Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome
Abstract
:1. Introduction
2. Change of Nomenclature
3. Underlying Mechanisms
3.1. Insulin Resistance: The Central Pathway
3.2. Adipose Tissue Dysfunction and Pro-Inflammatory State
3.3. ER Stress and Oxidative Stress: Amplifying Liver Injury
3.4. Gut Microbiota Dysbiosis and Metabolic-Endotoxemia
3.5. Innate Immune Response and Hepatic Inflammation
3.6. Fibrosis Pathways and Liver Disease Progression
4. Extrahepatic Co-Morbidities
4.1. Cardiovascular Morbidity and Mortality
4.2. Progression of Liver Disease
4.3. Type 2 Diabetes Mellitus
4.4. Extrahepatic Complications
4.4.1. Chronic Kidney Disease (CKD)
4.4.2. Obstructive Sleep Apnea (OSA)
4.4.3. Osteoporosis
4.4.4. Endocrine Disorders
Polycystic Ovary Syndrome
Thyroid Dysfunction
4.4.5. Implications for Clinical Management
5. Therapeutic Strategies and Future Directions
5.1. Lifestyle Modifications
5.2. Pharmacological Interventions
5.2.1. Insulin Sensitizers
5.2.2. GLP-1 Receptor Agonists
5.2.3. SGLT2 Inhibitors
5.2.4. Statins
6. Emerging Therapies
6.1. FXR Agonists
6.2. Acetyl-CoA Carboxylase (ACC) Inhibitors
6.3. Thyroid Hormone Receptor (THR)-β Agonists
6.4. Antibiotics
6.5. Novel Anti-Inflammatory and Anti-Fibrotic Agents
7. Future Directions
7.1. Precision Medicine Approaches
7.2. Microbiome Modulation
7.3. The Role of Artificial Intelligence in Prediction, Diagnosis, and Treatment
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
ACC | Acetyl-CoA carboxylase |
ACC1 | Acetyl-CoA carboxylase 1 (Cytosolic Isoform) |
ACC2 | Acetyl-CoA carboxylase 2 (Mitochondrial Isoform) |
AHI | Apnea–hypopnea index |
AKT | Protein kinase B |
ALD | Alcoholic liver disease |
ALT | Alanine aminotransferase |
AMPK | AMP-activated protein kinase |
Apo AI | Apolipoprotein A-I |
AST | Aspartate aminotransferase |
BA | Bile acid |
BALANCED | A study name related to FGF21 |
BMD | Bone mineral density |
BMI | Body mass index |
BSEP | Bile salt export pump |
CAD | Coronary artery disease |
CAFLD | Combined causes of fatty liver disease |
CAPE | Caffeic acid phenethyl ester |
CCN2 | Cellular communication network 2 (another name for CTGF) |
CCR2/CCR5 | C-C chemokine receptors 2 and 5 |
Ch-REBP | Carbohydrate response element-binding protein |
CIH | Chronic Intermittent Hypoxia |
CK18 | Cytokeratin 18 |
CKD | Chronic kidney disease |
CRP | C-reactive Protein |
CTGF | Connective tissue growth factor |
CVD | Cardiovascular disease |
CYP7A1 | Cholesterol 7 Alpha-Hydroxylase |
CYP8B1 | Cytochrome P450 Family 8 Subfamily B Member 1 |
DGAT2 | Diacylglycerol O-acyltransferase 2 |
DIO1 | Deiodinase Type 1 |
DNL | De novo lipogenesis |
DPP-4 | Dipeptidyl Peptidase-4 |
DSF | Disulfiram |
EAT | Epicardial adipose tissue |
ECM | Extracellular matrix |
Epigenome | The chemical changes to DNA that regulate gene expression |
ER | Endoplasmic reticulum |
ERK | Extracellular signal-regulated kinase |
ERK1/2 | Extracellular signal-regulated kinase ½ |
ESFLD | End-stage fatty liver disease |
ESKD | End-stage kidney disease |
FAFLD | Advanced stage fibrotic fatty liver disease |
FDA | Food and Drug Administration |
FFA s | Free fatty acids |
FGF15 | Fibroblast growth factor 15 |
FGF19 | Fibroblast growth factor 19 |
FGF21 | Fibroblast growth factor 21 |
FGFR4 | Fibroblast growth factor receptor 4 |
FLD | Fatty Liver Disease |
FMT | Fecal Microbiota Transplantation |
FXR | Farnesoid X receptor |
GAFLD | Genetics-associated fatty liver disease |
GCKR | Glucokinase regulatory protein (Gene) |
Genome | The complete set of genes or genetic material in an organism |
GIP | Glucose-Dependent Insulinotropic Polypeptide |
GLP-1 | Glucagon-Like Peptide-1 |
GS-0976 | Firsocostat (Gilead’s ACC inhibitor) |
H2S | Hydrogen sulphide |
HARMONY | A phase 2 trial name |
HCC | Hepatocellular carcinoma |
HDL-C | High-density lipoprotein cholesterol |
HMG-CoA | 3-Hydroxy-3-Methylglutaryl-Coenzyme A |
HOMA-IR | Homeostasis Model Assessment of Insulin Resistance |
HSC | Hepatic stellate cells |
Hs-CRP | Highly Sensitive C-Reactive Protein |
HSCs | Hepatic stellate cells |
IBS | Irritable bowel syndrome |
IL-18 | Interleukin-18 |
IL-1β | Interleukin-1β |
IL-31 | Interleukin 31 |
IL-6 | Interleukin-6 |
IR | Insulin resistance |
JNK | c-Jun N-terminal Kinase |
LAFLD | Lipodystrophy-associated fatty liver disease |
LDL-C | Low-density lipoprotein cholesterol |
LDL-R | Low-density lipoprotein receptor |
LOX | Lysyl oxidase |
LOXL2 | Lysyl oxidase Like 2 |
LPO | Lipid peroxidation |
LPS | Lipopolysaccharide |
LSECs | Liver sinusoidal endothelial cells |
M1 | Macrophage Type 1 (Pro-inflammatory) |
MASLD | Metabolic dysfunction-associated fatty liver disease |
MASH | Metabolic dysfunction-associated steatohepatitis |
lMASLD | Metabolic dysfunction-associated steatotic liver disease |
MBOAT7 | Membrane Bound O-acyltransferase Domain-Containing 7 (Gene) |
MCP-1 | Monocyte Chemoattractant Protein-1 |
Metabolome | The complete set of small-molecule metabolites in an organism |
MetS | Metabolic syndrome |
Microbiome | The community of microorganisms living in and on the body |
MK-4074 | An MSD drug (ACC inhibitor) |
mTOR | Mechanistic Target of Rapamycin |
MUFA | Mono-unsaturated fatty acids |
NAFLD | Non-alcoholic fatty liver disease |
NAS | Non-Alcoholic Fatty Liver Disease Activity Score |
NASH | Non-alcoholic steatohepatitis |
NLRP3 | NACHT, LRR and PYD domains-containing protein 3 (inflammasome) |
NO | Nitric oxide |
