1
|
Seko Y, Yamaguchi K, Shima T, Iwaki M, Takahashi H, Kawanaka M, Tanaka S, Mitsumoto Y, Yoneda M, Nakajima A, Okanoue T, Itoh Y. Clinical Utility of Genetic Variants in PNPLA3 and TM6SF2 to Predict Liver-Related Events in Metabolic Dysfunction-Associated Steatotic Liver Disease. Liver Int 2025; 45:e16124. [PMID: 39373247 DOI: 10.1111/liv.16124] [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: 05/11/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND AND AIMS Fibrosis-4 (FIB-4) index and genetic polymorphisms have been used in assessing the risk of liver-related events (LRE) in metabolic dysfunction-associated steatotic liver disease (MASLD). To establish a more efficient prediction strategy for LRE, we investigated a combined approach that uses the FIB-4 index and genetic polymorphisms. METHODS We enrolled 1304 Japanese patients with biopsy-proven MASLD in this longitudinal multicenter cohort study. PNPLA3, TM6SF2, GCKR and MBOAT7 genotypes were genotyped, and polygenic risk score high fat content (PRS-HFC) were calculated. RESULTS During the follow-up period of 8.1 year, 96 LRE occurred and 53 patients died. PNPLA3, TM6SF2 and GCKR genotypes were associated with LRE development. We divided patients into three groups based on the FIB-4 index and PNPLA3 and TM6SF2 genotype. The cumulative LRE development rate in each group was 2.1%/28.9%/53.5%, respectively, at 10 years. Multivariate analysis revealed hazard ratios (HRs) for LRE of 10.72 in the high-risk group and 4.80 in the intermediate-risk group. Overall survival in each group was 98.8%/85.2%/72.4%, respectively, at 10 years. HRs for prognosis were 8.74 in the high-risk group and 5.62 in the intermediate-risk group. Patients with FIB-4 index > 2.67 and high PRS-HFC had HR of 6.70 for LRE development and HR of 6.07 for prognosis compared to patients with FIB-4 ≤ 2.67. CONCLUSIONS The approach of measuring the FIB-4 index first followed by assessment of genetic polymorphisms efficiently detected patients at high risk of developing LRE. Therefore, this two-step strategy could be used as a screening method in large populations of patients with MASLD.
Collapse
Affiliation(s)
- Yuya Seko
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kanji Yamaguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshihide Shima
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Miwa Kawanaka
- General Internal Medicine 2, General Medical Center, Kawasaki Medical School, Okayama, Japan
| | - Saiyu Tanaka
- Center for Digestive and Liver Diseases, Nara City Hospital, Nara, Japan
| | - Yasuhide Mitsumoto
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
2
|
Petta S, Armandi A, Bugianesi E. Impact of PNPLA3 I148M on Clinical Outcomes in Patients With MASLD. Liver Int 2025; 45:e16133. [PMID: 39412170 PMCID: PMC11815615 DOI: 10.1111/liv.16133] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/23/2024] [Accepted: 10/02/2024] [Indexed: 01/03/2025]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a heterogenous clinical and histopathological entity, where multiple metabolic co-factors are intertwined with high interindividual variability. The impact and severity of each factor (including obesity and type 2 diabetes) define a systemic dysmetabolism that can lead to either advanced liver disease and its complication (including hepatocellular carcinoma and clinical events related to portal hypertension) or extrahepatic events: incident cardiovascular disease, chronic kidney disease and extrahepatic cancers. The balance between environmental factors and genetic susceptibility has unique implications in MASLD: the intermittent injury of metabolic co-factors, their fluctuation over time and their specific management, are counterbalanced by the presence of gene variants that can significantly impact the disease at multiple levels. The I148M variant in the PNPLA3 gene is the most investigated genetic susceptibility that induces a more severe steatohepatitis, enhanced fibrogenesis and can shape the incidence of long-term clinical events regardless of, or worsened by, other metabolic risk factors. METHODS AND RESULTS In this review, we will summarise the updated evidence on the natural history of MASLD accounting for classical metabolic risk factors, the role of PNPLA3 in clinical sub-phenotyping (e.g., 'lean MASLD'), impact on disease severity and fibrosis progression, as well as its role for prognostication, alone or in combination with non-invasive tools into polygenic risk scores.
Collapse
Affiliation(s)
- Salvatore Petta
- Sezione di Gastroenterologia, Di.Bi.M.I.SUniversity of PalermoPalermoItaly
| | - Angelo Armandi
- Division of Gastroenterology and Hepatology, Department of Medical SciencesUniversity of TurinTurinItaly
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical SciencesUniversity of TurinTurinItaly
| |
Collapse
|
3
|
Bornia Matavelli C, Echeverria LS, Pereira LM, Chrispim I, Mounzer DLS, Chaim FDM, Chaim EA, Utrini MP, Gestic MA, Callejas-Neto F, Cazzo E. Short-Term Evolution of MASLD Following Roux-en-Y Gastric Bypass: A Focus on Fibrotic MASH. Obes Surg 2025; 35:926-933. [PMID: 39870941 DOI: 10.1007/s11695-025-07688-w] [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: 10/03/2024] [Revised: 12/23/2024] [Accepted: 01/11/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) includes simple steatosis and metabolic dysfuncion-associated steatohepatitis (MASH), with fibrosis in MASH serving as a critical prognostic marker. This study investigates the effects of Roux-en-Y gastric bypass (RYGB) on fibrotic MASH, assessed using the fibrotic NASH index (FNI) and the non-invasive NASH detection score (NI-NASH-DS), as well as provides further data on the diagnostic accuracy of both scores. METHODS A retrospective cohort study was conducted involving 104 individuals (91.3% female, mean age 39.4 ± 8.6 years) who underwent RYGB. Histopathological evaluations during surgery identified fibrotic MASH, and FNI and NI-NASH-DS values were calculated at baseline and 1 year post-surgery. RESULTS At the time of surgery, participants had a mean BMI of 35.3 ± 2.8 kg/m2, which decreased to 27.1 ± 4.0 kg/m2 1 year after surgery. The mean % total weight loss (%TWL) was 23.8 ± 10.1%, and the mean % excess weight loss (%EWL) was 82.4 ± 37.3%. Fibrotic MASH was present in 17.3% of participants pre-operatively. The mean FNI score significantly decreased after surgery (p < 0.0001). Female gender (p < 0.001) and histological evidence of lobular inflammation (p < 0.001) were independently associated with the improvement of FNI. The FNI demonstrated high diagnostic accuracy (sensitivity: 61.1%, specificity: 96.4%, overall accuracy: 90.2%). NI-NASH-DS had a 60% accuracy in detecting MASH, alongside an 85.9% specificity. CONCLUSIONS RYGB seemingly promotes improvement of fibrotic MASH as evaluated by FNI, highlighting its potential as a therapeutic intervention to mitigate MASLD progression. Both FNI and NI-NASH-DS are helpful and inexpensive tools for assessing MASH.
Collapse
|
4
|
Chen VL, Vespasiani‐Gentilucci U. Integrating PNPLA3 into clinical risk prediction. Liver Int 2025; 45:e16103. [PMID: 39282715 PMCID: PMC11815612 DOI: 10.1111/liv.16103] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 02/05/2025]
Abstract
The PNPLA3-rs738409-G variant was the first common variant associated with hepatic fat accumulation and progression of metabolic dysfunction-associated steatotic liver disease (MASLD). Nevertheless, to date, the clinical translation of this discovery has been minimal because it has not yet been clearly demonstrated where the genetic information may play an independent and additional role in clinical risk prediction. In this mini-review, we will discuss the most relevant evidence regarding the potential integration of the PNPLA3 variant into scores and algorithms for liver disease diagnostics and risk stratification, specifically focusing on MASLD but also extending to liver diseases of other etiologies. The PNPLA3 variant adds little in diagnosing the current state of the disease, whether in terms of presence/absence of metabolic dysfunction-associated steatohepatitis or the stage of fibrosis. While it can play an important role in prediction, allowing for the early definition of risk profiles that enable tailored monitoring and interventions over time, this is most valuable when applied to populations with relatively high pre-test probability of having significant fibrosis based on either non-invasive tests (e.g. Fibrosis-4) or demographics (e.g. diabetes). Indeed, in this context, integrating FIB4 with the PNPLA3 genotype can refine risk stratification, though there is still no evidence that genetic information adds to liver stiffness determined by elastography. Similarly, in patients with known liver cirrhosis, knowing the PNPLA3 genotype can play a role in predicting the risk of hepatocellular carcinoma, while more doubts remain about the risk of decompensation.
Collapse
Affiliation(s)
- Vincent L. Chen
- Division of Gastroenterology and Hepatology, Department of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Umberto Vespasiani‐Gentilucci
- Research Unit of HepatologyUniversità Campus Bio‐Medico di RomeRomeItaly
- Hepatology and Clinical Medicine UnitFondazione Policlinico Universitario Campus‐Biomedico di RomaRomeItaly
| |
Collapse
|
5
|
Soma H, Yoshida R, Ishizuka S. Quantitative analysis of sterol balance in a mouse model of hepatic lipid accumulation induced by cholesterol and cholic acid supplementation. Biosci Biotechnol Biochem 2025; 89:438-445. [PMID: 39656874 DOI: 10.1093/bbb/zbae183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/27/2024] [Indexed: 12/17/2024]
Abstract
The cholesterol balance and bile acid metabolism in a mouse model of hepatic lipid accumulation induced by a diet supplemented with cholesterol and cholic acid (CA) were quantitatively evaluated. The mice were fed diets supplemented with different levels of cholesterol (0, 3, or 6 g/kg of diet) and CA (0.5 g/kg of diet) for 6 weeks. Cholesterol supplementation doubled the hepatic triglyceride concentration, regardless of the supplementation level, without inflammation or gallstone formation. Both cholesterol supplementations enhanced fecal excretion of muricholic acid. Additionally, the higher cholesterol supplementation led to an increase in fecal cholesterol excretion, accompanied by elevated expression of hepatic cholesterol exporters and a reduction in fecal bile acid excretion. In this mouse study, supplementation with 3 g cholesterol/kg diet and 0.5 g CA/kg diet was sufficient to induce hepatic lipid accumulation.
Collapse
Affiliation(s)
- Hinata Soma
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Ryo Yoshida
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| | - Satoshi Ishizuka
- Research Faculty of Agriculture, Hokkaido University, Sapporo, Japan
| |
Collapse
|
6
|
Jurado Vélez J, Anderson N, Datcher I, Foster C, Jackson P, Hidalgo B. Striving Towards Equity in Cardiovascular Genomics Research. Curr Atheroscler Rep 2025; 27:34. [PMID: 39964583 PMCID: PMC11836143 DOI: 10.1007/s11883-025-01277-z] [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] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
PURPOSE OF REVIEW Our review emphasizes recent advancements and persisting gaps in cardiovascular genomics, particularly highlighting how emerging studies involving underrepresented populations have uncovered new genetic variants associated with cardiovascular diseases. RECENT FINDINGS Initiatives like the H3Africa project, the Million Veterans Program, and the All of Us Research Program are working to address this gap by focusing on underrepresented groups. Additionally, emerging research is centering on the interplay between genetic factors and socio-environmental determinants of health, which disproportionately impact marginalized communities. As cardiovascular genomics research grows, increasing the inclusion of underrepresented populations is essential for gaining a more comprehensive understanding of genetic variability. This will lead to more accurate and clinically meaningful strategies for preventing and treating cardiovascular diseases across all ancestral backgrounds and diverse populations.
Collapse
Affiliation(s)
- Javier Jurado Vélez
- Marnix E Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nekayla Anderson
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ivree Datcher
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christy Foster
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Pamela Jackson
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bertha Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
7
|
Liu Z, Chen X, Yuan H, Jin L, Zhang T, Chen X. Dissecting the shared genetic architecture between nonalcoholic fatty liver disease and type 2 diabetes. Hum Mol Genet 2025; 34:338-346. [PMID: 39690818 DOI: 10.1093/hmg/ddae184] [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: 06/11/2024] [Revised: 09/04/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
Observational studies have reported a bidirectional correlation between nonalcoholic fatty liver disease (NAFLD) and type 2 diabetes (T2D), but the shared genetic basis between the two conditions remains unclear. Using genome-wide association study (GWAS) summary data from European-ancestry populations, we examined the cross-trait genetic correlation and identified genomic overlaps and shared risk loci. We employed a latent causal variable model and Mendelian randomization (MR) analysis to infer causal relationships. Colocalization analysis and conditional/conjunctional false discovery rate (condFDR/conjFDR) were used to identify genomic overlaps and shared risk loci. Two-step MR analysis was utilized to identify potential mediators. We observed a strong positive genomic correlation between NAFLD and T2D (rg = 0.652, P = 5.67 × 10-6) and identified tissue-specific transcriptomic correlations in the pancreas, liver, skeletal muscle, subcutaneous adipose, and blood. Genetic enrichment was observed in NAFLD conditional on associations with T2D and vice versa, indicating significant polygenic overlaps. We found robust evidence for the causal effect of NAFLD on T2D, particularly insulin-related T2D, rather than vice versa. Colocalization analysis identified shared genomic regions between NAFLD and T2D, including GCKR, FTO, MAU2-TM6SF2, and PNPLA3-SAMM50. High-density lipoprotein cholesterol and insulin were partly mediated the association between NAFLD and T2D. These findings unveil a close genetic link between NAFLD and T2D, shedding light on the biological mechanisms connecting NAFLD progression to T2D.
Collapse
Affiliation(s)
- Zhenqiu Liu
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Xiaochen Chen
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, 227 Chongqing South RD, Shanghai 200025, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
| | - Tiejun Zhang
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, 130 Dong'an RD, Shanghai 200032, China
- Department of Epidemiology, School of Public Health, Fudan University, 130 Dong'an RD, Shanghai 200032, China
- Yiwu Research Institute of Fudan University, 2 Chengbei RD, Yiwu 322000, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering and Human Phenome Institute, Fudan University, 825 Zhangheng RD, Pudong New Area, Shanghai 201203, China
- Fudan University Taizhou Institute of Health Sciences, 799 Yaocheng RD, Taizhou 225316, China
- Yiwu Research Institute of Fudan University, 2 Chengbei RD, Yiwu 322000, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, 12 Urumqi RD, Shanghai 200040, China
| |
Collapse
|
8
|
Xu J, Li QQ, Yang S, Teng HD, Lu ZY, Gu YZ, Xi JH, Mei ZN, Chen Y, Yang GZ. Garcibracgluinols A-C, structurally intriguing polycyclic polyprenylated acylphloroglucinols from Garcinia bracteata alleviate hepatocyte lipid accumulation and insulin resistance. J Mol Struct 2025; 1323:140755. [DOI: 10.1016/j.molstruc.2024.140755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Hur YK, Lee HE, Yoo JY, Park YN, Lee IH, Bae YS. NADPH oxidase 4-SH3 domain-containing YSC84-like 1 complex participates liver inflammation and fibrosis. Free Radic Biol Med 2025; 227:246-259. [PMID: 39645205 DOI: 10.1016/j.freeradbiomed.2024.12.021] [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: 08/31/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
There is growing evidence that NADPH oxidase 4 (Nox4) in hepatocytes contributes to liver inflammation and fibrosis during the development of metabolic dysfunction-associated steatohepatitis (MASH). However, how Nox4 is regulated and leads to liver pathogenesis is unclear. Our previous studies showed that the cytosolic protein SH3 domain-containing Ysc84-like 1 (SH3YL1) regulates Nox4 activity. Here, we asked whether SH3YL1 also participates in liver inflammation and fibrosis during MASH development. We generated that whole body SH3YL1 knockout (SH3YL1-/-), Nox4 knockout (Nox4-/-) mice, and the hepatocyte-specific SH3YL1 conditional knockout (Alb-Cre/SH3YL1fl/fl) mice were fed a methionine/choline-deficient (MCD) diet to induce liver inflammation and fibrosis in pathogenesis of MASH. Palmitate-stimulated primary SH3YL1-and Nox4-deficient hepatocytes and hepatic stellate cells (HSCs) did not generate H2O2. While the liver of MCD diet-fed wild type (WT) mice demonstrated elevated 3-nitrotyrosine as a protein oxidation and 4-hydroxynonenal adducts as a lipid oxidation and increased liver inflammation, hepatocyte apoptosis, and liver fibrosis, these events were markedly reduced in SH3YL1-/-, Nox4-/-, and Alb-Cre/SH3YL1fl/fl mice. The MCD diet-fed WT mice also showed elevated hepatocyte expression of SH3YL1 protein. Similarly, liver biopsies from MASH patients demonstrated strong hepatocyte SH3YL1 protein expression, whereas hepatocytes from patients with steatosis weakly expressed SH3YL1 and histologically normal patient hepatocytes exhibited very little SH3YL1 expression. The Nox4-SH3YL1 complex in murine hepatocytes elevates their H2O2 production, which promotes the liver inflammation, hepatocyte apoptosis, and liver fibrosis that characterize MASH. This axis may also participate in MASH in humans.
