to summarize the new drug candidates with promising efficacy identified in clinical trials for these diseases. These drug candidates may provide alternatives for patients with metabolic disorders and advance the progress of drug discovery for the large disease burden.
Collapse
Affiliation(s)
- Jinming Zhang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yixin Li
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, Anhui, China
| | - Liu Yang
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ningning Ma
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shengying Qian
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yingfen Chen
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, Anhui, China.
| | - Xiaogang Xiang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yong He
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
50
|
Govardhan B, Anand VK, Nagaraja Rao P, Balachandran Menon P, Mithun S, Sasikala M, Sowmya T, Anuradha S, Smita CP, Nageshwar Reddy D, Ravikanth V. 17-Beta-Hydroxysteroid Dehydrogenase 13 Loss of Function Does Not Confer Protection to Nonalcoholic Fatty Liver Disease in Indian Population. J Clin Exp Hepatol 2024; 14:101371. [PMID: 38523737 PMCID: PMC10956055 DOI: 10.1016/j.jceh.2024.101371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 02/15/2024] [Indexed: 03/26/2024] Open
Abstract
Background A splice variant in HSD17B13 gene is demonstrated to protect against nonalcoholic fatty liver disease (NAFLD), and mitigate the effect of Patatin-like phospholipase domain-containing 3 (PNPLA3-I148M). It is being explored as a putative drug target and in polygenic risk scores. Based on whole exome sequencing (WES) in our cohort of biopsy proven NAFLD and limited data on the variant in our ethnicity, we sought to explore its role. Methods This is a cross-sectional study that recruited 1,020 individuals with ultrasound/biopsy-confirmed NAFLD and matched controls. Liver enzymes and lipid profiles were estimated (Beckman coulter LX750/DXH800); WES was performed in NAFLD patients and controls (Illumina; HiSeqX); HSD17B13-A-INS/I148M-PNPLA3 variants were genotyped (sequencing/qR T-PCR); HSD17B13 protein expression was estimated (immunohistochemistry); the Student's t-test/Mann-Whitney U/Chi-square test and odds ratio (95% confidence interval) were used. Results There was no significant difference (Odds ratio = 0.76; 95% CI -0.57 to 1.03; P = 0.76) in the frequency of the rs72613567-A-INS between controls and patients (17.8% vs. 14.4%). No difference in the ALT (Alanine transaminase; 72.24 ± 65.13 vs. 73.70 ± 60.06; P = 0.51) and AST levels (Aspartate aminotransferase; 60.72 ± 55.59 vs. 61.63 ± 60.33; P = 0.91) between HSD17B13-wild and variant carriers were noted. Significantly elevated liver enzymes were seen in PNPLA3-148-variant/HSD17B13-wild compared with PNPLA3-148-variant/HSD17B13-variant (90.44 ± 59.0 vs. 112.32 ± 61.78; P = 0.02). No difference in steatosis (P = 0.51) between HSD17B13-wild and variant carriers was noted. No other variants in the intron-exon boundaries were identified. Although, protein expression differences were noted between wild and variant carriers, no difference in the extent of steatosis was seen. Conclusion Our study reports lack of association of the splice variant with reduced risk of NAFLD, and mitigating the effect of PNPLA3 variant. Ethnicity-based validation must be carried out before including it in assessing protection against NAFLD.
Collapse
Affiliation(s)
- Bale Govardhan
- Asian Healthcare Foundation, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| | - V. Kulkarni Anand
- AIG Hospitals, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| | - Padaki Nagaraja Rao
- AIG Hospitals, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| | - P. Balachandran Menon
- AIG Hospitals, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| | - Sharma Mithun
- AIG Hospitals, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| | - Mitnala Sasikala
- Asian Healthcare Foundation, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| | - T.R. Sowmya
- AIG Hospitals, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| | - Sekaran Anuradha
- AIG Hospitals, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| | - C. Pawar Smita
- Department of Genetics, Osmania University, Hyderabad, Telangana 500032, India
| | - D. Nageshwar Reddy
- AIG Hospitals, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| | - Vishnubhotla Ravikanth
- Asian Healthcare Foundation, Plot No 2/3/4/5, Survey No 136/1, Mindspace Road, Gachibowli, Hyderabad, Telangana 500032, India
| |
Collapse
|