NTCP | Sodium-Taurocholate Cotransporting Polypeptide |
OAFLD | Obesity-associated fatty liver disease |
OATP1 | Organic Anion Transporter Polypeptide 1 |
OCA | Obeticholic acid |
OSA | Obstructive Sleep Apnea |
PDGF | Platelet-derived growth factor |
PF-05175157 | A Pfizer drug (ACC1 inhibitor) |
PF-05221304 | A Pfizer drug (ACC2 inhibitor) |
PI3K | Phosphoinositide 3-Kinase |
PNPLA3 | Patatin-Like Phospholipase Domain-Containing 3 (Gene) |
PON1 | Paraoxonase 1 |
PPAR-α | Peroxisome Proliferator-Activated Receptor-α |
PROC3,4,8 | Proinflammatory Cytokine 3, 4, and 8 |
Proteome | The entire set of proteins produced by an organism |
PUFA | Polyunsaturated fatty acids |
Ras | Rat Sarcoma (a family of proteins) |
RhoA | Ras Homolog Family Member A |
ROS | Reactive oxygen species |
SAFLD | Sarcopenia-associated fatty liver disease |
SCFA s | Short-chain fatty acids |
SGLT | Sodium-Glucose Cotransporter |
SGLT1 | Sodium-Glucose Cotransporter 1 |
SGLT2 | Sodium-Glucose Cotransporter 2 |
SHP | Small heterodimer partner |
SIRT1 | Sirtuin 1 |
Smad | Suppressor of Mothers against Decapentaplegic |
SNPs | Single Nucleotide Polymorphisms (genetic variations) |
SREBP-1c | Sterol regulatory element-binding protein 1c |
SSB s | Sugar-sweetened beverages |
T2D | Type 2 diabetes |
T3 | Triiodothyronine |
T4 | Thyroxine |
TC | Total cholesterol |
TG | Triglycerides |
TGF-β | Transforming growth factor beta |
TGR5 | G Protein-Coupled Bile Acid Receptor 1 |
THR-α | Thyroid hormone receptor alpha |
THR-β | Thyroid hormone receptor beta |
TLR4 | Toll-Like Receptor 4 |
TM6SF2 | Transmembrane 6 Superfamily Member 2 (Gene) |
TNF-α | Tumor necrosis factor alpha |
Transcriptome | The full range of messenger RNA molecules expressed by an organism |
TSB | Total Stool Bacteria |
TSH | Thyroid-stimulating hormone |
TZD | Thiazolidinediones |
UPR | Unfolded protein response |
Vitamin D | Vitamin D (commonly referred in discussions about bone health) |
XAFLD | Fatty liver disease of multiple/unknown causes |
β-oxidation | Beta-Oxidation |
β-trophin | Beta-Trophin |
References
- Eslam, M.; Sanyal, A.J.; George, J.; on behalf of the International Consensus Panel. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014.e1. [Google Scholar] [CrossRef] [PubMed]
- Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12. [Google Scholar] [CrossRef] [PubMed]
- Rich, N.E.; Oji, S.; Mufti, A.R.; Browning, J.D.; Parikh, N.D.; Odewole, M.; Mayo, H.; Singal, A.G. Racial and Ethnic Disparities in Nonalcoholic Fatty Liver Disease Prevalence, Severity, and Outcomes in the United States: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2018, 16, 198–210.e2. [Google Scholar] [CrossRef] [PubMed]
- Lazo, M.; Hernaez, R.; Eberhardt, M.S.; Bonekamp, S.; Kamel, I.; Guallar, E.; Koteish, A.; Brancati, F.L.; Clark, J.M. Prevalence of nonalcoholic fatty liver disease in the United States: The Third National Health and Nutrition Examination Survey, 1988-1994. Am. J. Epidemiol. 2013, 178, 38–45. [Google Scholar] [CrossRef]
- Huang, Q.; Zou, X.; Wen, X.; Zhou, X.; Ji, L. NAFLD or MAFLD: Which Has Closer Association With All-Cause and Cause-Specific Mortality?-Results From NHANES III. Front. Med. 2021, 8, 693507. [Google Scholar] [CrossRef]
- Zou, B.; Yeo, Y.H.; Nguyen, V.H.; Cheung, R.; Ingelsson, E.; Nguyen, M.H. Prevalence, characteristics and mortality outcomes of obese, nonobese and lean NAFLD in the United States, 1999–2016. J. Intern. Med. 2020, 288, 139–151. [Google Scholar] [CrossRef]
- Li, J.; Zou, B.; Yeo, Y.H.; Feng, Y.; Xie, X.; Lee, D.H.; Fujii, H.; Wu, Y.; Kam, L.Y.; Ji, F.; et al. Prevalence, incidence, and outcome of non-alcoholic fatty liver disease in Asia, 1999-2019: A systematic review and meta-analysis. Lancet Gastroenterol. Hepatol. 2019, 4, 389–398. [Google Scholar] [CrossRef]
- Estes, C.; Anstee, Q.M.; Arias-Loste, M.T.; Bantel, H.; Bellentani, S.; Caballeria, J.; Colombo, M.; Craxi, A.; Crespo, J.; Day, C.P.; et al. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016–2030. J. Hepatol. 2018, 69, 896–904. [Google Scholar] [CrossRef]
- Huang, D.Q.; El-Serag, H.B.; Loomba, R. Global epidemiology of NAFLD-related HCC: Trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 223–238. [Google Scholar] [CrossRef]
- Byrne, C.D.; Olufadi, R.; Bruce, K.D.; Cagampang, F.R.; Ahmed, M.H. Metabolic disturbances in non-alcoholic fatty liver disease. Clin. Sci. Lond. Engl. 1979 2009, 116, 539–564. [Google Scholar] [CrossRef]
- Gastaldelli, A.; Cusi, K. From NASH to diabetes and from diabetes to NASH: Mechanisms and treatment options. JHEP Rep. Innov. Hepatol. 2019, 1, 312–328. [Google Scholar] [CrossRef]
- Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 2006, 444, 860–867. [Google Scholar] [CrossRef] [PubMed]
- Shulman, G.I. Cellular mechanisms of insulin resistance. J. Clin. Investig. 2000, 106, 171–176. [Google Scholar] [CrossRef] [PubMed]
- Czech, M.P. Mechanisms of insulin resistance related to white, beige, and brown adipocytes. Mol. Metab. 2020, 34, 27–42. [Google Scholar] [CrossRef]
- Ye, J. Mechanisms of insulin resistance in obesity. Front. Med. 2013, 7, 14–24. [Google Scholar] [CrossRef]
- Saltiel, A.R.; Olefsky, J.M. Inflammatory mechanisms linking obesity and metabolic disease. J. Clin. Investig. 2017, 127, 1–4. [Google Scholar] [CrossRef]
- Haas, J.T.; Francque, S.; Staels, B. Pathophysiology and Mechanisms of Nonalcoholic Fatty Liver Disease. Annu. Rev. Physiol. 2016, 78, 181–205. [Google Scholar] [CrossRef]