Collapse
Affiliation(s)
- Yeo Kyu Hur
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea
| | - Hye Eun Lee
- Celros Biotech, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea
| | - Jung-Yeon Yoo
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea
| | - Young Nyun Park
- Department of Pathology Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-Gu, Seoul, 03722, South Korea
| | - In Hye Lee
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea.
| | - Yun Soo Bae
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea; Celros Biotech, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea.
| |
Collapse
|
10
|
Li H, Liang J, Han M, Gao Z. Polyphenols synergistic drugs to ameliorate non-alcoholic fatty liver disease via signal pathway and gut microbiota: A review. J Adv Res 2025; 68:43-62. [PMID: 38471648 PMCID: PMC11785558 DOI: 10.1016/j.jare.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver disease with an increasing incidence worldwide. Single drug therapy may have toxic side effects and disrupt gut microbiota balance. Polyphenols are widely used in disease intervention due to their distinctive nutritional properties and medicinal value, which a potential gut microbiota modulator. However, there is a lack of comprehensive review to explore the efficacy and mechanism of combined therapy with drugs and polyphenols for NAFLD. AIM OF REVIEW Based on this, this review firstly discusses the link between NAFLD and gut microbiota, and outlines the effects of polyphenols and drugs on gut microbiota. Secondly, it examined recent advances in the treatment and intervention of NAFLD with drugs and polyphenols and the therapeutic effect of the combination of the two. Finally, we highlight the underlying mechanisms of polyphenol combined drug therapy in NAFLD. This is mainly in terms of signaling pathways (NF-κB, AMPK, Nrf2, JAK/STAT, PPAR, SREBP-1c, PI3K/Akt and TLR) and gut microbiota. Furthermore, some emerging mechanisms such as microRNA potential biomarker therapies may provide therapeutic avenues for NAFLD. KEY SCIENTIFIC CONCEPTS OF REVIEW Drawing inspiration from combination drug strategies, the use of active substances in combination with drugs for NAFLD intervention holds transformative and prospective potential, both improve NAFLD and restore gut microbiota balance while reducing the required drug dosage. This review systematically discusses the bidirectional interactions between gut microbiota and NAFLD, and summarizes the potential mechanisms of polyphenol synergistic drugs in the treatment of NAFLD by modulating signaling pathways and gut microbiota. Future researches should develop multi-omics technology to identify patients who benefit from polyphenols combination drugs and devising individualized treatment plans to enhance its therapeutic effect.
Collapse
Affiliation(s)
- Hongcai Li
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Jingjing Liang
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Mengzhen Han
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China
| | - Zhenpeng Gao
- College of Food Science and Engineering, Northwest A&F University, 712100 Yangling, Shaanxi, People's Republic of China.
| |
Collapse
|
11
|
Caddeo A, Romeo S. Precision medicine and nucleotide-based therapeutics to treat steatotic liver disease. Clin Mol Hepatol 2025; 31:S76-S93. [PMID: 39103998 PMCID: PMC11925435 DOI: 10.3350/cmh.2024.0438] [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: 06/08/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 08/07/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a complex multifactorial disease and becoming the leading cause of liver-related morbidity and mortality. MASLD spans from isolated steatosis to metabolic dysfunction-associated steatohepatitis (MASH), that may progress to cirrhosis and hepatocellular carcinoma (HCC). Genetic, metabolic, and environmental factors strongly contribute to the heterogeneity of MASLD. Lifestyle intervention and weight loss represent a viable treatment for MASLD. Moreover, Resmetirom, a thyroid hormone beta receptor agonist, has recently been approved for MASLD treatment. However, most individuals treated did not respond to this therapeutic, suggesting the need for a more tailored approach to treat MASLD. Oligonucleotide-based therapies, namely small-interfering RNA (siRNA) and antisense oligonucleotide (ASO), have been recently developed to tackle MASLD by reducing the expression of genes influencing MASH progression, such as PNPLA3 and HSD17B13. Here, we review the latest progress made in the synthesis and development of oligonucleotide-based agents targeting genetic determinants of MASH.
Collapse
Affiliation(s)
- Andrea Caddeo
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Stefano Romeo
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Medicine, Endocrinology (H7) Karolinska Institute and Hospital, Huddinge, Stockholm, Sweden
| |
Collapse
|
12
|
Wang Y, Zhang K, Wang B, Yu B, Zhang Z, Yu Y, Yu Y, Sun Y, Chen Y, Zhang W, Cai Y, Xiang Q, Xia F, Wang N, Lu Y. Early-life famine exposure, genetic susceptibility and risk of MAFLD in adulthood. J Nutr Health Aging 2025; 29:100443. [PMID: 39667094 DOI: 10.1016/j.jnha.2024.100443] [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: 10/23/2023] [Revised: 06/02/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
OBJECTIVES Early-famine exposure was reported to be associated with metabolic associated fatty liver disease (MAFLD); however, it has not been fully elucidated whether the gene-famine interaction exist in this association. We aimed to investigate the association between early-life famine exposure in different genetic risk stratifications and the risk of MAFLD in adulthood. DESIGN, SETTING, PARTICIPANTS, AND MEASUREMENTS The study included 8213 participants from the SPECT-China study. Famine exposure subgroups was defined according to the birth year. A genetic risk score (GRS) was constructed with single nucleotide polymorphisms associated with MAFLD in East Asians. Logistic models were used to examine the association of famine exposure and GRS with MAFLD. RESULTS Early-life famine exposure was positively associated with MAFLD after adjusting for multiple confounders (OR (95% CI): fetal-exposure 1.3(1.11-1.53), childhood exposure 1.12(1-1.25)). Meanwhile, with per SD increment of GRS (2.49 points), the OR(95%CI) of MAFLD was 1.1(1.04-1.16). In high GRS group, fetal-exposure was positively associated with 45% higher risk of MAFLD (1.45(1.15-1.83)). In men, neither in low or high GRS subgroups observed an association between early-life famine exposure and MAFLD. But in women with high GRS of MAFLD, fetal-exposure was positively associated with even higher risk of MAFLD (1.64(1.22-2.22)). CONCLUSION The positive association between early-life famine exposure and MAFLD is intensified by high genetic susceptibility of MAFLD in women and in general population in China; while this association does not exist in men or in those with low genetic risk scores.
Collapse
Affiliation(s)
- Yuying Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Kun Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Bin Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Bowei Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Ziteng Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Yuetian Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Yuefeng Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Ying Sun
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Yi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Wen Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Yan Cai
- Department of Endocrinology, The Fifth Affiliated Hospital of Kunming Medical University, Yunnan Honghe Prefecture Central Hospital (Ge Jiu People's Hospital), Yunnan, China
| | - Qian Xiang
- Department of Endocrinology, The Fifth Affiliated Hospital of Kunming Medical University, Yunnan Honghe Prefecture Central Hospital (Ge Jiu People's Hospital), Yunnan, China
| | - Fangzhen Xia
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China.
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China.
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China.
| |
Collapse
|
13
|
Hamed AM, Elbahy DA, Ahmed ARH, Thabet SA, Refaei RA, Ragab I, Elmahdy SM, Osman AS, Abouelella AMA. Comparison of the efficacy of curcumin and its nano formulation on dexamethasone-induced hepatic steatosis, dyslipidemia, and hyperglycemia in Wistar rats. Heliyon 2024; 10:e41043. [PMID: 39759349 PMCID: PMC11696662 DOI: 10.1016/j.heliyon.2024.e41043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/07/2025] Open
Abstract
Background and objective Insulin resistance is a primary feature of type 2 diabetes. This study compared the effects of curcumin and its nanoformulation on insulin resistance, fasting blood sugar, liver function, GLUT4, lipid profile, and oxidative stress in the liver and pancreas in a diabetic model. Methods Thirty male Wistar rats were divided into five groups: a control group, a diabetic group, a diabetic group treated with metformin (40 mg/kg), a diabetic group treated with curcumin (100 mg/kg), and a diabetic group treated with curcumin NPs (100 mg/kg). Diabetes was induced by injecting dexamethasone daily for 14 days. Treatment with curcumin and curcumin NPs was administered by gavage for 14 days. Body weight and fasting blood sugar levels were measured on days 1, 14, and 28. Results The metformin, curcumin, and curcumin NPs groups showed significantly greater body weight gain than the untreated diabetic group (P < 0.001). In diabetic rats treated with curcumin and curcumin NPs, insulin resistance decreased by approximately 40 %, while fasting blood sugar levels dropped by 35-40 % (P < 0.001). The levels of liver enzymes (AST, ALT), cholesterol, triglycerides, LDL, and the oxidative stress marker MDA in liver and pancreatic tissues were reduced by 30-50 %. Additionally, beneficial markers such as albumin, HDL, antioxidants (GSH, SOD), and GLUT4 levels were increased by 25-45 % (P < 0.001). Nano-curcumin consistently showed greater improvements than curcumin, especially in reducing oxidative stress and supporting liver and pancreatic health. Conclusion This study demonstrates that curcumin NPs has a superior effect on reducing oxidative stress and improving metabolic parameters in diabetes compared to curcumin. by enhancing the bioavailability and stability of curcumin, the nanoformulation showed stronger therapeutic potential for managing high blood sugar, cholesterol issues, and liver health, positioning curcumin NPs as a promising alternative to conventional treatments for diabetes and its complications.
Collapse
Affiliation(s)
- Amany M. Hamed
- Chemistry Department, Faculty of Science, Sohag University, Sohag, Egypt
| | - Dalia A. Elbahy
- Department of Clinical Pharmacology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Ahmed RH. Ahmed
- Department of Pathology, faculty of medicine, Sohag University, Sohag, Egypt
| | - Shymaa A. Thabet
- Central Research Center, Faculty of Medicine, Sohag University, Sohag, Egypt
| | | | - Islam Ragab
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| | | | - Ahmed S. Osman
- Department of Biochemistry, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Azza MA. Abouelella
- Department of Clinical Pharmacology, Faculty of Medicine, Sohag University, Sohag, Egypt
| |
Collapse
|
14
|
Giardoglou P, Gavra I, Amanatidou AI, Kalafati IP, Symianakis P, Kafyra M, Moulos P, Dedoussis GV. Development of a Polygenic Risk Score for Metabolic Dysfunction-Associated Steatotic Liver Disease Prediction in UK Biobank. Genes (Basel) 2024; 16:33. [PMID: 39858580 PMCID: PMC11765347 DOI: 10.3390/genes16010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) is the leading cause of liver-related morbidity and mortality. Although the invasive liver biopsy remains the golden standard for MASLD diagnosis, Magnetic Resonance Imaging-derived Proton Density Fat Fraction (MRI-PDFF) is an accurate, non-invasive method for the assessment of treatment response. This study aimed at developing a Polygenic Risk Score (PRS) to improve MRI-PDFF prediction using UK Biobank data to assess an individual's genetic liability to MASLD. METHODS We iteratively sequestered 10% of MRI-PDFF samples as a validation set and split the rest of each dataset into base and target partitions, containing GWAS summary statistics and raw genotype data, respectively. PRSice2 was deployed to derive PRS candidates. Based on the frequency of SNP appearances along the PRS candidates, we generated different SNP sets according to variable frequency cutoffs. By applying the PRSs to the validation set, we identified the optimal SNP set, which was then applied to a Greek nonalcoholic fatty liver disease (NAFLD) study. RESULTS Data from 3553 UK Biobank participants yielded 49 different SNP sets. After calculating the PRS on the validation set for every SNP set, an optimal PRS with 75 SNPs was selected (incremental R2 = 0.025, p-value = 0.00145). Interestingly, 43 SNPs were successfully mapped to MASLD-related known genes. The selected PRS could predict traits, like LDL cholesterol and diastolic blood pressure in the UK Biobank, as also disease outcome in the Greek NAFLD study. CONCLUSIONS Our findings provide strong evidence that PRS is a powerful prediction model for MASLD, while it can also be applied on populations of different ethnicity.
Collapse
Affiliation(s)
- Panagiota Giardoglou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Ioanna Gavra
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Athina I. Amanatidou
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Ioanna Panagiota Kalafati
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
- Department of Nutrition and Dietetics, School of Physical Education, Sport Science and Dietetics, University of Thessaly, 42132 Trikala, Greece
| | - Panagiotis Symianakis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Maria Kafyra
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| | - Panagiotis Moulos
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center ‘Alexander Fleming’, 16672 Vari, Greece;
| | - George V. Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, 17671 Athens, Greece; (P.G.); (M.K.)
| |
Collapse
|
15
|
Ahmed A, Cule M, Bell JD, Sattar N, Yaghootkar H. Differing genetic variants associated with liver fat and their contrasting relationships with cardiovascular diseases and cancer. J Hepatol 2024; 81:921-929. [PMID: 38960375 DOI: 10.1016/j.jhep.2024.06.030] [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: 11/22/2023] [Revised: 06/14/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND & AIMS The mechanisms underlying the association of steatotic liver disease with cardiovascular and cancer outcomes are poorly understood. We aimed to use MRI-derived measures of liver fat and genetics to investigate causal mechanisms that link higher liver fat to various health outcomes. METHODS We conducted a genome-wide association study on 37,358 UK Biobank participants to identify genetic variants associated with liver fat measured from MRI scans. We used a Mendelian randomisation approach to investigate the causal effect of liver fat on health outcomes independent of BMI, alcohol consumption and lipids using data from published genome-wide association studies and FinnGen. RESULTS We identified 13 genetic variants associated with liver fat that had differing effects on the risks of health outcomes. Genetic variants associated with impaired hepatic triglyceride export showed liver fat-increasing alleles to be correlated with a reduced risk of coronary artery disease and myocardial infarction but an elevated risk of type 2 diabetes, while variants associated with enhanced de novo lipogenesis showed liver fat-increasing alleles to be linked to a higher risk of myocardial infarction and coronary artery disease. Genetically higher liver fat content increased the risk of non-alcohol-related cirrhosis, hepatocellular carcinoma, and intrahepatic bile duct and gallbladder cancers, exhibiting a dose-dependent relationship, irrespective of the mechanism. CONCLUSION This study provides fresh insight into the heterogeneous effect of liver fat on health outcomes. It challenges the notion that liver fat per se is an independent risk factor for cardiovascular disease, underscoring the dependency of this association on the specific mechanisms that drive fat accumulation in the liver. However, excess liver fat, regardless of the underlying mechanism, appears to be causally linked to cirrhosis and cancers in a dose-dependent manner. IMPACT AND IMPLICATION This research advances our understanding of the heterogeneity in mechanisms influencing liver fat accumulation, providing new insights into how liver fat accumulation may impact various health outcomes. The findings challenge the notion that liver fat is an independent risk factor for cardiovascular disease and highlight the mechanistic effect of some genetic variants on fat accumulation and the development of cardiovascular diseases. This study is of particular importance for healthcare professionals including physicians and researchers, as well as patients, as it allows for more targeted and personalised treatment by understanding the relationship between liver fat and various health outcomes. The findings emphasise the need for a personalised management approach and a reshaping of risk assessment criteria. It also provides room for prioritising a clinical intervention aimed at reducing liver fat content (likely via intentional weight loss) that could help protect against liver-related fibrosis and cancer.
Collapse
Affiliation(s)
- Altayeb Ahmed
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, UK
| | | | - Jimmy D Bell
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Hanieh Yaghootkar
- Joseph Banks Laboratories, College of Health and Science, University of Lincoln, Lincoln, UK.
| |
Collapse
|
16
|
Jamialahmadi O, De Vincentis A, Tavaglione F, Malvestiti F, Li-Gao R, Mancina RM, Alvarez M, Gelev K, Maurotti S, Vespasiani-Gentilucci U, Rosendaal FR, Kozlitina J, Pajukanta P, Pattou F, Valenti L, Romeo S. Partitioned polygenic risk scores identify distinct types of metabolic dysfunction-associated steatotic liver disease. Nat Med 2024; 30:3614-3623. [PMID: 39653778 PMCID: PMC11645285 DOI: 10.1038/s41591-024-03284-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 08/30/2024] [Indexed: 12/15/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by an excess of lipids, mainly triglycerides, in the liver and components of the metabolic syndrome, which can lead to cirrhosis and liver cancer. While there is solid epidemiological evidence that MASLD clusters with cardiometabolic disease, several leading genetic risk factors for MASLD do not increase the risk of cardiovascular disease, suggesting no causal relationship between MASLD and cardiometabolic derangement. In this work, we leveraged measurements of visceral adiposity identifying 27 previously unknown genetic loci associated with MASLD (n = 36,394), six replicated in four independent cohorts (n = 3,903). Next, we generated two partitioned polygenic risk scores based on the presence of lipoprotein retention in the liver. The two polygenic risk scores suggest the presence of at least two distinct types of MASLD, one confined to the liver resulting in a more aggressive liver disease and one that is systemic and results in a higher risk of cardiometabolic disease. These findings shed light on the heterogeneity of MASLD and have the potential to improve the prediction of clinical trajectories and inform precision medicine approaches.
Collapse
Grants
- 777377 EC | Horizon 2020 Framework Programme (EU Framework Programme for Research and Innovation H2020)
- 22 2270 Pj Cancerfonden (Swedish Cancer Society)
- R01 DK132775 NIDDK NIH HHS
- 2023-02079 Vetenskapsrådet (Swedish Research Council)
- R01 HG010505 NHGRI NIH HHS
- R01 HL170604 NHLBI NIH HHS
- the Swedish state under the Agreement between the Swedish government and the county councils (the ALF agreement, ALFGBG-965360); Swedish Heart Lung Foundation (20220334); Wallenberg Academy Fellows from the Knut and Alice Wallenberg Foundation (KAW 2017.0203); Novonordisk Distinguished Investigator Grant - Endocrinology and Metabolism (NNF23OC0082114; Novonordisk Project grants in Endocrinology and Metabolism (NNF20OC0063883).
- NIH grants R01HG010505, R01DK132775, and R01HL170604
- Italian Ministry of Health (Ministero della Salute), Ricerca Finalizzata 2016, RF-2016-02364358; Italian Ministry of Health, Ricerca Finalizzata 2021 (TERS) RF-2021-12373889; Italian Ministry of Health (national coordinator) (2023-2026) Ricerca Finalizzata PNRR 2022 (PNRR-MAD-2022-12375656); Italian Ministry of Health (Ministero della Salute), Rete Cardiologica “CV-PREVITAL”; Fondazione Patrimonio Ca’ Granda, “Liver BIBLE” (PR-0361); The European Union, H2020-ICT-2018-20/H2020-ICT-2020-2 programme “Photonics” under grant agreement No. 101016726-REVEAL,Gilead_IN-IT-989-5790;The European Union, HORIZON-MISS-2021-CANCER-02-03 programme “Genial” under grant agreement “101096312#x201D;; Italian Ministry of University and Research, PNRR – M4 - C2 “di R&S su alcune Key Enabling Technologies” “National Center for Gene Therapy and Drugs based on RNA Technology” CN3 Spoke 4, group ASSET: A sex-specific approach to NAFLD targeting.
Collapse
Affiliation(s)
- Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden.
| | - Antonio De Vincentis
- Operative Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Internal Medicine, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Federica Tavaglione
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Malvestiti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rosellina M Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Rome, Italy
| | - Marcus Alvarez
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Kyla Gelev
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Samantha Maurotti
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Umberto Vespasiani-Gentilucci
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Frits Richard Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Julia Kozlitina
- The Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
- Institute for Precision Health, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - François Pattou
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France
- European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Precision Medicine - Biological Resource Center, Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden.
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.
- Department of Medicine (H7), Karolinska Institute, Huddinge, Stockholm, Sweden.
- Department of Endocrinology, Karolinska University Hospital, Huddinge, Stockholm, Sweden.
| |
Collapse
|
17
|
Xue H, Wang L, Wu Y, Liu X, Jiang J, Chan SO, Chen X, Ling W, Yu C. Association of moderate alcohol intake with the risks of cirrhosis and steatotic liver disease: Results from a large population-based cohort study. Clin Nutr 2024; 43:75-83. [PMID: 39427474 DOI: 10.1016/j.clnu.2024.10.014] [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: 07/30/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND&AIMS There is uncertainty about the associations between moderate alcohol consumption and liver-related outcomes. We aimed to explore the associations of moderate drinking with cirrhosis, steatotic liver disease (SLD), and liver cancer in a large cohort study. METHODS A total of 215,559 non-drinkers and moderate drinkers (<20 g/day alcohol for females or < 30 g/day for males) were enrolled between 2006 and 2010 and followed up to 2022. The primary outcome is incident cirrhosis, and the secondary outcomes are the incidence of steatotic liver disease and liver cancer. Hazard ratios (HRs) and 95 % confidence intervals (CIs) were calculated for liver-related outcomes in relation to moderate drinkers, as well as the quantity and type of their alcohol intake. All analyses were stratified by sex. RESULTS A total of 705 cirrhosis, 2010 SLD, and 350 liver cancer cases were documented during a median follow-up period of 12.7 years. Compared with non-drinkers, moderate drinkers had a lower risk of SLD (HR: 0.77; 95 % CI: 0.66, 0.89). Among the moderate drinkers, alcohol intake [per standard deviation (SD) increment] was associated with an increased risk of incident cirrhosis (HR: 1.11; 95 % CI: 1.02, 1.20), but the association was attenuated after restricting alcohol intake to no more than 16 g/day. Wine consumption (per SD increment of the percentage of wine consumption of total alcohol intake) had an inverse association with incident cirrhosis and SLD (HR: 0.82; 95 % CI: 0.75, 0.89 for cirrhosis; HR: 0.91; 95 % CI: 0.87, 0.96 for SLD). The inverse associations between moderate wine use and SLD were likely to be sex-dependent (P for interaction = 0.01). CONCLUSIONS The excessive alcohol threshold of 30 g/day for males may be set high for liver health. Further work is needed to make sex-specific recommendations on moderate drinking for liver health.
Collapse
Affiliation(s)
- Hongliang Xue
- Department of Nutrition, School of Public Health, Guangzhou Medical University, Guangzhou, China; The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Liqing Wang
- Department of Neurology, Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuankai Wu
- Department of Infectious Diseases, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xinyu Liu
- Department of Nutrition, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Jingcheng Jiang
- Department of Intergrative Physiology, University of Colorado, Boulder, USA
| | - Sun On Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Xu Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China; Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, USA; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China.
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| | - Chao Yu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China; Medical Examination Center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
18
|
Raverdy V, Tavaglione F, Chatelain E, Lassailly G, De Vincentis A, Vespasiani-Gentilucci U, Qadri SF, Caiazzo R, Verkindt H, Saponaro C, Kerr-Conte J, Baud G, Marciniak C, Chetboun M, Oukhouya-Daoud N, Blanck S, Vandel J, Olsson L, Chakaroun R, Gnemmi V, Leteurtre E, Lefebvre P, Haas JT, Yki-Järvinen H, Francque S, Staels B, Le Roux CW, Tremaroli V, Mathurin P, Marot G, Romeo S, Pattou F. Data-driven cluster analysis identifies distinct types of metabolic dysfunction-associated steatotic liver disease. Nat Med 2024; 30:3624-3633. [PMID: 39653777 PMCID: PMC11645276 DOI: 10.1038/s41591-024-03283-1] [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: 01/29/2024] [Accepted: 08/30/2024] [Indexed: 12/15/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) exhibits considerable variability in clinical outcomes. Identifying specific phenotypic profiles within MASLD is essential for developing targeted therapeutic strategies. Here we investigated the heterogeneity of MASLD using partitioning around medoids clustering based on six simple clinical variables in a cohort of 1,389 individuals living with obesity. The identified clusters were applied across three independent MASLD cohorts with liver biopsy (totaling 1,099 participants), and in the UK Biobank to assess the incidence of chronic liver disease, cardiovascular disease and type 2 diabetes. Results unveiled two distinct types of MASLD associated with steatohepatitis on histology and liver imaging. The first cluster, liver-specific, was genetically linked and showed rapid progression of chronic liver disease but limited risk of cardiovascular disease. The second cluster, cardiometabolic, was primarily associated with dysglycemia and high levels of triglycerides, leading to a similar incidence of chronic liver disease but a higher risk of cardiovascular disease and type 2 diabetes. Analyses of samples from 831 individuals with available liver transcriptomics and 1,322 with available plasma metabolomics highlighted that these two types of MASLD exhibited distinct liver transcriptomic profiles and plasma metabolomic signatures, respectively. In conclusion, these data provide preliminary evidence of the existence of two distinct types of clinically relevant MASLD with similar liver phenotypes at baseline, but each with specific underlying biological profiles and different clinical trajectories, suggesting the need for tailored therapeutic strategies.
Collapse
Affiliation(s)
- Violeta Raverdy
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Federica Tavaglione
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Estelle Chatelain
- US 41 - UAR 2014 - PLBS Bilille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | - Guillaume Lassailly
- Department of Hepato-Gastroenterology CHU Lille, University of Lille, Inserm INFINITE-U1286, Lille, France
| | - Antonio De Vincentis
- Operative Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Internal Medicine, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Umberto Vespasiani-Gentilucci
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Sami F Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Robert Caiazzo
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Helene Verkindt
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Chiara Saponaro
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
| | - Julie Kerr-Conte
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
| | - Gregory Baud
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Camille Marciniak
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Mikael Chetboun
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Naima Oukhouya-Daoud
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Samuel Blanck
- ULR 2694 METRICS: Évaluation des technologies de santé et des pratiques médicales, University of Lille, CHU Lille, F-59000, Lille, France
| | - Jimmy Vandel
- US 41 - UAR 2014 - PLBS Bilille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | - Lisa Olsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rima Chakaroun
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Viviane Gnemmi
- Cancer Heterogeneity Plasticity and Resistance to Therapies, CANTHER-UMR9020-U1277 - CNRS, Inserm, CHU Lille, University of Lille, Lille, France
- Department of Pathology, CHU Lille, University of Lille, Lille, France
| | - Emmanuelle Leteurtre
- Cancer Heterogeneity Plasticity and Resistance to Therapies, CANTHER-UMR9020-U1277 - CNRS, Inserm, CHU Lille, University of Lille, Lille, France
- Department of Pathology, CHU Lille, University of Lille, Lille, France
| | - Philippe Lefebvre
- Nuclear Receptors, Metabolic and Cardiovascular Diseases - U1011, University of Lille, Inserm, CHU Lille, Institut Pasteur Lille, Lille, France
| | - Joel T Haas
- Nuclear Receptors, Metabolic and Cardiovascular Diseases - U1011, University of Lille, Inserm, CHU Lille, Institut Pasteur Lille, Lille, France
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Edegem, Belgium
- InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Bart Staels
- Nuclear Receptors, Metabolic and Cardiovascular Diseases - U1011, University of Lille, Inserm, CHU Lille, Institut Pasteur Lille, Lille, France
| | - Carel W Le Roux
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - Valentina Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Philippe Mathurin
- Department of Hepato-Gastroenterology CHU Lille, University of Lille, Inserm INFINITE-U1286, Lille, France
| | - Guillemette Marot
- ULR 2694 METRICS: Évaluation des technologies de santé et des pratiques médicales, University of Lille, CHU Lille, F-59000, Lille, France
- MODAL: Models for Data Analysis and Learning, Inria, F-59000, Lille, France
| | - Stefano Romeo
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Department of Medicine Huddinge (H7), Karolinska Institutet and University Hospital, Stockholm, Sweden.
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden.
| | - François Pattou
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France.
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France.
| |
Collapse
|
19
|
Silva-Luis CC, Lopes MS, Gomes SM, Cantalice Matias PK, Brandini FP, Costa PCT, de Moraes RCS, Baccin Martins VJ, de Brito Alves JL. Ultra-Processed Food Consumption and Cardiometabolic Risk Factors in Children Living in Northeastern Brazil. Nutrients 2024; 16:3944. [PMID: 39599729 PMCID: PMC11597345 DOI: 10.3390/nu16223944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVE To analyze the association between ultra-processed food (UPF) consumption and cardiometabolic, biochemical, and inflammatory risk factors in children in a metropolis in Northeast Brazil. METHODS A cross-sectional study using baseline data from a community-based controlled trial was carried out with 151 children from public schools in João Pessoa, Paraíba, Brazil aged 7 to 10 years. Dietary consumption was assessed using 24 h food recall, and UPF consumption was estimated using the NOVA classification system. Anthropometry (BMI for age), blood pressure, biochemical parameters (ALT, AST, GGT, cholesterol, LDL-c, HDL-c, triglycerides, fasting glucose, HbA1c, HOMA-IR, creatinine, urea, hs-CRP), and cytokines (IL-2, IL-4, IL-6, IL-10, IL-17a, IFN-γ, and TNF-α) were also assessed. RESULTS Children in the third tertile (highest UPF consumption) had higher serum concentrations of LDL-c (p-value = 0.04) and ALT (p-value = 0.01), with a trend towards higher AST (p-value = 0.06). Total energy (p-value = 0.01), trans fatty acid (p-value = 0.02), and sodium (p-value = 0.04) intakes were higher in the highest tertile, whereas protein (p-value < 0.01) and fiber (p-value < 0.01) intakes were lower. Concentrations of IL-17A (p-value = 0.01) and IL-10 (p-value = 0.04) were significantly higher in the second tertile. Multiple linear regression showed that UPF consumption was significantly associated with increased LDL-c, ALT, and AST concentrations. CONCLUSIONS High intake of UPFs was associated with dyslipidemia, elevated liver enzymes, and inflammatory changes in children. Dietary interventions are needed to reduce UPF consumption and prevent cardiometabolic and liver disease in childhood.