- Stefan, N.; Häring, H.-U.; Cusi, K. Non-alcoholic fatty liver disease: Causes, diagnosis, cardiometabolic consequences, and treatment strategies. Lancet Diabetes Endocrinol. 2019, 7, 313–324. [Google Scholar] [CrossRef]
- Willner, I.R.; Waters, B.; Patil, S.R.; Reuben, A.; Morelli, J.; Riely, C.A. Ninety patients with nonalcoholic steatohepatitis: Insulin resistance, familial tendency, and severity of disease. Am. J. Gastroenterol. 2001, 96, 2957–2961. [Google Scholar] [CrossRef]
- Struben, V.M.D.; Hespenheide, E.E.; Caldwell, S.H. Nonalcoholic steatohepatitis and cryptogenic cirrhosis within kindreds. Am. J. Med. 2000, 108, 9–13. [Google Scholar] [CrossRef]
- Zeng, J.; Yang, R.-X.; Sun, C.; Pan, Q.; Zhang, R.-N.; Chen, G.-Y.; Hu, Y.; Fan, J.-G. Prevalence, clinical characteristics, risk factors, and indicators for lean Chinese adults with nonalcoholic fatty liver disease. World J. Gastroenterol. 2020, 26, 1792–1804. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Du, X.; Kuppa, A.; Feitosa, M.F.; Bielak, L.F.; O’Connell, J.R.; Musani, S.K.; Guo, X.; Kahali, B.; Chen, V.L.; et al. Genome-wide association meta-analysis identifies 17 loci associated with nonalcoholic fatty liver disease. Nat. Genet. 2023, 55, 1640–1650. [Google Scholar] [CrossRef] [PubMed]
- Wong, V.W.-S.; Adams, L.A.; de Lédinghen, V.; Wong, G.L.-H.; Sookoian, S. Noninvasive biomarkers in NAFLD and NASH — current progress and future promise. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 461–478. [Google Scholar] [CrossRef] [PubMed]
- Vilar-Gomez, E.; Yasells-Garcia, A.; Martinez-Perez, Y.; Calzadilla-Bertot, L.; Torres-Gonzalez, A.; Gra-Oramas, B.; Gonzalez-Fabian, L.; Villa-Jimenez, O.; Friedman, S.L.; Diago, M.; et al. Development and validation of a noninvasive prediction model for nonalcoholic steatohepatitis resolution after lifestyle intervention. Hepatology 2016, 63, 1875–1887. [Google Scholar] [CrossRef]
- Nam, D.; Chapiro, J.; Paradis, V.; Seraphin, T.P.; Kather, J.N. Artificial intelligence in liver diseases: Improving diagnostics, prognostics and response prediction. JHEP Rep. 2022, 4, 100443. [Google Scholar] [CrossRef]
- Lim, S.; Kim, J.-W.; Targher, G. Links between metabolic syndrome and metabolic dysfunction-associated fatty liver disease. Trends Endocrinol. Metab. 2021, 32, 500–514. [Google Scholar] [CrossRef]
- Ludwig, J.; Viggiano, T.R.; McGILL, D.B.; Ott, B.J. Nonalcoholic Steatohepatitis Mayo Clinic Experiences With a Hitherto Unnamed Disease. Mayo Clin. Proc. 1980, 55, 434–438. [Google Scholar] [CrossRef]
- Alberti, K.G.M.M.; Eckel, R.H.; Grundy, S.M.; Zimmet, P.Z.; Cleeman, J.I.; Donato, K.A.; Fruchart, J.-C.; James, W.P.T.; Loria, C.M.; Smith, S.C.; et al. Harmonizing the metabolic syndrome: A joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation 2009, 120, 1640–1645. [Google Scholar] [CrossRef]
- Younossi, Z.M.; Rinella, M.E.; Sanyal, A.J.; Harrison, S.A.; Brunt, E.M.; Goodman, Z.; Cohen, D.E.; Loomba, R. From NAFLD to MAFLD: Implications of a Premature Change in Terminology. Hepatology 2021, 73, 1194–1198. [Google Scholar] [CrossRef]
- Stefan, N.; Häring, H.-U. The role of hepatokines in metabolism. Nat. Rev. Endocrinol. 2013, 9, 144–152. [Google Scholar] [CrossRef]
- Meex, R.C.R.; Watt, M.J. Hepatokines: Linking nonalcoholic fatty liver disease and insulin resistance. Nat. Rev. Endocrinol. 2017, 13, 509–520. [Google Scholar] [CrossRef] [PubMed]
- Spahis, S.; Delvin, E.; Borys, J.-M.; Levy, E. Oxidative Stress as a Critical Factor in Nonalcoholic Fatty Liver Disease Pathogenesis. Antioxid. Redox Signal. 2017, 26, 519–541. [Google Scholar] [CrossRef] [PubMed]
- Mridha, A.R.; Wree, A.; Robertson, A.A.B.; Yeh, M.M.; Johnson, C.D.; Van Rooyen, D.M.; Haczeyni, F.; Teoh, N.C.-H.; Savard, C.; Ioannou, G.N.; et al. NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J. Hepatol. 2017, 66, 1037–1046. [Google Scholar] [CrossRef] [PubMed]
- Polyzos, S.A.; Kountouras, J.; Mantzoros, C.S. Adipokines in nonalcoholic fatty liver disease. Metabolism 2016, 65, 1062–1079. [Google Scholar] [CrossRef]
- Lonardo, A.; Nascimbeni, F.; Mantovani, A.; Targher, G. Hypertension, diabetes, atherosclerosis and NASH: Cause or consequence? J. Hepatol. 2018, 68, 335–352. [Google Scholar] [CrossRef]
- Longo, M.; Zatterale, F.; Naderi, J.; Parrillo, L.; Formisano, P.; Raciti, G.A.; Beguinot, F.; Miele, C. Adipose Tissue Dysfunction as Determinant of Obesity-Associated Metabolic Complications. Int. J. Mol. Sci. 2019, 20, 2358. [Google Scholar] [CrossRef]
- Simões, I.C.M.; Fontes, A.; Pinton, P.; Zischka, H.; Wieckowski, M.R. Mitochondria in non-alcoholic fatty liver disease. Int. J. Biochem. Cell Biol. 2018, 95, 93–99. [Google Scholar] [CrossRef]
- Grattagliano, I.; de Bari, O.; Bernardo, T.C.; Oliveira, P.J.; Wang, D.Q.-H.; Portincasa, P. Role of mitochondria in nonalcoholic fatty liver disease--from origin to propagation. Clin. Biochem. 2012, 45, 610–618. [Google Scholar] [CrossRef]
- Leung, C.; Rivera, L.; Furness, J.B.; Angus, P.W. The role of the gut microbiota in NAFLD. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 412–425. [Google Scholar] [CrossRef]
- Arab, J.P.; Karpen, S.J.; Dawson, P.A.; Arrese, M.; Trauner, M. Bile acids and nonalcoholic fatty liver disease: Molecular insights and therapeutic perspectives. Hepatology 2017, 65, 350–362. [Google Scholar] [CrossRef]