Collapse
Affiliation(s)
- Cristiane Cosmo Silva-Luis
- Health Sciences Center, Department of Nutrition, Campus I, Federal University of Paraíba, João Pessoa 58059-900, PB, Brazil; (C.C.S.-L.); (M.S.L.); (S.M.G.); (P.K.C.M.); (F.P.B.); (P.C.T.C.); (R.C.S.d.M.)
| | - Mariana Souza Lopes
- Health Sciences Center, Department of Nutrition, Campus I, Federal University of Paraíba, João Pessoa 58059-900, PB, Brazil; (C.C.S.-L.); (M.S.L.); (S.M.G.); (P.K.C.M.); (F.P.B.); (P.C.T.C.); (R.C.S.d.M.)
| | - Sávio Marcelino Gomes
- Health Sciences Center, Department of Nutrition, Campus I, Federal University of Paraíba, João Pessoa 58059-900, PB, Brazil; (C.C.S.-L.); (M.S.L.); (S.M.G.); (P.K.C.M.); (F.P.B.); (P.C.T.C.); (R.C.S.d.M.)
| | - Palloma Karlla Cantalice Matias
- Health Sciences Center, Department of Nutrition, Campus I, Federal University of Paraíba, João Pessoa 58059-900, PB, Brazil; (C.C.S.-L.); (M.S.L.); (S.M.G.); (P.K.C.M.); (F.P.B.); (P.C.T.C.); (R.C.S.d.M.)
| | - Fernando Paiva Brandini
- Health Sciences Center, Department of Nutrition, Campus I, Federal University of Paraíba, João Pessoa 58059-900, PB, Brazil; (C.C.S.-L.); (M.S.L.); (S.M.G.); (P.K.C.M.); (F.P.B.); (P.C.T.C.); (R.C.S.d.M.)
| | - Paulo César Trindade Costa
- Health Sciences Center, Department of Nutrition, Campus I, Federal University of Paraíba, João Pessoa 58059-900, PB, Brazil; (C.C.S.-L.); (M.S.L.); (S.M.G.); (P.K.C.M.); (F.P.B.); (P.C.T.C.); (R.C.S.d.M.)
| | - Rúbia Cartaxo Squizato de Moraes
- Health Sciences Center, Department of Nutrition, Campus I, Federal University of Paraíba, João Pessoa 58059-900, PB, Brazil; (C.C.S.-L.); (M.S.L.); (S.M.G.); (P.K.C.M.); (F.P.B.); (P.C.T.C.); (R.C.S.d.M.)
| | - Vinícius José Baccin Martins
- Health Sciences Center, Department of Biomedicine Science, Campus I, Federal University of Paraíba, João Pessoa 58059-900, PB, Brazil;
| | - José Luiz de Brito Alves
- Health Sciences Center, Department of Nutrition, Campus I, Federal University of Paraíba, João Pessoa 58059-900, PB, Brazil; (C.C.S.-L.); (M.S.L.); (S.M.G.); (P.K.C.M.); (F.P.B.); (P.C.T.C.); (R.C.S.d.M.)
| |
Collapse
|
20
|
Markowska J, Kasprzak-Drozd K, Niziński P, Dragan M, Kondracka A, Gondek E, Oniszczuk T, Oniszczuk A. Quercetin: A Promising Candidate for the Management of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Molecules 2024; 29:5245. [PMID: 39598636 PMCID: PMC11596905 DOI: 10.3390/molecules29225245] [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: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents a chronic liver disease. The development of MASLD is influenced by a multitude of diseases associated with modern lifestyles, including but not limited to diabetes mellitus, hypertension, hyperlipidaemia and obesity. These conditions are often consequences of the adoption of unhealthy habits, namely a sedentary lifestyle, a lack of physical activity, poor dietary choices and excessive alcohol consumption. The treatment of MASLD is primarily based on modifying the patient's lifestyle and pharmacological intervention. Despite the absence of FDA-approved pharmacological agents for the treatment of MASLD, several potential therapeutic modalities have demonstrated efficacy in reversing the histopathological features of the disease. Among the botanical ingredients belonging to the flavonoid group is quercetin (QE). QE has been demonstrated to possess a number of beneficial physiological effects, including anti-inflammatory, anticancer and antifungal properties. Additionally, it functions as a natural antioxidant. Preclinical evidence indicates that QE may play a beneficial role in reducing liver damage and improving metabolic health. Early human studies also suggest that QE may be an effective treatment for MASLD due to its antioxidant, anti-inflammatory, and lipid-regulating properties. This review aims to summarize the available information on the therapeutic effects of QE in MASLD.
Collapse
Affiliation(s)
- Julia Markowska
- Science Circle of the Department of Inorganic Chemistry, Medical University of Lublin, Dr. Witolda Chodźki 4a, 20-093 Lublin, Poland; (J.M.); (M.D.)
| | - Kamila Kasprzak-Drozd
- Department of Inorganic Chemistry, Medical University of Lublin, Dr. Witolda Chodźki 4a, 20-093 Lublin, Poland;
| | - Przemysław Niziński
- Department of Pharmacology, Medical University of Lublin, Radziwiłłowska 11, 20-080 Lublin, Poland;
| | - Magdalena Dragan
- Science Circle of the Department of Inorganic Chemistry, Medical University of Lublin, Dr. Witolda Chodźki 4a, 20-093 Lublin, Poland; (J.M.); (M.D.)
| | - Adrianna Kondracka
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Ewa Gondek
- Department of Food Engineering and Process Management, Institute of Food Science, Warsaw University of Life Sciences, Nowoursynowska 159C, 02-776 Warsaw, Poland
| | - Tomasz Oniszczuk
- Department of Thermal Technology and Food Process Engineering, University of Life Sciences in Lublin, Głęboka 31, 20-612 Lublin, Poland;
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, Dr. Witolda Chodźki 4a, 20-093 Lublin, Poland;
| |
Collapse
|
21
|
Sookoian S, Rotman Y, Valenti L. Genetics of Metabolic Dysfunction-associated Steatotic Liver Disease: The State of the Art Update. Clin Gastroenterol Hepatol 2024; 22:2177-2187.e3. [PMID: 39094912 PMCID: PMC11512675 DOI: 10.1016/j.cgh.2024.05.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/18/2024] [Accepted: 05/28/2024] [Indexed: 08/04/2024]
Abstract
Recent advances in the genetics of metabolic dysfunction-associated steatotic liver disease (MASLD) are gradually revealing the mechanisms underlying the heterogeneity of the disease and have shown promising results in patient stratification. Genetic characterization of the disease has been rapidly developed using genome-wide association studies, exome-wide association studies, phenome-wide association studies, and whole exome sequencing. These advances have been powered by the increase in computational power, the development of new analytical algorithms, including some based on artificial intelligence, and the recruitment of large and well-phenotyped cohorts. This review presents an update on genetic studies that emphasize new biological insights from next-generation sequencing approaches. Additionally, we discuss innovative methods for discovering new genetic loci for MASLD, including rare variants. To comprehensively manage MASLD, it is important to stratify risks. Therefore, we present an update on phenome-wide association study associations, including extreme phenotypes. Additionally, we discuss whether polygenic risk scores and targeted sequencing are ready for clinical use. With particular focus on precision medicine, we introduce concepts such as the interplay between genetics and the environment in modulating genetic risk with lifestyle or standard therapies. A special chapter is dedicated to gene-based therapeutics. The limitations of approved pharmacological approaches are discussed, and the potential of gene-related mechanisms in therapeutic development is reviewed, including the decision to perform genetic testing in patients with MASLD.
Collapse
Affiliation(s)
- Silvia Sookoian
- Clinical and Molecular Hepatology. Translational Health Research Center (CENITRES). Maimónides University. Buenos Aires, Argentina
- Faculty of Health Science. Maimónides University. Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - 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
| | - Luca Valenti
- Precision Medicine - Biological Resource Center, Department of Transfusion Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
22
|
Reid MV, Fredickson G, Mashek DG. Mechanisms coupling lipid droplets to MASLD pathophysiology. Hepatology 2024:01515467-990000000-01067. [PMID: 39475114 DOI: 10.1097/hep.0000000000001141] [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: 08/15/2024] [Accepted: 10/17/2024] [Indexed: 01/03/2025]
Abstract
Hepatic steatosis, the buildup of neutral lipids in lipid droplets (LDs), is commonly referred to as metabolic dysfunction-associated steatotic liver disease when alcohol or viral infections are not involved. Metabolic dysfunction-associated steatotic liver disease encompasses simple steatosis and the more severe metabolic dysfunction-associated steatohepatitis, characterized by inflammation, hepatocyte injury, and fibrosis. Previously viewed as inert markers of disease, LDs are now understood to play active roles in disease etiology and have significant nonpathological and pathological functions in cell signaling and function. These dynamic properties of LDs are tightly regulated by hundreds of proteins that coat the LD surface, controlling lipid metabolism, trafficking, and signaling. The following review highlights various facets of LD biology with the primary goal of discussing key mechanisms through which LDs promote the development of advanced liver diseases, including metabolic dysfunction-associated steatohepatitis.
Collapse
Affiliation(s)
- Mari V Reid
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gavin Fredickson
- Department of Integrated Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Douglas G Mashek
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
23
|
Moretti V, Romeo S, Valenti L. The contribution of genetics and epigenetics to MAFLD susceptibility. Hepatol Int 2024; 18:848-860. [PMID: 38662298 PMCID: PMC11450136 DOI: 10.1007/s12072-024-10667-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/25/2024] [Indexed: 04/26/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is the most common liver disease worldwide. The risk of developing MAFLD varies among individuals, due to a combination of environmental inherited and acquired genetic factors. Genome-wide association and next-generation sequencing studies are leading to the discovery of the common and rare genetic determinants of MAFLD. Thanks to the great advances in genomic technologies and bioinformatics analysis, genetic and epigenetic factors involved in the disease can be used to develop genetic risk scores specific for liver-related complications, which can improve risk stratification. Genetic and epigenetic factors lead to the identification of specific sub-phenotypes of MAFLD, and predict the individual response to a pharmacological therapy. Moreover, the variant transcripts and protein themselves represent new therapeutic targets. This review will discuss the current status of research into genetic as well as epigenetic modifiers of MAFLD development and progression.
Collapse
Affiliation(s)
- Vittoria Moretti
- Precision Medicine Lab, Biological Resource Center and Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Via F Sforza 35, 20122, Milan, Italy
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Luca Valenti
- Precision Medicine Lab, Biological Resource Center and Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Via F Sforza 35, 20122, Milan, Italy.
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
24
|
Tacke F, Horn P, Wai-Sun Wong V, Ratziu V, Bugianesi E, Francque S, Zelber-Sagi S, Valenti L, Roden M, Schick F, Yki-Järvinen H, Gastaldelli A, Vettor R, Frühbeck G, Dicker D. EASL-EASD-EASO Clinical Practice Guidelines on the management of metabolic dysfunction-associated steatotic liver disease (MASLD). J Hepatol 2024; 81:492-542. [PMID: 38851997 DOI: 10.1016/j.jhep.2024.04.031] [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: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 06/10/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed non-alcoholic fatty liver disease (NAFLD), is defined as steatotic liver disease (SLD) in the presence of one or more cardiometabolic risk factor(s) and the absence of harmful alcohol intake. The spectrum of MASLD includes steatosis, metabolic dysfunction-associated steatohepatitis (MASH, previously NASH), fibrosis, cirrhosis and MASH-related hepatocellular carcinoma (HCC). This joint EASL-EASD-EASO guideline provides an update on definitions, prevention, screening, diagnosis and treatment for MASLD. Case-finding strategies for MASLD with liver fibrosis, using non-invasive tests, should be applied in individuals with cardiometabolic risk factors, abnormal liver enzymes, and/or radiological signs of hepatic steatosis, particularly in the presence of type 2 diabetes (T2D) or obesity with additional metabolic risk factor(s). A stepwise approach using blood-based scores (such as FIB-4) and, sequentially, imaging techniques (such as transient elastography) is suitable to rule-out/in advanced fibrosis, which is predictive of liver-related outcomes. In adults with MASLD, lifestyle modification - including weight loss, dietary changes, physical exercise and discouraging alcohol consumption - as well as optimal management of comorbidities - including use of incretin-based therapies (e.g. semaglutide, tirzepatide) for T2D or obesity, if indicated - is advised. Bariatric surgery is also an option in individuals with MASLD and obesity. If locally approved and dependent on the label, adults with non-cirrhotic MASH and significant liver fibrosis (stage ≥2) should be considered for a MASH-targeted treatment with resmetirom, which demonstrated histological effectiveness on steatohepatitis and fibrosis with an acceptable safety and tolerability profile. No MASH-targeted pharmacotherapy can currently be recommended for the cirrhotic stage. Management of MASH-related cirrhosis includes adaptations of metabolic drugs, nutritional counselling, surveillance for portal hypertension and HCC, as well as liver transplantation in decompensated cirrhosis.
Collapse
|
25
|
Zhan H, Nong X, Zhu S, Luo T, Li T, Cao M, Li Q, He Z, Hu J, Liu X. Clinical value of γ-glutamyl transpeptidase to platelet ratio and triglyceride measurement in the diagnosis of nonalcoholic fatty liver disease: A cross-sectional study. Heliyon 2024; 10:e36193. [PMID: 39224338 PMCID: PMC11367536 DOI: 10.1016/j.heliyon.2024.e36193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Objective In clinical practice, there are few effective biomarkers for identifying non-alcoholic fatty liver disease (NAFLD). The aim of this study is to investigate the diagnostic value of γ-glutamyl transpeptidase to platelet ratio (GPR) combined with triglyceride (TG) in NAFLD. Methods A total of 14,415 individuals participated in the annual physical examination. Multivariate logistic regression analysis was conducted to investigate the exposure factors associated with NAFLD. Spearman's analysis was performed to assess the correlation among the exposure factors of NAFLD. Furthermore, the diagnostic efficacy of the combination of GPR and TG in NAFLD was analyzed using the receiver operating characteristic curve (ROC). Results The results of the multivariate logistic regression analysis showed that BMI (OR = 1.619), Systolic Blood Pressure (SBP) (OR = 1.014), Diastolic Blood Pressure (DBP) (OR = 1.028), GPR (OR = 12.809), and TG (OR = 2.936) were all risk factors for NAFLD, while HDL-C (OR = 0.215) was a protective factor. Spearman correlation analysis revealed significant positive correlations between GPR and SBP, DBP, BMI, TG (p < 0.01), but a negative correlation between GPR and HDL-C (p < 0.01). TG was only positively correlated with GPR (p < 0.001). ROC curve analysis demonstrated that the area under the curve (AUC) of GPR combined with TG for diagnosis of NAFLD was 0.855 (95 % CI: 0.819-0.891), sensitivity was 83.45 % and specificity was 73.56 %. Conclusion This study indicated that high levels of GPR and TG were risk factors for NAFLD and demonstrated good clinical value in diagnosing NAFLD.
Collapse
Affiliation(s)
- Haohong Zhan
- Department of Emergency Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoli Nong
- Department of Ultrasound, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Senzhi Zhu
- Department of Emergency Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ting Luo
- Department of Emergency Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Mingjing Cao
- Department of Emergency Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qi Li
- Department of Emergency Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhuosen He
- Department of Emergency Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Junyan Hu
- Department of Emergency Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xi Liu
- Department of Emergency Medicine, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
26
|
Gad AI, Ibrahim NF, Almadani N, Mahfouz R, Nofal HA, El-Rafey DS, Ali HT, El-Hawary AT, Sadek AMEM. Therapeutic Effects of Semaglutide on Nonalcoholic Fatty Liver Disease with Type 2 Diabetes Mellitus and Obesity: An Open-Label Controlled Trial. Diseases 2024; 12:186. [PMID: 39195185 DOI: 10.3390/diseases12080186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/11/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND GLP-1 receptor agonists (GLP-1 RAs) have been shown to improve glycemic control and insulin sensitivity and reduce body weight in obese patients with type 2 diabetes mellitus (T2D). This trial sought to evaluate the therapeutic effect of oral and subcutaneous semaglutide in NAFLD and its sequelae in obesity and/or T2D. METHODS In an open-labelled intervention study, the sample was 180 patients classified into three parallel groups (1:1:1): group I received oral semaglutide, group II patients received injectable semaglutide, and group III received pioglitazone and/or vitamin E. Patients were evaluated at 6 and 12 months. RESULTS There was a substantial improvement in lipid profile, liver enzymes, and body mass index, especially in group II. As for HDL, only group II showed a consistent increase at both 6 months (51 ± 4.62 mg/dL) and 12 months (50.08 ± 2.45 mg/dL) compared with baseline (45.6 ± 6.37 mg/dL) (p-value < 0.001). Despite the non-significant difference in NAFLD fibrosis score (NFS) (p-value = 0.45 and 0.63), group II had significantly lower scores of the fibrosis-4 score (FIB-4), liver stiffness measurement (LSM), and controlled attenuation parameter (CAP) at 6 and 12 months (p-value < 0.001). Conclusions: Semaglutide improves lipid profile, liver steatosis, and fibrosis parameters and reduces the BMI in T2D and obese patients with NAFLD.
Collapse
Affiliation(s)
- Ahmed I Gad
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Nevin F Ibrahim
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Noura Almadani
- Community and Psychiatric Mental Health Nursing Department, College of Nursing, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Rasha Mahfouz
- Community and Psychiatric Mental Health Nursing Department, College of Nursing, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Hanaa A Nofal
- Community, Environmental Occupational Medicine Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Dina S El-Rafey
- Community, Environmental Occupational Medicine Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | | | - Amr T El-Hawary
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Ayman M E M Sadek
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
27
|
Xiong Y, Shi X, Xiong X, Li S, Zhao H, Song H, Wang J, Zhang L, You S, Ji G, Liu B, Wu N. A systematic review and meta-analysis of randomized controlled trials: effects of mediterranean diet and low-fat diet on liver enzymes and liver fat content of NAFLD. Food Funct 2024; 15:8248-8257. [PMID: 39076035 DOI: 10.1039/d4fo01461h] [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: 07/31/2024]
Abstract
Background: Non-alcoholic fatty liver disease (NAFLD) has emerged as a leading cause of several chronic diseases, imposing a significant global economic burden. The Mediterranean diet (MD) and low-fat diet (LFD) are the two primary recommended dietary patterns that exhibit distinct positive effects on treating NAFLD. Objective: To investigate which of the two diets, MD and LFD, is more effective in the treatment of NAFLD. Methods: Randomized controlled trials (RCTs) up to April 2024 were searched for in PubMed, Web of Science, Medline, Scopus and Embase. Interventions included MD or LFD, with primary outcome measures being intrahepatic lipid, liver stiffness, triglycerides, total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, alanine aminotransferase, gamma-glutamyl transferase, and homeostasis model assessment of insulin resistance. Secondary outcomes included weight, waist circumference, and body mass index. Use of random effects meta-analysis to assess outcomes of interest. Results: meta-analysis revealed no significant differences between MD and LFD in improving liver enzymes, liver fat, and related indices in NAFLD patients. Our findings provide compelling evidence for patients and healthcare professionals, allowing patients to choose a dietary pattern that aligns with their preferences and disease conditions. In summary, both MD and LFD can equivalently ameliorate NAFLD in the short term. Conclusions: Our results show that MD and LFD have similar therapeutic effects on liver enzymes and liver fat content in patients with NAFLD in the short term. Furthermore, our meta-analysis results have also opened up a new avenue of thought as to whether similar effects are achieved by alternating MD and LFD on alternate days.
Collapse
Affiliation(s)
- Yalan Xiong
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xinyu Shi
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xinying Xiong
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Shenyu Li
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hanhua Zhao
- Department of Sport Science, College of Education, Zhejiang University, Hangzhou, China
| | - Hualing Song
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jianying Wang
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Lei Zhang
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Shengfu You
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Guang Ji
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Baocheng Liu
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Na Wu
- School of Public Health, Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
28
|
Cho SH, Kim S, Oh R, Kim JY, Lee YB, Jin SM, Hur KY, Kim G, Kim JH. Metabolic dysfunction-associated fatty liver disease and heavy alcohol consumption increase mortality:A nationwide study. Hepatol Int 2024; 18:1168-1177. [PMID: 38806774 DOI: 10.1007/s12072-024-10671-9] [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: 11/28/2023] [Accepted: 03/17/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND The effects of excessive alcohol consumption on the prognosis of metabolic dysfunction-associated fatty liver disease (MAFLD) remain unclear. We investigated all-cause and cause-specific mortality according to the amount of alcohol consumed by Asian individuals with MAFLD. METHODS This nationwide retrospective study included 996,508 adults aged 40-79 years who underwent health check-ups between 2009 and 2012. Participants were categorized by the alcohol consumption-non-alcohol, moderate alcohol, and heavy alcohol group (≥ 30 g/day for men, ≥ 20 g/day for women) and by the combination of the presence or absence of MAFLD. Hepatic steatosis was defined as the fatty liver index ≥ 30. Cox analyses were used to analyze the association between alcohol consumption and MAFLD and all-cause and cause-specific mortality. RESULTS MAFLD significantly increased all-cause, liver-, and cancer-related mortality. Individuals with both MAFLD and heavy alcohol consumption expressed the highest mortality risk in liver-related mortality compared to non-MAFLD and non-alcohol group (adjusted hazard ratio (HR), 9.8; 95% confidence interval (CI), 8.20-12.29). Regardless of MAFLD, heavy alcohol consumption increased the risk of liver- and cancer-related mortality. CONCLUSIONS MAFLD and heavy alcohol consumption increased all-cause, liver-, and cancer-related mortality. Heavy alcohol consumption and MAFLD synergistically increase liver-related mortality.
Collapse
Affiliation(s)
- So Hyun Cho
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Seohyun Kim
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06355, Republic of Korea
| | - Rosa Oh
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Ji Yoon Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - You-Bin Lee
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Sang-Man Jin
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Kyu Yeon Hur
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea
| | - Gyuri Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea.
| | - Jae Hyeon Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-Ro, Gangnam-Gu, Seoul, 06351, Republic of Korea.
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, 06355, Republic of Korea.
| |
Collapse
|
29
|
Seko Y, Yamaguchi K, Shima T, Iwaki M, Takahashi H, Kawanaka M, Tanaka S, Mitsumoto Y, Yoneda M, Nakajima A, Okanoue T, Itoh Y. Differential Effects of Genetic Polymorphism on Comorbid Disease in Metabolic Dysfunction-Associated Steatotic Liver Disease. Clin Gastroenterol Hepatol 2024; 22:1436-1443.e4. [PMID: 38604296 DOI: 10.1016/j.cgh.2024.02.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/31/2024] [Accepted: 02/29/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND & AIMS PNPLA3 rs738409, TM6SF2 rs58542926, and HSD17B13 rs72613567 have been associated with an increased risk of liver-related events (LREs) in patients with metabolic dysfunction-associated steatotic liver disease (MASLD). In this study, we investigated the combined effects of these variants on LREs. METHODS The longitudinal multicenter cohort study enrolled 1178 patients with biopsy-proven MASLD. We calculated the genetic risk of hepatic fibrosis and LRE according to the impact of these variants. RESULTS Patients with genetic fibrosis scores of 2, 3, and 4 or 5 were at greater risk than patients with scores of 0 or 1, with odds ratios of 2.45 (95% CI, 1.27-4.74), 2.14 (95% CI, 1.17-3.94), and 2.54 (95% CI, 1.35-4.77), respectively. Multivariate analysis revealed that PNPLA3 and TM6SF2, but not HSD17B13, were associated significantly with LRE development. The hazard ratio of the genetic high-risk group for LRE was 1.91 (95% CI, 1.20-3.04). The higher risk of LRE development in the genetic high-risk group also was seen in patients with F ≥ 3 or Fibrosis-4 index > 2.67. The hazard ratios of the genetic high-risk group for LRE were greater in patients without obesity, without diabetes, and of younger age compared with patients with obesity, with diabetes, or of older age, respectively. CONCLUSIONS This combination of MASLD-related genetic variants is useful for predicting LREs in Japanese patients with MASLD. The genetic risk according to these variants is useful for LRE risk assessment, especially in patients without metabolic risk factors or in younger patients in Japan.
Collapse
Affiliation(s)
- Yuya Seko
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyou-ku, Kyoto, Japan
| | - Kanji Yamaguchi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyou-ku, Kyoto, Japan
| | - Toshihide Shima
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama, Japan
| | | | - Miwa Kawanaka
- General Internal Medicine 2, General Medical Center, Kawasaki Medical School, Kita-ku, Okayama, Japan
| | - Saiyu Tanaka
- Center for Digestive and Liver Diseases, Nara City Hospital, Nara, Japan
| | - Yasuhide Mitsumoto
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama, Japan
| | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Kanazawa-ku, Yokohama, Japan
| | - Takeshi Okanoue
- Department of Gastroenterology and Hepatology, Saiseikai Suita Hospital, Suita, Japan.
| | - Yoshito Itoh
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyou-ku, Kyoto, Japan
| |
Collapse
|
30
|
Kumada T, Toyoda H, Ogawa S, Gotoh T, Suzuki Y, Sugimoto K, Yoshida Y, Kuroda H, Kamada Y, Sumida Y, Ito T, Akita T, Tanaka J. Severe hepatic steatosis promotes increased liver stiffness in the early stages of metabolic dysfunction-associated steatotic liver disease. Liver Int 2024; 44:1700-1714. [PMID: 38558221 DOI: 10.1111/liv.15920] [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: 10/31/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND & AIMS The predictors of progression from steatosis to more advanced stages of metabolic dysfunction-associated steatotic liver disease (MASLD) remain unclear. We evaluated the association between the quantity of hepatic steatosis and longitudinal changes in liver stiffness measurements (LSMs) using magnetic resonance elastography (MRE) in patients with MASLD. METHODS We retrospectively analysed patients with MASLD who underwent at least two serial MRE and magnetic resonance imaging-based proton density fat fraction (MRI-PDFF) examinations at least 1 year apart. Fine-Gray competitive proportional hazard regression was used to identify LSM progression and regression factors. RESULTS A total of 471 patients were enrolled. Factors linked to LSM progression were steatosis grade 3 (MRI-PDFF ≥17.1%, adjusted hazard ratio [aHR] 2.597; 95% confidence interval [CI] 1.483-4.547) and albumin-bilirubin grade 2 or 3 (aHR 2.790; 95% CI 1.284-6.091), while the only factor linked to LSM regression was % decrease rate of MRI-PDFF ≥5% (aHR 2.781; 95% CI 1.584-4.883). Steatosis grade 3 correlated with a higher incidence rate of LSM progression than steatosis grade 1 (MRI-PDFF <11.3%) in patients with LSM stage 0 (<2.5 kilopascal [kPa]), and a % annual decrease rate of MRI-PDFF ≥5% correlated with a higher incidence rate of LSM regression than that of MRI-PDFF >-5% and <5% in patients with LSM stage 1 or 2-4 (≥2.5 kPa). CONCLUSIONS Severe hepatic steatosis was linked to significant LSM progression in patients with MASLD and low LSM (<2.5 kPa).
Collapse
Affiliation(s)
- Takashi Kumada
- Department of Nursing, Faculty of Nursing, Gifu Kyoritsu University, Gifu, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Ogaki, Japan
| | - Sadanobu Ogawa
- Department of Imaging Diagnosis, Ogaki Municipal Hospital, Ogaki, Japan
| | - Tatsuya Gotoh
- Department of Imaging Diagnosis, Ogaki Municipal Hospital, Ogaki, Japan
| | - Yasuaki Suzuki
- Department of Gastroenterology, Nayoro City General Hospital, Nayoro, Japan
| | - Katsutoshi Sugimoto
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Yuichi Yoshida
- Department of Gastroenterology and Hepatology, Suita Municipal Hospital, Osaka, Japan
| | - Hidekatsu Kuroda
- Division of Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
| | - Yoshihiro Kamada
- Department of Advanced Metabolic Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshio Sumida
- Department of Healthcare Management, International University of Health and Welfare, Tokyo, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoyuki Akita
- Department of Epidemiology, Infectious Disease Control, and Prevention, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Junko Tanaka
- Department of Epidemiology, Infectious Disease Control, and Prevention, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
31
|
Petta S, Targher G, Romeo S, Pajvani UB, Zheng MH, Aghemo A, Valenti LVC. The first MASH drug therapy on the horizon: Current perspectives of resmetirom. Liver Int 2024; 44:1526-1536. [PMID: 38578141 DOI: 10.1111/liv.15930] [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: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/06/2024]
Abstract
The rising prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) poses a significant global health challenge, affecting over 30% of adults worldwide. MASLD is linked to increased mortality rates and substantial healthcare costs, primarily driven by its progression to metabolic dysfunction-associated steatohepatitis (MASH), which can lead to severe liver complications including cirrhosis and hepatocellular carcinoma. Despite its growing burden, effective pharmacotherapy for MASLD/MASH has been lacking until the recent conditional approval of resmetirom by the FDA. Resmetirom, a liver-targeted thyroid hormone receptor-β selective drug, has shown promise in clinical trials for treating non-cirrhotic MASH with moderate to advanced fibrosis. It has demonstrated efficacy in reducing hepatic fat content, improving liver histology (both MASH resolution and fibrosis improvement), and ameliorating biomarkers of liver damage without significant effects on body weight or glucose metabolism. Notably, resmetirom also exhibits favourable effects on circulating lipids, potentially reducing cardiovascular risk in MASLD/MASH patients. The safety profile of resmetirom appears acceptable, with gastrointestinal adverse events being the most common, though generally mild or moderate. However, long-term surveillance is warranted to monitor for potential risks related to thyroid, gonadal, or bone diseases. Clinical implementation of resmetirom faces challenges in patient selection and monitoring treatment response, and will heavily rely on non-invasive tests for liver fibrosis assessment. Nonetheless, resmetirom represents a landmark breakthrough in MASLD/MASH treatment, paving the way for future therapeutic strategies aiming to mitigate the multifaceted risks associated with this complex metabolic liver disease.