- Koh, A.; Molinaro, A.; Ståhlman, M.; Khan, M.T.; Schmidt, C.; Mannerås-Holm, L.; Wu, H.; Carreras, A.; Jeong, H.; Olofsson, L.E.; et al. Microbially Produced Imidazole Propionate Impairs Insulin Signaling through mTORC1. Cell 2018, 175, 947–961.e17. [Google Scholar] [CrossRef]
- Albillos, A.; de Gottardi, A.; Rescigno, M. The gut-liver axis in liver disease: Pathophysiological basis for therapy. J. Hepatol. 2020, 72, 558–577. [Google Scholar] [CrossRef] [PubMed]
- Ekstedt, M.; Hagström, H.; Nasr, P.; Fredrikson, M.; Stål, P.; Kechagias, S.; Hultcrantz, R. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 2015, 61, 1547–1554. [Google Scholar] [CrossRef] [PubMed]
- Younossi, Z.M.; Golabi, P.; Paik, J.M.; Henry, A.; Van Dongen, C.; Henry, L. The global epidemiology of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH): A systematic review. Hepatology 2023, 77, 1335–1347. [Google Scholar] [CrossRef] [PubMed]
- Eslam, M.; Newsome, P.N.; Sarin, S.K.; Anstee, Q.M.; Targher, G.; Romero-Gomez, M.; Zelber-Sagi, S.; Wai-Sun Wong, V.; Dufour, J.-F.; Schattenberg, J.M.; et al. A new definition for metabolic dysfunction-associated fatty liver disease: An international expert consensus statement. J. Hepatol. 2020, 73, 202–209. [Google Scholar] [CrossRef]
- Mantovani, A.; Petracca, G.; Beatrice, G.; Csermely, A.; Tilg, H.; Byrne, C.D.; Targher, G. Non-alcoholic fatty liver disease and increased risk of incident extrahepatic cancers: A meta-analysis of observational cohort studies. Gut 2022, 71, 778–788. [Google Scholar] [CrossRef]
- Mottillo, S.; Filion, K.B.; Genest, J.; Joseph, L.; Pilote, L.; Poirier, P.; Rinfret, S.; Schiffrin, E.L.; Eisenberg, M.J. The metabolic syndrome and cardiovascular risk a systematic review and meta-analysis. J. Am. Coll. Cardiol. 2010, 56, 1113–1132. [Google Scholar] [CrossRef]
- Targher, G.; Byrne, C.D.; Lonardo, A.; Zoppini, G.; Barbui, C. Non-alcoholic fatty liver disease and risk of incident cardiovascular disease: A meta-analysis. J. Hepatol. 2016, 65, 589–600. [Google Scholar] [CrossRef]
- Brunner, K.T.; Pedley, A.; Massaro, J.M.; Hoffmann, U.; Benjamin, E.J.; Long, M.T. Increasing liver fat is associated with progression of cardiovascular risk factors. Liver Int. Off. J. Int. Assoc. Study Liver 2020, 40, 1339–1343. [Google Scholar] [CrossRef]
- Basheer, M.; Saad, E.; Jeries, H.; Assy, N. Liver Fat Storage Is a Better Predictor of Coronary Artery Disease than Visceral Fat. Metabolites 2023, 13, 896. [Google Scholar] [CrossRef]
- Eslam, M.; George, J. Refining the role of epicardial adipose tissue in non-alcoholic fatty liver disease. Hepatol. Int. 2019, 13, 662–664. [Google Scholar] [CrossRef]
- Gutiérrez-Cuevas, J.; Santos, A.; Armendariz-Borunda, J. Pathophysiological Molecular Mechanisms of Obesity: A Link between MAFLD and NASH with Cardiovascular Diseases. Int. J. Mol. Sci. 2021, 22, 11629. [Google Scholar] [CrossRef] [PubMed]
- Adler, M.; Schaffner, F. Fatty liver hepatitis and cirrhosis in obese patients. Am. J. Med. 1979, 67, 811–816. [Google Scholar] [CrossRef]
- Ballestri, S.; Lonardo, A.; Bonapace, S.; Byrne, C.D.; Loria, P.; Targher, G. Risk of cardiovascular, cardiac and arrhythmic complications in patients with non-alcoholic fatty liver disease. World J. Gastroenterol. 2014, 20, 1724–1745. [Google Scholar] [CrossRef]
- Gaggini, M.; Morelli, M.; Buzzigoli, E.; DeFronzo, R.A.; Bugianesi, E.; Gastaldelli, A. Non-alcoholic fatty liver disease (NAFLD) and its connection with insulin resistance, dyslipidemia, atherosclerosis and coronary heart disease. Nutrients 2013, 5, 1544–1560. [Google Scholar] [CrossRef]
- Santos, R.D.; Valenti, L.; Romeo, S. Does nonalcoholic fatty liver disease cause cardiovascular disease? Current knowledge and gaps. Atherosclerosis 2019, 282, 110–120. [Google Scholar] [CrossRef]
- Anstee, Q.M.; Mantovani, A.; Tilg, H.; Targher, G. Risk of cardiomyopathy and cardiac arrhythmias in patients with nonalcoholic fatty liver disease. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 425–439. [Google Scholar] [CrossRef]
- Taylor, R.S.; Taylor, R.J.; Bayliss, S.; Hagström, H.; Nasr, P.; Schattenberg, J.M.; Ishigami, M.; Toyoda, H.; Wai-Sun Wong, V.; Peleg, N.; et al. Association Between Fibrosis Stage and Outcomes of Patients With Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis. Gastroenterology 2020, 158, 1611–1625.e12. [Google Scholar] [CrossRef]
- Vilar-Gomez, E.; Athinarayanan, S.J.; Adams, R.N.; Hallberg, S.J.; Bhanpuri, N.H.; McKenzie, A.L.; Campbell, W.W.; McCarter, J.P.; Phinney, S.D.; Volek, J.S.; et al. Post hoc analyses of surrogate markers of non-alcoholic fatty liver disease (NAFLD) and liver fibrosis in patients with type 2 diabetes in a digitally supported continuous care intervention: An open-label, non-randomised controlled study. BMJ Open 2019, 9, e023597. [Google Scholar] [CrossRef]
- Friedman, S.L.; Neuschwander-Tetri, B.A.; Rinella, M.; Sanyal, A.J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 2018, 24, 908–922. [Google Scholar] [CrossRef]
- Marra, F.; Svegliati-Baroni, G. Lipotoxicity and the gut-liver axis in NASH pathogenesis. J. Hepatol. 2018, 68, 280–295. [Google Scholar] [CrossRef] [PubMed]
- Schwabe, R.F.; Tabas, I.; Pajvani, U.B. Mechanisms of Fibrosis Development in Nonalcoholic Steatohepatitis. Gastroenterology 2020, 158, 1913–1928. [Google Scholar] [CrossRef] [PubMed]
- Arab, J.P.; Izzy, M.; Leggio, L.; Bataller, R.; Shah, V.H. Management of alcohol use disorder in patients with cirrhosis in the setting of liver transplantation. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 45–59. [Google Scholar] [CrossRef] [PubMed]