Collapse
Affiliation(s)
- Salvatore Petta
- Gastroenterology and Hepatology, PROMISE, Università di Palermo, Palermo, Italy
| | - Giovanni Targher
- Department of Medicine, University of Verona, Verona, Italy
- Metabolic Diseases Research Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Negrar di Valpolicella, Italy
| | - Stefano Romeo
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Nutrition Unit, Department of Medical and Surgical Science, University Magna Graecia, Catanzaro, Italy
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Utpal B Pajvani
- Department of Medicine and Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Ming-Hua Zheng
- Department of Hepatology, MAFLD Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for The Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Alessio Aghemo
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Luca V C Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Precision Medicine, Biological Resource Center Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
32
|
Bril F, Kalavalapalli S, Lomonaco R, Frye R, Godinez Leiva E, Cusi K. Insulin resistance is an integral feature of MASLD even in the presence of PNPLA3 variants. JHEP Rep 2024; 6:101092. [PMID: 39022386 PMCID: PMC11252529 DOI: 10.1016/j.jhepr.2024.101092] [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: 03/04/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 07/20/2024] Open
Abstract
Background & Aims It has been postulated that carriers of PNPLA3 I148M (CG [Ile/Met] or GG [Met/Met]) develop metabolic dysfunction-associated steatotic liver disease (MASLD) in the absence of insulin resistance or metabolic syndrome. However, the relationship between insulin resistance and MASLD according to the PNPLA3 allele has not been carefully assessed. Methods A total of 204 participants were recruited and underwent PNPLA3 genotyping, an oral glucose tolerance test, liver proton magnetic resonance spectroscopy and percutaneous liver biopsy if diagnosed with MASLD. A subgroup of patients (n = 55) had an euglycemic hyperinsulinemic clamp with glucose tracer infusion. Results As expected, patients with the CG/GG genotype had worse intrahepatic triglyceride content and worse liver histology. However, regardless of PNPLA3 genotype, patients with a diagnosis of MASLD had severe whole-body insulin resistance (Matsuda index, an estimation of insulin resistance in glucose metabolic pathways) and fasting and postprandial adipose tissue insulin resistance (Adipo-IR index and free fatty acid suppression during the oral glucose tolerance test, respectively, as measures of insulin resistance in lipolytic metabolic pathways) compared to patients without MASLD. Moreover, for the same amount of liver fat accumulation, insulin resistance was similar in patients with genotypes CC vs. CG/GG. In multiple regression analyses, A1c and Adipo-IR were associated with the presence of MASLD and advanced liver fibrosis, independently of PNPLA3 genotype. Conclusions PNPLA3 variant carriers with MASLD are equally insulin resistant as non-carriers with MASLD at the level of the liver, muscle, and adipose tissue. This calls for reframing "PNPLA3 MASLD" as an insulin-resistant condition associated with increased hepatic susceptibility to metabolic insults, such as obesity or diabetes, wherein early identification and aggressive intervention are warranted to reverse metabolic dysfunction and prevent disease progression. Impact and implications It has been proposed that the PNPLA3 G allele is associated with the presence of metabolic dysfunction-associated steatotic liver disease (MASLD) in the absence of insulin resistance. However, our results suggest that regardless of PNPLA3 alleles, the presence of insulin resistance is necessary for the development of MASLD. This calls for reframing patients with "PNPLA3 MASLD" not as insulin sensitive, but on the contrary, as an insulin-resistant population with increased hepatic susceptibility to metabolic insults, such as obesity or diabetes.
Collapse
Affiliation(s)
- Fernando Bril
- Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham. 510 20 Street South, FOT 825A, 35233, Birmingham, AL, USA
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA. 1600 Archer Road, Room H2, 32610, Gainesville, FL, USA
| | - Srilaxmi Kalavalapalli
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA. 1600 Archer Road, Room H2, 32610, Gainesville, FL, USA
| | - Romina Lomonaco
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA. 1600 Archer Road, Room H2, 32610, Gainesville, FL, USA
| | - Reginald Frye
- Center for Pharmacogenomics, University of Florida, Gainesville, FL, USA; Currently at College of Pharmacy, University of Tennessee Health Science Center, USA
| | - Eddison Godinez Leiva
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA. 1600 Archer Road, Room H2, 32610, Gainesville, FL, USA
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA. 1600 Archer Road, Room H2, 32610, Gainesville, FL, USA
| |
Collapse
|
33
|
Ferenc K, Jarmakiewicz-Czaja S, Sokal-Dembowska A, Stasik K, Filip R. Common Denominator of MASLD and Some Non-Communicable Diseases. Curr Issues Mol Biol 2024; 46:6690-6709. [PMID: 39057041 PMCID: PMC11275402 DOI: 10.3390/cimb46070399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Currently, steatohepatitis has been designated as metabolic dysfunction-associated steatohepatitis (MASLD). MASLD risk factors mainly include metabolic disorders but can also include genetic, epigenetic, and environmental factors. Disease entities such as obesity, diabetes, cardiovascular disease, and MASLD share similar pathomechanisms and risk factors. Moreover, a bidirectional relationship is observed between the occurrence of certain chronic diseases and MASLD. These conditions represent a global public health problem that is responsible for poor quality of life and high mortality. It seems that paying holistic attention to these problems will not only help increase the chances of reducing the incidence of these diseases but also assist in the prevention, treatment, and support of patients.
Collapse
Affiliation(s)
- Katarzyna Ferenc
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (K.F.)
| | - Sara Jarmakiewicz-Czaja
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (S.J.-C.); (A.S.-D.)
| | - Katarzyna Stasik
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (K.F.)
- IBD Unit, Department of Gastroenterology, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| | - Rafał Filip
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland; (K.F.)
- IBD Unit, Department of Gastroenterology, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
34
|
Yan L, Hu X, Wu S, Cui C, Zhao S. Association between the cardiometabolic index and NAFLD and fibrosis. Sci Rep 2024; 14:13194. [PMID: 38851771 PMCID: PMC11162484 DOI: 10.1038/s41598-024-64034-3] [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: 09/20/2023] [Accepted: 06/04/2024] [Indexed: 06/10/2024] Open
Abstract
Composed of obesity and lipid parameters, the cardiometabolic index (CMI) has emerged as a novel diagnostic tool. Originally developed for diabetes diagnosis, its application has expanded to identifying patients with cardiovascular diseases, such as atherosclerosis and hypertension. However, the relationship between CMI and non-alcoholic fatty liver disease (NAFLD) and liver fibrosis in the US population remains unclear. This cross-sectional study analyzed data from the National Health and Nutrition Examination Survey (NHANES) spanning 2017-2020, involving 2996 participants aged 20 years or older. Vibration controlled transient elastography using a FibroScan® system (model 502, V2 Touch) with controlled attenuation parameter measurements identified NAFLD at a threshold of ≥ 274 dB/m, while liver stiffness measurement (LSM) results (median, ≥ 8.2 kPa) indicated fibrosis. A multifactorial logistic regression model explored the relationship between CMI and NAFLD and fibrosis. The effectiveness of CMI in detecting NAFLD and liver fibrosis was assessed through receiver operating characteristic curve analysis. Controlling for potential confounders, CMI showed a significant positive association with NAFLD (adjusted OR = 1.44, 95% CI 1.44-1.45) and liver fibrosis (adjusted OR = 1.84, 95% CI 1.84-1.85). The Areas Under the Curve for predicting NAFLD and fibrosis were 0.762 (95% CI 0.745 ~ 0.779) and 0.664(95% CI 0.633 ~ 0.696), respectively, with optimal cut-off values of 0.462 and 0.527. There is a positive correlation between CMI and NAFLD and fibrosis, which is a suitable and simple predictor of NAFLD and fibrosis.
Collapse
Affiliation(s)
- Laisha Yan
- Department of Cardio Surgery Intensive Care Unit, Ningbo Medical Centre Li Huili Hospital, Ningbo, China
| | - Xiaoyan Hu
- Department of Cardio Surgery Intensive Care Unit, Ningbo Medical Centre Li Huili Hospital, Ningbo, China
| | - Shanshan Wu
- Department of Cardio Surgery Intensive Care Unit, Ningbo Medical Centre Li Huili Hospital, Ningbo, China
| | - Can Cui
- Department of Cardio Surgery Intensive Care Unit, Ningbo Medical Centre Li Huili Hospital, Ningbo, China
| | - Shunying Zhao
- Department of Cardio Surgery Intensive Care Unit, Ningbo Medical Centre Li Huili Hospital, Ningbo, China.
| |
Collapse
|
35
|
EASL-EASD-EASO Clinical Practice Guidelines on the Management of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Obes Facts 2024; 17:374-444. [PMID: 38852583 PMCID: PMC11299976 DOI: 10.1159/000539371] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously termed non-alcoholic fatty liver disease (NAFLD), is defined as steatotic liver disease (SLD) in the presence of one or more cardiometabolic risk factor(s) and the absence of harmful alcohol intake. The spectrum of MASLD includes steatosis, metabolic dysfunction-associated steatohepatitis (MASH, previously NASH), fibrosis, cirrhosis and MASH-related hepatocellular carcinoma (HCC). This joint EASL-EASD-EASO guideline provides an update on definitions, prevention, screening, diagnosis and treatment for MASLD. Case-finding strategies for MASLD with liver fibrosis, using non-invasive tests, should be applied in individuals with cardiometabolic risk factors, abnormal liver enzymes, and/or radiological signs of hepatic steatosis, particularly in the presence of type 2 diabetes (T2D) or obesity with additional metabolic risk factor(s). A stepwise approach using blood-based scores (such as FIB-4) and, sequentially, imaging techniques (such as transient elastography) is suitable to rule-out/in advanced fibrosis, which is predictive of liver-related outcomes. In adults with MASLD, lifestyle modification - including weight loss, dietary changes, physical exercise and discouraging alcohol consumption - as well as optimal management of comorbidities - including use of incretin-based therapies (e.g. semaglutide, tirzepatide) for T2D or obesity, if indicated - is advised. Bariatric surgery is also an option in individuals with MASLD and obesity. If locally approved and dependent on the label, adults with non-cirrhotic MASH and significant liver fibrosis (stage ≥2) should be considered for a MASH-targeted treatment with resmetirom, which demonstrated histological effectiveness on steatohepatitis and fibrosis with an acceptable safety and tolerability profile. No MASH-targeted pharmacotherapy can currently be recommended for the cirrhotic stage. Management of MASH-related cirrhosis includes adaptations of metabolic drugs, nutritional counselling, surveillance for portal hypertension and HCC, as well as liver transplantation in decompensated cirrhosis.
Collapse
|
36
|
Caddeo A, Maurotti S, Kovooru L, Romeo S. 3D culture models to study pathophysiology of steatotic liver disease. Atherosclerosis 2024; 393:117544. [PMID: 38677899 DOI: 10.1016/j.atherosclerosis.2024.117544] [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: 11/21/2023] [Revised: 03/19/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024]
Abstract
Steatotic liver disease (SLD) refers to a spectrum of diseases caused by hepatic lipid accumulation. SLD has emerged as the leading cause of chronic liver disease worldwide. Despite this burden and many years, understanding the pathophysiology of this disease is challenging due to the inaccessibility to human liver specimens. Therefore, cell-based in vitro systems are widely used as models to investigate the pathophysiology of SLD. Culturing hepatic cells in monolayers causes the loss of their hepatocyte-specific phenotype and, consequently, tissue-specific function and architecture. Hence, three-dimensional (3D) culture models allow cells to mimic the in vivo microenvironment and spatial organization of the liver unit. The utilization of 3D in vitro models minimizes the drawbacks of two-dimensional (2D) cultures and aligns with the 3Rs principles to alleviate the number of in vivo experiments. This article provides an overview of liver 3D models highlighting advantages and limitations, and culminates by discussing their applications in pharmaceutical and biomedical research.
Collapse
Affiliation(s)
- Andrea Caddeo
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy.
| | - Samantha Maurotti
- Department of Clinical and Experimental Medicine, University Magna Graecia, Catanzaro, Italy
| | - Lohitesh Kovooru
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.
| |
Collapse
|
37
|
Tas E, Sundararajan D, Lo JS, Morelli N, Garcia-Reyes Y, Ware MA, Rahat H, Ou X, Na X, Sundaram S, Severn C, Pyle LL, Børsheim E, Vajravelu ME, Muzumdar R, Dranoff JA, Cree MG. Diagnostic Accuracy of Transient Elastography in Hepatosteatosis in Youth With Obesity. J Endocr Soc 2024; 8:bvae110. [PMID: 38895640 PMCID: PMC11185182 DOI: 10.1210/jendso/bvae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Indexed: 06/21/2024] Open
Abstract
Context Steatotic liver disease is common but overlooked in childhood obesity; diagnostic methods are invasive or expensive. Objective We sought to determine the diagnostic accuracy of vibration-controlled transient elastography (VCTE) compared with magnetic resonance imaging (MRI) in adolescents with obesity and high risk for hepatosteatosis. Methods Baseline data in 3 clinical trials enrolling adolescents with obesity were included (NCT03919929, NCT03717935, NCT04342390). Liver fat was assessed using MRI fat fraction and VCTE-based controlled attenuation parameter (CAP). Hepatosteatosis was defined as MRI fat fraction ≥5.0%. The area under the receiver-operating characteristic curves (AUROCs) for CAP against MRI was calculated, and optimal CAP using the Youden index for hepatosteatosis diagnosis was determined. Results Data from 82 adolescents (age 15.6 ± 1.4 years, body mass index 36.5 ± 5.9 kg/m2, 81% female) were included. Fifty youth had hepatosteatosis by MRI (fat fraction 9.3% ; 95% CI 6.7, 14.0), and 32 participants did not have hepatosteatosis (fat fraction 3.1%; 95% CI 2.2, 3.9; P < .001). The hepatosteatosis group had higher mean CAP compared with no hepatosteatosis (293 dB/m; 95% CI 267, 325 vs 267 dB/m; 95% CI 248, 282; P = .0120). A CAP of 281 dB/m had the highest sensitivity (60%) and specificity (74%) with AUROC of 0.649 (95% CI 0.51-0.79; P = .04) in the entire cohort. In a subset of participants with polycystic ovary syndrome (PCOS), a CAP of 306 dB/m had the highest sensitivity (78%) and specificity (52%) and AUROC of 0.678 (95% CI 0.45-0.90; P = .108). Conclusion CAP of 281 dB/m has modest diagnostic performance for hepatosteatosis compared with MRI in youth with significant obesity. A higher CAP in youth with PCOS suggests that comorbidities might affect optimal CAP in hepatosteatosis diagnosis.
Collapse
Affiliation(s)
- Emir Tas
- Pediatric Endocrinology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
- Center for Childhood Obesity Prevention, Arkansas Children's Research Institute, Little Rock, AR 72202, USA
| | - Divya Sundararajan
- Pediatric Endocrinology, University of Colorado Anschutz, Aurora, CO 80045, USA
| | - Jaclyn S Lo
- Pediatric Endocrinology, University of Colorado Anschutz, Aurora, CO 80045, USA
| | - Nazeen Morelli
- Pediatric Endocrinology, University of Colorado Anschutz, Aurora, CO 80045, USA
| | | | - Meredith A Ware
- Pediatric Endocrinology, University of Colorado Anschutz, Aurora, CO 80045, USA
| | - Haseeb Rahat
- Pediatric Endocrinology, University of Colorado Anschutz, Aurora, CO 80045, USA
| | - Xiawei Ou
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Xiaoxu Na
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Shikha Sundaram
- Pediatric Gastroenterology, University of Colorado Anschutz, Aurora, CO 80045, USA
| | - Cameron Severn
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Laura L Pyle
- Pediatric Endocrinology, University of Colorado Anschutz, Aurora, CO 80045, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Elisabet Børsheim
- Center for Childhood Obesity Prevention, Arkansas Children's Research Institute, Little Rock, AR 72202, USA
| | - Mary Ellen Vajravelu
- Pediatric Endocrinology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Radhika Muzumdar
- Pediatric Endocrinology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Jonathan A Dranoff
- Section of Digestive Diseases, Yale School of Medicine, New Haven, CT 06520, USA
| | - Melanie G Cree
- Pediatric Endocrinology, University of Colorado Anschutz, Aurora, CO 80045, USA
- Ludeman Center for Women's Health, Aurora, CO 80045, USA
| |
Collapse
|
38
|
Meroni M, Longo M, Dongiovanni P. Cardiometabolic risk factors in MASLD patients with HCC: the other side of the coin. Front Endocrinol (Lausanne) 2024; 15:1411706. [PMID: 38846491 PMCID: PMC11153718 DOI: 10.3389/fendo.2024.1411706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/10/2024] [Indexed: 06/09/2024] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) constitutes the commonest cause of chronic liver disorder worldwide, whereby affecting around one third of the global population. This clinical condition may evolve into Metabolic Dysfunction-Associated Steatohepatitis (MASH), fibrosis, cirrhosis and hepatocellular carcinoma (HCC), in a predisposed subgroup of patients. The complex pathogenesis of MASLD is severely entangled with obesity, dyslipidemia and type 2 diabetes (T2D), so far so nutritional and lifestyle recommendations may be crucial in influencing the risk of HCC and modifying its prognosis. However, the causative association between HCC onset and the presence of metabolic comorbidities is not completely clarified. Therefore, the present review aimed to summarize the main literature findings that correlate the presence of inherited or acquired hyperlipidemia and metabolic risk factors with the increased predisposition towards liver cancer in MASLD patients. Here, we gathered the evidence underlining the relationship between circulating/hepatic lipids, cardiovascular events, metabolic comorbidities and hepatocarcinogenesis. In addition, we reported previous studies supporting the impact of triglyceride and/or cholesterol accumulation in generating aberrancies in the intracellular membranes of organelles, oxidative stress, ATP depletion and hepatocyte degeneration, influencing the risk of HCC and its response to therapeutic approaches. Finally, our pursuit was to emphasize the link between HCC and the presence of cardiometabolic abnormalities in our large cohort of histologically-characterized patients affected by MASLD (n=1538), of whom 86 had MASLD-HCC by including unpublished data.
Collapse
|
39
|
Zhang X, Zhou C, Hu J, Hu J, Ding Y, Chen S, Wang X, Xu L, Gou Z, Zhang S, Shi W. Six-gene prognostic signature for non-alcoholic fatty liver disease susceptibility using machine learning. Medicine (Baltimore) 2024; 103:e38076. [PMID: 38728481 PMCID: PMC11081587 DOI: 10.1097/md.0000000000038076] [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: 12/28/2023] [Accepted: 04/10/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND nonalcoholic fatty liver disease (NAFLD) is a common liver disease affecting the global population and its impact on human health will continue to increase. Genetic susceptibility is an important factor influencing its onset and progression, and there is a lack of reliable methods to predict the susceptibility of normal populations to NAFLD using appropriate genes. METHODS RNA sequencing data relating to nonalcoholic fatty liver disease was analyzed using the "limma" package within the R software. Differentially expressed genes were obtained through preliminary intersection screening. Core genes were analyzed and obtained by establishing and comparing 4 machine learning models, then a prediction model for NAFLD was constructed. The effectiveness of the model was then evaluated, and its applicability and reliability verified. Finally, we conducted further gene correlation analysis, analysis of biological function and analysis of immune infiltration. RESULTS By comparing 4 machine learning algorithms, we identified SVM as the optimal model, with the first 6 genes (CD247, S100A9, CSF3R, DIP2C, OXCT 2 and PRAMEF16) as predictive genes. The nomogram was found to have good reliability and effectiveness. Six genes' receiver operating characteristic curves (ROC) suggest an essential role in NAFLD pathogenesis, and they exhibit a high predictive value. Further analysis of immunology demonstrated that these 6 genes were closely connected to various immune cells and pathways. CONCLUSION This study has successfully constructed an advanced and reliable prediction model based on 6 diagnostic gene markers to predict the susceptibility of normal populations to NAFLD, while also providing insights for potential targeted therapies.
Collapse
Affiliation(s)
- Xiang Zhang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Chunzi Zhou
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingwen Hu
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingwen Hu
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Yueping Ding
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shiqi Chen
- Lishui Hospital of Traditional Chinese Medicine, Lishui, China
| | - Xu Wang
- Shanghai Jinshan TCM-Integrated Hospital, Shanghai, China
| | - Lei Xu
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Gou
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuqiao Zhang
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiqun Shi
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
40
|
Dutta T, Sasidharan K, Ciociola E, Pennisi G, Noto FR, Kovooru L, Kroon T, Lindblom A, Du Y, Pirmoradian M, Wallin S, Mancina RM, Lindén D, Romeo S. Mitochondrial amidoxime-reducing component 1 p.Ala165Thr increases protein degradation mediated by the proteasome. Liver Int 2024; 44:1219-1232. [PMID: 38375985 DOI: 10.1111/liv.15857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 02/21/2024]
Abstract
OBJECTIVE Metabolic dysfunction-associated steatotic liver disease (MASLD) is a global health concern with no effective and specific drug treatment available. The rs2642438 minor allele in mitochondrial amidoxime-reducing component 1 (MARC1) results in an aminoacidic substitution (p.Ala165Thr) and associates with protection against MASLD. However, the mechanisms behind this protective effect are unknown. In this study, we examined the consequences of this aminoacidic substitution on protein stability and subcellular localization. METHODS We overexpressed the human MARC1 A165 (wild-type) or 165T (mutant) in vivo in mice and in vitro in human hepatoma cells (HepG2 and HuH-7), generated several mutants at position 165 by in situ mutagenesis and then examined protein levels. We also generated HepG2 cells stably overexpressing MARC1 A165 or 165T to test the effect of this substitution on MARC1 subcellular localization. RESULTS MARC1 165T overexpression resulted in lower protein levels than A165 both in vivo and in vitro. Similarly, any mutant at position 165 showed lower protein levels compared to the wild-type protein. We showed that the 165T mutant protein is polyubiquitinated and its degradation is accelerated through lysine-48 ubiquitin-mediated proteasomal degradation. We also showed that the 165T substitution does not affect the MARC1 subcellular localization. CONCLUSIONS This study shows that alanine at position 165 in MARC1 is crucial for protein stability, and the threonine substitution at this position leads to a hypomorphic protein variant due to lower protein levels. Our result supports the notion that lowering hepatic MARC1 protein level may be a successful therapeutic strategy for treating MASLD.
Collapse
Affiliation(s)
- Tanmoy Dutta
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Kavitha Sasidharan
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Ester Ciociola
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Grazia Pennisi
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
- Section of Gastroenterology and Hepatology, Dipartimento Di Promozione Della Salute, Materno Infantile, Medicina Interna e Specialistica Di Eccellenza (PROMISE), University of Palermo, Palermo, Italy
| | - Francesca R Noto
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
- Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Lohitesh Kovooru
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Tobias Kroon
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Lindblom
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Yue Du
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Mohammad Pirmoradian
- Translational Science and Experimental Medicine, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Simonetta Wallin
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rosellina M Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Daniel Lindén
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Division of Endocrinology, Department of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, The Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
- Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
41
|
Meroni M, De Caro E, Chiappori F, Longo M, Paolini E, Mosca E, Merelli I, Lombardi R, Badiali S, Maggioni M, Orro A, Mezzelani A, Valenti L, Fracanzani AL, Dongiovanni P. Hepatic and adipose tissue transcriptome analysis highlights a commonly deregulated autophagic pathway in severe MASLD. Obesity (Silver Spring) 2024; 32:923-937. [PMID: 38439203 DOI: 10.1002/oby.23996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 03/06/2024]
Abstract
OBJECTIVE The incidence of metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly ramping up due to the spread of obesity, which is characterized by expanded and dysfunctional visceral adipose tissue (VAT). Previous studies have investigated the hepatic transcriptome across MASLD, whereas few studies have focused on VAT. METHODS We performed RNA sequencing in 167 hepatic samples from patients with obesity and in a subset of 79 matched VAT samples. Circulating cathepsin D (CTSD), a lysosomal protease, was measured by ELISA, whereas the autophagy-lysosomal pathway was assessed by Western blot in hepatic and VAT samples (n = 20). RESULTS Inflammation, extracellular matrix remodeling, and mitochondrial dysfunction were upregulated in severe MASLD in both tissues, whereas autophagy and oxidative phosphorylation were reduced. Tissue comparative analysis revealed 13 deregulated genes, including CTSD, which showed the most robust diagnostic accuracy in discriminating mild and severe MASLD. CTSD expression correlated with circulating protein, whose increase was further validated in 432 histologically characterized MASLD patients, showing a high accuracy in foreseeing severe liver injury. In addition, the assessment of serum CTSD increased the performance of fibrosis 4 in diagnosing advanced disease. CONCLUSIONS By comparing the hepatic and VAT transcriptome during MASLD, we refined the concept by which CTSD may represent a potential biomarker of severe disease.
Collapse
Affiliation(s)
- Marica Meroni
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Emilia De Caro
- Life and Medical Sciences Institute (LIMES), University of Bonn, Germany/System Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Federica Chiappori
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Miriam Longo
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ettore Mosca
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Ivan Merelli
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Rosa Lombardi
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Sara Badiali
- Department of Surgery, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Maggioni
- Department of Pathology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessandro Orro
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Alessandra Mezzelani
- Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Precision Medicine Lab, Biological Resource Center, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Ludovica Fracanzani
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
42
|
Raverdy V, Tavaglione F, Chatelain E, Caiazzo R, Saponaro C, Lassailly G, Verkindt H, Baud G, Marciniak C, Chetboun M, Oukhouya-Daoud N, Gnemmi V, Leteurtre E, Duhamel A, Philippe M, Marot G, Romeo S, Pattou F. Performance of non-invasive tests for liver fibrosis resolution after bariatric surgery. Metabolism 2024; 153:155790. [PMID: 38219973 DOI: 10.1016/j.metabol.2024.155790] [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: 08/01/2023] [Revised: 12/05/2023] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND & AIMS The value of non-invasive tests for monitoring the resolution of significant liver fibrosis after treatment is poorly investigated. We compared the performances of six non-invasive tests to predict the resolution of significant fibrosis after bariatric surgery. METHODS Participants were individuals with obesity submitted to needle liver biopsy at the time of bariatric surgery, and 12 and/or 60 months after surgery. We calculated the fibrosis-4 index (FIB-4), NAFLD fibrosis score (NFS), AST to platelet ratio index (APRI), Hepatic fibrosis score (HFS), Fibrotic NASH index (FNI), and Liver risk score (LRS) at each time point, and compared their performances for predicting significant fibrosis (F ≥ 2) and its resolution following surgery. RESULTS At baseline, 2436 patients had liver biopsy, including 261 (10.7 %) with significant fibrosis. Overall, 672 patients had pre- and post-operative biopsies (564 at M12 and 328 at M60). The fibrosis stage decreased at M12 and M60 (p < 0.001 vs M0). Resolution of significant fibrosis occurred in 58/121 (47.9 %) at M12 and 32/50 (64 %) at M60. The mean value of all tests decreased after surgery, except for FIB-4. Performances for predicting fibrosis resolution was higher at M60 than at M12 for all tests, and maximal at M60 for FNI and LRS: area under the curve 0.843 (95%CI 0.71-0.95) and 0.92 (95%CI 0.84-1.00); positive likelihood ratio 3.75 (95 % CI 1.33-10.59) and 4.58 (95 % CI 1.65-12.70), respectively. CONCLUSIONS Results showed the value and limits of non-invasive tests for monitoring the evolution of liver fibrosis after an intervention. Following bariatric surgery, the best performances to predict the resolution of significant fibrosis were observed at M60 with tests combining liver and metabolic traits, namely FNI and LRS.
Collapse
Affiliation(s)
- Violeta Raverdy
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Federica Tavaglione
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy; Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Estelle Chatelain
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41-UAR 2014-PLBS, billille, F-59000 Lille, France
| | - Robert Caiazzo
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Chiara Saponaro
- European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Guillaume Lassailly
- CHU Lille, Univ. Lille, Inserm INFINITE-U1286, Department of Hepato-Gastroenterology, F-59000 Lille, France
| | - Helene Verkindt
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Gregory Baud
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Camille Marciniak
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Mikael Chetboun
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Naima Oukhouya-Daoud
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France
| | - Viviane Gnemmi
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France; Department of Pathology, CHU Lille, Univ. Lille, Lille, France
| | - Emmanuelle Leteurtre
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France; Department of Pathology, CHU Lille, Univ. Lille, Lille, France
| | - Alain Duhamel
- Univ. Lille, CHU Lille, ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales, F-59000 Lille, France
| | - Mathurin Philippe
- CHU Lille, Univ. Lille, Inserm INFINITE-U1286, Department of Hepato-Gastroenterology, F-59000 Lille, France
| | - Guillemette Marot
- Univ. Lille, CHU Lille, ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales, F-59000 Lille, France; Inria, MODAL: Models for Data Analysis and Learning, F-59000 Lille, France
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy; Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - François Pattou
- Service de chirurgie générale et endocrinienne, Centre Hospitalier Universitaire de Lille, Lille, France; European Genomic Institute for Diabetes, UMR 1190 Translational Research for Diabetes, Inserm, CHU Lille, University of Lille, Lille, France.
| |
Collapse
|
43
|
Gabriel-Medina P, Ferrer-Costa R, Ciudin A, Augustin S, Rivera-Esteban J, Pericàs JM, Selva DM, Rodriguez-Frias F. Accuracy of a sequential algorithm based on FIB-4 and ELF to identify high-risk advanced liver fibrosis at the primary care level. Intern Emerg Med 2024; 19:745-756. [PMID: 37952070 PMCID: PMC11039533 DOI: 10.1007/s11739-023-03441-2] [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: 12/02/2022] [Accepted: 06/21/2023] [Indexed: 11/14/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease, and liver fibrosis is the strongest predictor of morbimortality. We aimed to assess the performance of a sequential algorithm encompassing the Fibrosis 4 (FIB-4) and Enhanced Liver Fibrosis (ELF) scores for identifying patients at risk of advanced fibrosis. This cross-sectional study included one hospital-based cohort with biopsy-proven NAFLD (n = 140) and two primary care cohorts from different clinical settings: Type 2 Diabetes (T2D) follow-up (n = 141) and chronic liver disease (CLD) initial study (n = 138). Logistic regression analysis was performed to assess liver fibrosis diagnosis models based on FIB-4 and ELF biomarkers. The sequential algorithm retrieved the following accuracy parameters in predicting stages F3-4 in the biopsy-confirmed cohort: sensitivity (85%), specificity (73%), negative predictive value (79%) and positive predictive value (81%). In both T2D and CLD cohorts, a total of 28% of patients were classified as stages F3-4. Furthermore, of all F3-4 classified patients in the T2D cohort, 80% had a diagnosis of liver disease and 44% were referred to secondary care. Likewise, of all F3-4 classified patients in the CLD cohort, 71% had a diagnosis of liver disease and 44% were referred to secondary care. These results suggest the potential utility of this algorithm as a liver fibrosis stratifying tool in primary care, where updating referral protocols to detect high-risk F3-4 is needed. FIB-4 and ELF sequential measurement is an efficient strategy to prioritize patients with high risk of F3-4 in populations with metabolic risk factors.
Collapse
Affiliation(s)
- Pablo Gabriel-Medina
- Clinical Biochemistry Department, Vall d'Hebron University Hospital, 08035, Barcelona, Spain.
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), 08193, Barcelona, Spain.
- Clinical Biochemistry Research Team, Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain.
| | - Roser Ferrer-Costa
- Clinical Biochemistry Department, Vall d'Hebron University Hospital, 08035, Barcelona, Spain.