- Kleiner, D.E.; Brunt, E.M.; Wilson, L.A.; Behling, C.; Guy, C.; Contos, M.; Cummings, O.; Yeh, M.; Gill, R.; Chalasani, N.; et al. Association of Histologic Disease Activity With Progression of Nonalcoholic Fatty Liver Disease. JAMA Netw. Open 2019, 2, e1912565. [Google Scholar] [CrossRef]
- Zhao, Y.; Zhao, W.; Ma, J.; Toshiyoshi, M.; Zhao, Y. Patatin-like phospholipase domain-containing 3 gene (PNPLA3) polymorphic (rs738409) single nucleotide polymorphisms and susceptibility to nonalcoholic fatty liver disease: A meta-analysis of twenty studies. Medicine 2023, 102, e33110. [Google Scholar] [CrossRef]
- Tian, L.-Y.; Smit, D.J.; Jücker, M. The Role of PI3K/AKT/mTOR Signaling in Hepatocellular Carcinoma Metabolism. Int. J. Mol. Sci. 2023, 24, 2652. [Google Scholar] [CrossRef]
- Anstee, Q.M.; Darlay, R.; Cockell, S.; Meroni, M.; Govaere, O.; Tiniakos, D.; Burt, A.D.; Bedossa, P.; Palmer, J.; Liu, Y.-L.; et al. Genome-wide association study of non-alcoholic fatty liver and steatohepatitis in a histologically characterised cohort☆. J. Hepatol. 2020, 73, 505–515. [Google Scholar] [CrossRef]
- Wong, V.W.-S.; Wong, G.L.-H.; Woo, J.; Abrigo, J.M.; Chan, C.K.-M.; Shu, S.S.-T.; Leung, J.K.-Y.; Chim, A.M.-L.; Kong, A.P.-S.; Lui, G.C.-Y.; et al. Impact of the New Definition of Metabolic Associated Fatty Liver Disease on the Epidemiology of the Disease. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2021, 19, 2161–2171.e5. [Google Scholar] [CrossRef]
- Mantovani, A.; Byrne, C.D.; Bonora, E.; Targher, G. Nonalcoholic Fatty Liver Disease and Risk of Incident Type 2 Diabetes: A Meta-analysis. Diabetes Care 2018, 41, 372–382. [Google Scholar] [CrossRef]
- Bhat, N.; Mani, A. Dysregulation of Lipid and Glucose Metabolism in Nonalcoholic Fatty Liver Disease. Nutrients 2023, 15, 2323. [Google Scholar] [CrossRef]
- Sakurai, Y.; Kubota, N.; Yamauchi, T.; Kadowaki, T. Role of Insulin Resistance in MAFLD. Int. J. Mol. Sci. 2021, 22, 4156. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.-D.; Cai, J.; Targher, G.; Byrne, C.D.; Shapiro, M.D.; Sung, K.-C.; Somers, V.K.; Chahal, C.A.A.; George, J.; Chen, L.-L.; et al. Metabolic dysfunction-associated fatty liver disease and implications for cardiovascular risk and disease prevention. Cardiovasc. Diabetol. 2022, 21, 270. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.; Lee, D.S.; An, T.H.; Park, H.-J.; Kim, W.K.; Bae, K.-H.; Oh, K.-J. Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC. Int. J. Mol. Sci. 2021, 22, 4495. [Google Scholar] [CrossRef] [PubMed]
- Hudish, L.I.; Reusch, J.E.; Sussel, L. β Cell dysfunction during progression of metabolic syndrome to type 2 diabetes. J. Clin. Investig. 2019, 129, 4001–4008. [Google Scholar] [CrossRef]
- Heindel, J.J.; Blumberg, B.; Cave, M.; Machtinger, R.; Mantovani, A.; Mendez, M.A.; Nadal, A.; Palanza, P.; Panzica, G.; Sargis, R.; et al. Metabolism disrupting chemicals and metabolic disorders. Reprod. Toxicol. 2017, 68, 3–33. [Google Scholar] [CrossRef]
- Vilas-Boas, E.A.; Almeida, D.C.; Roma, L.P.; Ortis, F.; Carpinelli, A.R. Lipotoxicity and β-Cell Failure in Type 2 Diabetes: Oxidative Stress Linked to NADPH Oxidase and ER Stress. Cells 2021, 10, 3328. [Google Scholar] [CrossRef]
- Lu, Y.; Wang, W.; Liu, J.; Xie, M.; Liu, Q.; Li, S. Vascular complications of diabetes: A narrative review. Medicine 2023, 102, e35285. [Google Scholar] [CrossRef]
- Zhou, Z.; Sun, B.; Huang, S.; Zhu, C.; Bian, M. Glycemic variability: Adverse clinical outcomes and how to improve it? Cardiovasc. Diabetol. 2020, 19, 102. [Google Scholar] [CrossRef]
- Ferdous, S.-E.; Ferrell, J.M. Pathophysiological Relationship between Type 2 Diabetes Mellitus and Metabolic Dysfunction-Associated Steatotic Liver Disease: Novel Therapeutic Approaches. Int. J. Mol. Sci. 2024, 25, 8731. [Google Scholar] [CrossRef]
- Musso, G.; Gambino, R.; Tabibian, J.H.; Ekstedt, M.; Kechagias, S.; Hamaguchi, M.; Hultcrantz, R.; Hagström, H.; Yoon, S.K.; Charatcharoenwitthaya, P.; et al. Association of non-alcoholic fatty liver disease with chronic kidney disease: A systematic review and meta-analysis. PLoS Med. 2014, 11, e1001680. [Google Scholar] [CrossRef]
- Chen, S.; Pang, J.; Huang, R.; Xue, H.; Chen, X. Association of MAFLD with end-stage kidney disease: A prospective study of 337,783 UK Biobank participants. Hepatol. Int. 2023, 17, 595–605. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.-J.; Wang, Y.-Y.; Chen, C.; Lu, Y.-L.; Wang, N.-J. Cardiovascular and renal burdens of metabolic associated fatty liver disease from serial US national surveys, 1999–2016. Chin. Med. J. 2021, 134, 1593–1601. [Google Scholar] [CrossRef] [PubMed]
- Byrne, C.D.; Targher, G. NAFLD as a driver of chronic kidney disease. J. Hepatol. 2020, 72, 785–801. [Google Scholar] [CrossRef]
- Tang, H.; Lv, F.; Zhang, P.; Liu, J.; Mao, J. The impact of obstructive sleep apnea on nonalcoholic fatty liver disease. Front. Endocrinol. 2023, 14, 1254459. [Google Scholar] [CrossRef]
- Aron-Wisnewsky, J.; Clement, K.; Pépin, J.-L. Nonalcoholic fatty liver disease and obstructive sleep apnea. Metabolism 2016, 65, 1124–1135. [Google Scholar] [CrossRef]
- Influence of Obstructive Sleep Apnea on Fatty Liver Disease: Role of Chronic Intermittent Hypoxia—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/21762556/ (accessed on 6 February 2025).
- Sookoian, S.; Pirola, C.J. Obstructive sleep apnea is associated with fatty liver and abnormal liver enzymes: A meta-analysis. Obes. Surg. 2013, 23, 1815–1825. [Google Scholar] [CrossRef]
- Aron-Wisnewsky, J.; Minville, C.; Tordjman, J.; Lévy, P.; Bouillot, J.-L.; Basdevant, A.; Bedossa, P.; Clément, K.; Pépin, J.-L. Chronic intermittent hypoxia is a major trigger for non-alcoholic fatty liver disease in morbid obese. J. Hepatol. 2012, 56, 225–233. [Google Scholar] [CrossRef]
- Wang, D.; Si, D.; Li, G.; Ding, Z.; Yang, X.; Gao, C. Dysregulated autophagic activity induced in response to chronic intermittent hypoxia contributes to the pathogenesis of NAFLD. Front. Physiol. 2022, 13, 941706. [Google Scholar] [CrossRef]
- Global ETD Search. Available online: http://search.ndltd.org/show.php?id=oai%3Aunion.ndltd.org%3Abu.edu%2Foai%3Aopen.bu.edu%3A2144%2F31230&back=http%3A%2F%2Fsearch.ndltd.org%2Fsearch.php%3Fq%3Dsubject%253A%2522OSA%2522%26start%3D10 (accessed on 6 February 2025).
- NASH in Lean Individuals—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/30654392/ (accessed on 6 February 2025).
- Mesarwi, O.A.; Shin, M.-K.; Drager, L.F.; Bevans-Fonti, S.; Jun, J.C.; Putcha, N.; Torbenson, M.S.; Pedrosa, R.P.; Lorenzi-Filho, G.; Steele, K.E.; et al. Lysyl Oxidase as a Serum Biomarker of Liver Fibrosis in Patients with Severe Obesity and Obstructive Sleep Apnea. Sleep 2015, 38, 1583–1591. [Google Scholar] [CrossRef]
- Ji, Y.; Liang, Y.; Mak, J.C.W.; Ip, M.S.M. Obstructive sleep apnea, intermittent hypoxia and non-alcoholic fatty liver disease. Sleep Med. 2022, 95, 16–28. [Google Scholar] [CrossRef]
- Briançon-Marjollet, A.; Weiszenstein, M.; Henri, M.; Thomas, A.; Godin-Ribuot, D.; Polak, J. The impact of sleep disorders on glucose metabolism: Endocrine and molecular mechanisms. Diabetol. Metab. Syndr. 2015, 7, 25. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Grigoryev, D.N.; Ye, S.Q.; Thorne, L.; Schwartz, A.R.; Smith, P.L.; O’Donnell, C.P.; Polotsky, V.Y. Chronic intermittent hypoxia upregulates genes of lipid biosynthesis in obese mice. J. Appl. Physiol. Bethesda Md 1985 2005, 99, 1643–1648. [Google Scholar] [CrossRef]
- Liu, J.; Li, W.; Zhu, W.; He, W.; Zhao, H.; Xiang, Y.; Liu, C.; Wu, W. Chronic intermittent hypoxia promotes the development of experimental non-alcoholic steatohepatitis by modulating Treg/Th17 differentiation. Acta Biochim. Biophys. Sin. 2018, 50, 1200–1210. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Zhang, J.; Lu, G.; Qiu, Y.; Yang, D.; Yin, G.; Zhang, X. The effect of oxidative stress in myocardial cell injury in mice exposed to chronic intermittent hypoxia. Chin. Med. J. 2010, 123, 74–78. [Google Scholar]
- Yan, Y.R.; Zhang, L.; Lin, Y.N.; Sun, X.W.; Ding, Y.J.; Li, N.; Li, H.P.; Li, S.Q.; Zhou, J.P.; Li, Q.Y. Chronic intermittent hypoxia-induced mitochondrial dysfunction mediates endothelial injury via the TXNIP/NLRP3/IL-1β signaling pathway. Free Radic. Biol. Med. 2021, 165, 401–410. [Google Scholar] [CrossRef]
- Aron-Wisnewsky, J.; Pepin, J.-L. New insights in the pathophysiology of chronic intermittent hypoxia-induced NASH: The role of gut-liver axis impairment. Thorax 2015, 70, 713–715. [Google Scholar] [CrossRef]
- Pardee, P.E.; Dunn, W.; Schwimmer, J.B. Non-alcoholic fatty liver disease is associated with low bone mineral density in obese children. Aliment. Pharmacol. Ther. 2012, 35, 248–254. [Google Scholar] [CrossRef]
- Moon, S.-S.; Lee, Y.-S.; Kim, S.W. Association of nonalcoholic fatty liver disease with low bone mass in postmenopausal women. Endocrine 2012, 42, 423–429. [Google Scholar] [CrossRef]
- Cui, R.; Sheng, H.; Rui, X.-F.; Cheng, X.-Y.; Sheng, C.-J.; Wang, J.-Y.; Qu, S. Low bone mineral density in chinese adults with nonalcoholic Fatty liver disease. Int. J. Endocrinol. 2013, 2013, 396545. [Google Scholar] [CrossRef]
- Barchetta, I.; Cimini, F.A.; Cavallo, M.G. Vitamin D and Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD): An Update. Nutrients 2020, 12, 3302. [Google Scholar] [CrossRef]
- Kwok, R.M.; Torres, D.M.; Harrison, S.A. Vitamin D and nonalcoholic fatty liver disease (NAFLD): Is it more than just an association? Hepatology 2013, 58, 1166–1174. [Google Scholar] [CrossRef] [PubMed]
- Drapkina, O.M.; Elkina, A.Y.; Sheptulina, A.F.; Kiselev, A.R. Non-Alcoholic Fatty Liver Disease and Bone Tissue Metabolism: Current Findings and Future Perspectives. Int. J. Mol. Sci. 2023, 24, 8445. [Google Scholar] [CrossRef] [PubMed]
- Filip, R.; Radzki, R.P.; Bieńko, M. Novel insights into the relationship between nonalcoholic fatty liver disease and osteoporosis. Clin. Interv. Aging 2018, 13, 1879–1891. [Google Scholar] [CrossRef] [PubMed]