- Clinical Biochemistry Research Team, Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain.
| | - Andreea Ciudin
- Endocrinology and Nutrition Department, Vall d'Hebron University Hospital, 08035, Barcelona, Spain
- Diabetes and Metabolism Department, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029, Madrid, Spain
| | - Salvador Augustin
- Liver Unit, Internal Medicine Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron University Hospital, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029, Madrid, Spain
| | - Jesus Rivera-Esteban
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), 08193, Barcelona, Spain
- Liver Unit, Internal Medicine Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron University Hospital, 08035, Barcelona, Spain
| | - J M Pericàs
- Liver Unit, Internal Medicine Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron University Hospital, 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029, Madrid, Spain
| | - D M Selva
- Diabetes and Metabolism Department, Vall d'Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029, Madrid, Spain
| | - Francisco Rodriguez-Frias
- Clinical Biochemistry Department, Vall d'Hebron University Hospital, 08035, Barcelona, Spain
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), 08193, Barcelona, Spain
- Clinical Biochemistry Research Team, Vall d'Hebron Institut de Recerca (VHIR), 08035, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029, Madrid, Spain
| |
Collapse
|
44
|
Bianco C, Pelusi S, Margarita S, Tavaglione F, Jamialahmadi O, Malvestiti F, Periti G, Rondena J, Tomasi M, Carpani R, Ronzoni L, Vidali M, Ceriotti F, Fraquelli M, Vespasiani‐Gentilucci U, Romeo S, Prati D, Valenti L. Predictors of controlled attenuation parameter in metabolic dysfunction. United European Gastroenterol J 2024; 12:364-373. [PMID: 38141028 PMCID: PMC11017762 DOI: 10.1002/ueg2.12513] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/26/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND & AIMS Hepatic fat content can be non-invasively estimated by controlled attenuation parameter (CAP) during transient elastography. The aim of this study was to examine the determinants and predictors of CAP values in individuals with metabolic dysfunction. METHODS We enrolled 1230 consecutive apparently healthy individuals (Liver-Bible-2022 cohort) with ≥3 metabolic dysfunction features. CAP was measured by Fibroscan. CAP determinants and predictors were identified using backward stepwise analysis and introduced in generalized linear models. RESULTS Participants were predominantly males (82.9%), mean age was 53.8 ± 6.4 years, 600 (48.8%) had steatosis (CAP ≥ 275 dB/m), and 27 had liver stiffness measurement (LSM) ≥ 8 kPa. CAP values correlated with LSM (p < 10-22). In multivariable analysis, fasting insulin and abdominal circumference (AC) were the main determinants of CAP (p < 10-6), together with body mass index (BMI; p < 10-4), age, diabetes, triglycerides, ferritin, and lower HDL and thyroid stimulating hormone (TSH; p < 0.05 for all). In a subset of 592 participants with thyroid hormone measurement, we found an association between higher free triiodothyronine levels, correlating with lower TSH, and CAP values, independent of TSH and of levothyroxine treatment (p = 0.0025). A clinical CAP score based on age, BMI, AC, HbA1c, ALT, and HDL predicted CAP ≥ 275 dB/m with moderate accuracy (AUROC = 0.73), which was better than that of the Fatty Liver Index and of ALT (AUROC = 0.70/0.61, respectively) and validated it in multiple cohorts. CONCLUSION Abdominal adiposity and insulin resistance severity were the main determinants of CAP in individuals with metabolic dysfunction and may improve steatotic liver disease risk stratification. CAP values were modulated by the hypophysis-thyroid axis.
Collapse
Affiliation(s)
- Cristiana Bianco
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Serena Pelusi
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Sara Margarita
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Federica Tavaglione
- Clinical Medicine and Hepatology UnitDepartment of Internal Medicine and GeriatricsFondazione Policlinico Campus Bio‐MedicoRomeItaly
- Department of Medicine and SurgeryUniversità Campus Bio‐Medico di RomaRomeItaly
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyWallenberg LaboratoryUniversity of GothenburgGothenburgSweden
| | - Francesco Malvestiti
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
| | - Giulia Periti
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Jessica Rondena
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Melissa Tomasi
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Rossana Carpani
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Luisa Ronzoni
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Matteo Vidali
- Clinical Chemistry Unit and Laboratory MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Ferruccio Ceriotti
- Clinical Chemistry Unit and Laboratory MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Mirella Fraquelli
- Gastroenterology and Endoscopy UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Umberto Vespasiani‐Gentilucci
- Clinical Medicine and Hepatology UnitDepartment of Internal Medicine and GeriatricsFondazione Policlinico Campus Bio‐MedicoRomeItaly
- Department of Medicine and SurgeryUniversità Campus Bio‐Medico di RomaRomeItaly
| | - Stefano Romeo
- Department of Molecular and Clinical MedicineInstitute of MedicineSahlgrenska AcademyWallenberg LaboratoryUniversity of GothenburgGothenburgSweden
- Clinical Nutrition UnitDepartment of Medical and Surgical SciencesUniversity Magna GraeciaCatanzaroItaly
- Cardiology DepartmentSahlgrenska University HospitalGothenburgSweden
| | - Daniele Prati
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Luca Valenti
- Precision Medicine LabBiological Resource Center and Department of Transfusion MedicineFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanItaly
| |
Collapse
|
45
|
Rong S, Xia M, Vale G, Wang S, Kim CW, Li S, McDonald JG, Radhakrishnan A, Horton JD. DGAT2 inhibition blocks SREBP-1 cleavage and improves hepatic steatosis by increasing phosphatidylethanolamine in the ER. Cell Metab 2024; 36:617-629.e7. [PMID: 38340721 PMCID: PMC10939742 DOI: 10.1016/j.cmet.2024.01.011] [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: 08/10/2023] [Revised: 11/28/2023] [Accepted: 01/18/2024] [Indexed: 02/12/2024]
Abstract
Diacylglycerol acyltransferase 2 (DGAT2) catalyzes the final step of triglyceride (TG) synthesis. DGAT2 deletion in mice lowers liver TGs, and DGAT2 inhibitors are under investigation for the treatment of fatty liver disease. Here, we show that DGAT2 inhibition also suppressed SREBP-1 cleavage, reduced fatty acid synthesis, and lowered TG accumulation and secretion from liver. DGAT2 inhibition increased phosphatidylethanolamine (PE) levels in the endoplasmic reticulum (ER) and inhibited SREBP-1 cleavage, while DGAT2 overexpression lowered ER PE concentrations and increased SREBP-1 cleavage in vivo. ER enrichment with PE blocked SREBP-1 cleavage independent of Insigs, which are ER proteins that normally retain SREBPs in the ER. Thus, inhibition of DGAT2 shunted diacylglycerol into phospholipid synthesis, increasing the PE content of the ER, resulting in reduced SREBP-1 cleavage and less hepatic steatosis. This study reveals a new mechanism that regulates SREBP-1 activation and lipogenesis that is independent of sterols and SREBP-2 in liver.
Collapse
Affiliation(s)
- Shunxing Rong
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Mingfeng Xia
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Goncalo Vale
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Simeng Wang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Chai-Wan Kim
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Shili Li
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Jeffrey G McDonald
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Arun Radhakrishnan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA
| | - Jay D Horton
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9046, USA.
| |
Collapse
|
46
|
Tavaglione F, Flagiello V, Terracciani F, Gallo P, Capparelli E, Spiezia C, De Vincentis A, Palermo A, Scriccia S, Galati G, Napoli N, Daniels SJ, Blau JE, Carlsson B, Khazrai YM, Incalzi RA, Picardi A, Vespasiani-Gentilucci U. Non-invasive assessment of hepatic steatosis by ultrasound-derived fat fraction in individuals at high-risk for metabolic dysfunction-associated steatotic liver disease. Diabetes Metab Res Rev 2024; 40:e3787. [PMID: 38461408 DOI: 10.1002/dmrr.3787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/05/2024] [Accepted: 02/15/2024] [Indexed: 03/11/2024]
Abstract
AIMS Given the increasing number of individuals developing metabolic dysfunction-associated steatotic liver disease (MASLD) and the low rate of those with progressive liver disease, there is a pressing need to conceive affordable biomarkers to assess MASLD in general population settings. Herein, we aimed to investigate the performance of the ultrasound-derived fat fraction (UDFF) for hepatic steatosis in high-risk individuals. METHODS A total of 302 Europeans with obesity, type 2 diabetes, or a clinical history of hepatic steatosis were included in the analyses. Clinical, laboratory, and imaging data were collected using standardized procedures during a single screening visit in Rome, Italy. Hepatic steatosis was defined by controlled attenuation parameter (CAP) or ultrasound-based Hamaguchi's score. UDFF performance for hepatic steatosis was estimated by the area under the receiver operating characteristic curve (AUC). RESULTS Overall, median (IQR) UDFF was 12% (7-20). UDFF was positively correlated with CAP (ρ = 0.73, p < 0.0001) and Hamaguchi's score (ρ = 0.79, p < 0.0001). Independent predictors of UDFF were circulating triglycerides, alanine aminotransferase (ALT), and ultrasound-measured visceral adipose tissue (VAT). UDFF AUC was 0.89 (0.85-0.93) and 0.92 (0.88-0.95) for CAP- and ultrasound-diagnosed hepatic steatosis, respectively. UDFF AUC for hepatic steatosis was higher than those of fatty liver index (FLI), hepatic steatosis index (HSI), CAP-score (CAPS), and ALT (p < 0.0001). Lower age, ALT, and VAT were associated with discordance between UDFF and ultrasound. CONCLUSIONS UDFF may be a simple and accurate imaging biomarker to assess hepatic steatosis and monitor changes in hepatic fat content over time or in response to therapeutic interventions beyond clinical trials.
Collapse
Affiliation(s)
- Federica Tavaglione
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Valentina Flagiello
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Francesca Terracciani
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Paolo Gallo
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Emma Capparelli
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Spiezia
- Research Unit of Food Science and Human Nutrition, Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Antonio De Vincentis
- Research Unit of Internal Medicine, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Andrea Palermo
- Research Unit of Metabolic Bone and Thyroid Disorders, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Metabolic Bone and Thyroid Disorders, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Sara Scriccia
- Check-up Center, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Giovanni Galati
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Nicola Napoli
- Research Unit of Metabolic Bone and Thyroid Disorders, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Metabolic Bone and Thyroid Disorders, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Samuel J Daniels
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jenny E Blau
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Björn Carlsson
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Yeganeh M Khazrai
- Research Unit of Food Science and Human Nutrition, Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Research Unit of Nutrition and Prevention, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Raffaele Antonelli Incalzi
- Research Unit of Internal Medicine, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Antonio Picardi
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Umberto Vespasiani-Gentilucci
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| |
Collapse
|
47
|
Nogueira JP, Cusi K. Role of Insulin Resistance in the Development of Nonalcoholic Fatty Liver Disease in People With Type 2 Diabetes: From Bench to Patient Care. Diabetes Spectr 2024; 37:20-28. [PMID: 38385099 PMCID: PMC10877218 DOI: 10.2337/dsi23-0013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Insulin resistance is implicated in both the pathogenesis of nonalcoholic fatty liver disease (NAFLD) and its progression from steatosis to steatohepatitis, cirrhosis, and even hepatocellular carcinoma, which is known to be more common in people with type 2 diabetes. This article reviews the role of insulin resistance in the metabolic dysfunction observed in obesity, type 2 diabetes, atherogenic dyslipidemia, and hypertension and how it is a driver of the natural history of NAFLD by promoting glucotoxicity and lipotoxicity. The authors also review the genetic and environmental factors that stimulate steatohepatitis and fibrosis progression and their relationship with cardiovascular disease and summarize guidelines supporting the treatment of NAFLD with diabetes medications that reduce insulin resistance, such as pioglitazone or glucagon-like peptide 1 receptor agonists.
Collapse
Affiliation(s)
- Juan Patricio Nogueira
- Universidad del Pacifico, Asunción, Paraguay
- Centro de Investigación en Endocrinología, Nutrición y Metabolismo, Facultad de Ciencias de la Salud, Universidad Nacional de Formosa, Formosa, Argentina
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL
| |
Collapse
|
48
|
Romeo S, Jamialahmadi O, De Vincentis A, Tavaglione F, Malvestiti F, Li-Gao R, Mancina R, Alvarez M, Gelev K, Maurotti S, Vespasiani-Gentilucci U, Rosendaal F, Kozlitina J, Pajukanta P, Pattou F, Valenti L. Partitioned polygenic risk scores identify distinct types of metabolic dysfunction-associated steatotic liver disease. RESEARCH SQUARE 2024:rs.3.rs-3878807. [PMID: 38405802 PMCID: PMC10889080 DOI: 10.21203/rs.3.rs-3878807/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) encompasses an excess of triglycerides in the liver, which can lead to cirrhosis and liver cancer. While there is solid epidemiological evidence of MASLD coexisting with cardiometabolic disease, several leading genetic risk factors for MASLD do not increase the risk of cardiovascular disease, suggesting no causal relationship between MASLD and cardiometabolic derangement. In this work, we leveraged measurements of visceral adiposity and identified 27 novel genetic loci associated with MASLD. Among these loci, we replicated 6 in several independent cohorts. Next, we generated two partitioned polygenic risk scores (PRS) based on the mechanism of genetic association with MASLD encompassing intra-hepatic lipoprotein retention. The two PRS suggest the presence of at least two distinct types of MASLD, one confined to the liver resulting in a more aggressive liver disease and one that is systemic and results in a higher risk of cardiometabolic disease.
Collapse
Affiliation(s)
- Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg
| | | | - Antonio De Vincentis
- Operative Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | | | | | | | - Rosellina Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Chung SW, Choi J. Editorial: Paradox of liver fat content in the spectrum of non-alcoholic fatty liver disease. Aliment Pharmacol Ther 2024; 59:569-570. [PMID: 38247152 DOI: 10.1111/apt.17809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
LINKED CONTENTThis article is linked to Loomba et al paper. To view this article, visit https://doi.org/10.1111/apt.17783
Collapse
Affiliation(s)
- Sung Won Chung
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jonggi Choi
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
50
|
de Souza Echeverria L, Mounzer DLS, Gestic MA, Utrini MP, Chaim FDM, Callejas-Neto F, Chaim EA, Cazzo E. Fibrotic NASH in Individuals with Obesity: a Cross-sectional Analysis of the Prevalence of this Significant Milestone of Disease Progression and Accuracy of a Non-invasive Marker for its Screening. Obes Surg 2024; 34:389-395. [PMID: 38110785 DOI: 10.1007/s11695-023-06998-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Fibrotic non-alcoholic steatohepatitis (NASH), i.e., the concomitant presence of active inflammation and fibrosis, represents a milestone in the natural history of NAFLD and a critical time point in its progression. The purpose of this study was to analyze the diagnostic accuracy of the non-invasive Fibrotic NASH Index (FNI) in individuals with obesity undergoing bariatric surgery. METHODS This is a cross-sectional study, enrolling individuals who underwent bariatric surgery with liver biopsy at a tertiary university hospital. FNI was calculated, and a cutoff value was determined. Its diagnostic accuracy was then calculated through comparison with the gold standard test for this analysis (histopathological examination). RESULTS Of 128 participants, 83.6% were female, and the average age was 39.8 ± 8.7 years. The mean BMI was 38.7 ± 5.7 kg/m2. NAFLD was histologically confirmed in 76.6%, of which 81.6% had NASH. Histologically confirmed fibrotic NASH was observed in 22.7% of the general study population, 29.6% of individuals with NAFLD, and 36.3% of those with NASH. The mean FNI was 0.18 ± 0.19. An optimal cutoff point of 0.21 was determined, with an overall accuracy of 90.1%, an 82.8% sensitivity, a 90.8% specificity, a 72.6% positive predictive value, and a 94.7% negative predictive value. CONCLUSIONS FNI provided adequate accuracy in detecting and ruling out fibrotic NASH. Considering the importance of fibrotic NASH within the natural history of NAFLD progression and the fact that this marker uses simple variables, it may be of great importance in high-risk populations, and its external validation and use should be encouraged.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Everton Cazzo
- Dept. of Surgery, State University of Campinas (UNICAMP), Campinas, Brazil.
| |
Collapse
|