- Santos, L.A.A.; Romeiro, F.G. Diagnosis and Management of Cirrhosis-Related Osteoporosis. BioMed Res. Int. 2016, 2016, 1423462. [Google Scholar] [CrossRef]
- Nakchbandi, I.A. Osteoporosis and fractures in liver disease: Relevance, pathogenesis and therapeutic implications. World J. Gastroenterol. 2014, 20, 9427–9438. [Google Scholar] [CrossRef]
- Azziz, R.; Carmina, E.; Chen, Z.; Dunaif, A.; Laven, J.S.E.; Legro, R.S.; Lizneva, D.; Natterson-Horowtiz, B.; Teede, H.J.; Yildiz, B.O. Polycystic ovary syndrome. Nat. Rev. Dis. Primers 2016, 2, 16057. [Google Scholar] [CrossRef]
- Hutchison, A.L.; Tavaglione, F.; Romeo, S.; Charlton, M. Endocrine aspects of metabolic dysfunction-associated steatotic liver disease (MASLD): Beyond insulin resistance. J. Hepatol. 2023, 79, 1524–1541. [Google Scholar] [CrossRef]
- Liu, D.; Gao, X.; Pan, X.-F.; Zhou, T.; Zhu, C.; Li, F.; Fan, J.-G.; Targher, G.; Zhao, J. The hepato-ovarian axis: Genetic evidence for a causal association between non-alcoholic fatty liver disease and polycystic ovary syndrome. BMC Med. 2023, 21, 62. [Google Scholar] [CrossRef]
- Mullur, R.; Liu, Y.-Y.; Brent, G.A. Thyroid Hormone Regulation of Metabolism. Physiol. Rev. 2014, 94, 355–382. [Google Scholar] [CrossRef]
- Zhang, J.-Z.; Cai, J.-J.; Yu, Y.; She, Z.-G.; Li, H. Nonalcoholic Fatty Liver Disease: An Update on the Diagnosis. Gene Expr. 2019, 19, 187–198. [Google Scholar] [CrossRef]
- Josloff, K.; Beiriger, J.; Khan, A.; Gawel, R.J.; Kirby, R.S.; Kendrick, A.D.; Rao, A.K.; Wang, R.X.; Schafer, M.M.; Pearce, M.E.; et al. Comprehensive Review of Cardiovascular Disease Risk in Nonalcoholic Fatty Liver Disease. J. Cardiovasc. Dev. Dis. 2022, 9, 419. [Google Scholar] [CrossRef] [PubMed]
- Pirola, C.J.; Sookoian, S. Personalized medicine in nonalcoholic fatty liver disease. Clin. Mol. Hepatol. 2022, 28, 935–938. [Google Scholar] [CrossRef] [PubMed]
- Rosato, V.; Masarone, M.; Dallio, M.; Federico, A.; Aglitti, A.; Persico, M. NAFLD and Extra-Hepatic Comorbidities: Current Evidence on a Multi-Organ Metabolic Syndrome. Int. J. Environ. Res. Public. Health 2019, 16, 3415. [Google Scholar] [CrossRef]
- Vilar-Gomez, E.; Martinez-Perez, Y.; Calzadilla-Bertot, L.; Torres-Gonzalez, A.; Gra-Oramas, B.; Gonzalez-Fabian, L.; Friedman, S.L.; Diago, M.; Romero-Gomez, M. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology 2015, 149, 367–378.e5; quiz e14–e15. [Google Scholar] [CrossRef]
- European Association for the Study of the Liver (EASL); European Association for the Study of Diabetes (EASD); European Association for the Study of Obesity (EASO) EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J. Hepatol. 2016, 64, 1388–1402. [CrossRef]
- Zelber-Sagi, S.; Salomone, F.; Mlynarsky, L. The Mediterranean dietary pattern as the diet of choice for non-alcoholic fatty liver disease: Evidence and plausible mechanisms. Liver Int. Off. J. Int. Assoc. Study Liver 2017, 37, 936–949. [Google Scholar] [CrossRef]
- Montemayor, S.; Mascaró, C.M.; Ugarriza, L.; Casares, M.; Llompart, I.; Abete, I.; Zulet, M.Á.; Martínez, J.A.; Tur, J.A.; Bouzas, C. Adherence to Mediterranean Diet and NAFLD in Patients with Metabolic Syndrome: The FLIPAN Study. Nutrients 2022, 14, 3186. [Google Scholar] [CrossRef]
- Plaz Torres, M.C.; Aghemo, A.; Lleo, A.; Bodini, G.; Furnari, M.; Marabotto, E.; Miele, L.; Giannini, E.G. Mediterranean Diet and NAFLD: What We Know and Questions That Still Need to Be Answered. Nutrients 2019, 11, 2971. [Google Scholar] [CrossRef]
- Anton, S.D.; Moehl, K.; Donahoo, W.T.; Marosi, K.; Lee, S.; Mainous, A.G.; Leeuwenburgh, C.; Mattson, M.P. Flipping the Metabolic Switch: Understanding and Applying Health Benefits of Fasting. Obes. Silver Spring Md. 2018, 26, 254–268. [Google Scholar] [CrossRef]
- Diab, R.; Dimachkie, L.; Zein, O.; Dakroub, A.; Eid, A.H. Intermittent Fasting Regulates Metabolic Homeostasis and Improves Cardiovascular Health. Cell Biochem. Biophys. 2024, 82, 1583–1597. [Google Scholar] [CrossRef]
- von Loeffelholz, C.; Roth, J.; Coldewey, S.M.; Birkenfeld, A.L. The Role of Physical Activity in Nonalcoholic and Metabolic Dysfunction Associated Fatty Liver Disease. Biomedicines 2021, 9, 1853. [Google Scholar] [CrossRef] [PubMed]
- Mika, A.; Macaluso, F.; Barone, R.; Di Felice, V.; Sledzinski, T. Effect of Exercise on Fatty Acid Metabolism and Adipokine Secretion in Adipose Tissue. Front. Physiol. 2019, 10, 26. [Google Scholar] [CrossRef]
- Gleeson, M.; Bishop, N.C.; Stensel, D.J.; Lindley, M.R.; Mastana, S.S.; Nimmo, M.A. The anti-inflammatory effects of exercise: Mechanisms and implications for the prevention and treatment of disease. Nat. Rev. Immunol. 2011, 11, 607–615. [Google Scholar] [CrossRef] [PubMed]
- Hickman, I.J.; Jonsson, J.R.; Prins, J.B.; Ash, S.; Purdie, D.M.; Clouston, A.D.; Powell, E.E. Modest weight loss and physical activity in overweight patients with chronic liver disease results in sustained improvements in alanine aminotransferase, fasting insulin, and quality of life. Gut 2004, 53, 413–419. [Google Scholar] [CrossRef]
- Kirwan, J.P.; Sacks, J.; Nieuwoudt, S. The essential role of exercise in the management of type 2 diabetes. Cleve. Clin. J. Med. 2017, 84, S15–S21. [Google Scholar] [CrossRef]
- Van Der Heijden, G.-J.; Wang, Z.J.; Chu, Z.; Toffolo, G.; Manesso, E.; Sauer, P.J.J.; Sunehag, A.L. Strength exercise improves muscle mass and hepatic insulin sensitivity in obese youth. Med. Sci. Sports Exerc. 2010, 42, 1973–1980. [Google Scholar] [CrossRef]
- Eckard, C.; Cole, R.; Lockwood, J.; Torres, D.M.; Williams, C.D.; Shaw, J.C.; Harrison, S.A. Prospective histopathologic evaluation of lifestyle modification in nonalcoholic fatty liver disease: A randomized trial. Ther. Adv. Gastroenterol. 2013, 6, 249–259. [Google Scholar] [CrossRef]
- Mazza, A.; Fruci, B.; Garinis, G.A.; Giuliano, S.; Malaguarnera, R.; Belfiore, A. The role of metformin in the management of NAFLD. Exp. Diabetes Res. 2012, 2012, 716404. [Google Scholar] [CrossRef]
- Huang, Y.; Wang, X.; Yan, C.; Li, C.; Zhang, L.; Zhang, L.; Liang, E.; Liu, T.; Mao, J. Effect of metformin on nonalcoholic fatty liver based on meta-analysis and network pharmacology. Medicine 2022, 101, e31437. [Google Scholar] [CrossRef]
- Haukeland, J.W.; Konopski, Z.; Eggesbø, H.B.; von Volkmann, H.L.; Raschpichler, G.; Bjøro, K.; Haaland, T.; Løberg, E.M.; Birkeland, K. Metformin in patients with non-alcoholic fatty liver disease: A randomized, controlled trial. Scand. J. Gastroenterol. 2009, 44, 853–860. [Google Scholar] [CrossRef]
- Bugianesi, E.; McCullough, A.J.; Marchesini, G. Insulin resistance: A metabolic pathway to chronic liver disease. Hepatology 2005, 42, 987–1000. [Google Scholar] [CrossRef] [PubMed]
- Horn, C.L.; Morales, A.L.; Savard, C.; Farrell, G.C.; Ioannou, G.N. Role of Cholesterol-Associated Steatohepatitis in the Development of NASH. Hepatol. Commun. 2022, 6, 12–35. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Chi, X.; Wang, Y.; Setrerrahmane, S.; Xie, W.; Xu, H. Trends in insulin resistance: Insights into mechanisms and therapeutic strategy. Signal Transduct. Target. Ther. 2022, 7, 1–25. [Google Scholar] [CrossRef] [PubMed]
- Feng, W.; Gao, C.; Bi, Y.; Wu, M.; Li, P.; Shen, S.; Chen, W.; Yin, T.; Zhu, D. Randomized trial comparing the effects of gliclazide, liraglutide, and metformin on diabetes with non-alcoholic fatty liver disease. J. Diabetes 2017, 9, 800–809. [Google Scholar] [CrossRef]
- Yan, J.; Yao, B.; Kuang, H.; Yang, X.; Huang, Q.; Hong, T.; Li, Y.; Dou, J.; Yang, W.; Qin, G.; et al. Liraglutide, Sitagliptin, and Insulin Glargine Added to Metformin: The Effect on Body Weight and Intrahepatic Lipid in Patients With Type 2 Diabetes Mellitus and Nonalcoholic Fatty Liver Disease. Hepatology 2019, 69, 2414–2426. [Google Scholar] [CrossRef]
- Koureta, E.; Cholongitas, E. Combination therapies in nonalcoholic fatty liver disease using antidiabetic and disease-specific drugs. Ann. Gastroenterol. 2023, 36, 378–391. [Google Scholar] [CrossRef]
- Kersten, S. Integrated physiology and systems biology of PPARα. Mol. Metab. 2014, 3, 354–371. [Google Scholar] [CrossRef]
- Molecular Actions of PPARα in Lipid Metabolism and Inflammation|Endocrine Reviews|Oxford Academic. Available online: https://academic.oup.com/edrv/article/39/5/760/5055100 (accessed on 7 February 2025).
- Nakagawa, Y.; Satoh, A.; Tezuka, H.; Han, S.; Takei, K.; Iwasaki, H.; Yatoh, S.; Yahagi, N.; Suzuki, H.; Iwasaki, Y.; et al. CREB3L3 controls fatty acid oxidation and ketogenesis in synergy with PPARα. Sci. Rep. 2016, 6, 39182. [Google Scholar] [CrossRef]
- Orasanu, G.; Ziouzenkova, O.; Devchand, P.R.; Nehra, V.; Hamdy, O.; Horton, E.S.; Plutzky, J. The PPARγ Agonist Pioglitazone Represses Inflammation In A PPARα-Dependent Manner In Vitro and In Vivo In Mice. J. Am. Coll. Cardiol. 2008, 52, 869–881. [Google Scholar] [CrossRef]
- Ito, D.; Shimizu, S.; Inoue, K.; Saito, D.; Yanagisawa, M.; Inukai, K.; Akiyama, Y.; Morimoto, Y.; Noda, M.; Shimada, A. Comparison of Ipragliflozin and Pioglitazone Effects on Nonalcoholic Fatty Liver Disease in Patients With Type 2 Diabetes: A Randomized, 24-Week, Open-Label, Active-Controlled Trial. Diabetes Care 2017, 40, 1364–1372. [Google Scholar] [CrossRef]
- Nagashima, K.; Lopez, C.; Donovan, D.; Ngai, C.; Fontanez, N.; Bensadoun, A.; Fruchart-Najib, J.; Holleran, S.; Cohn, J.S.; Ramakrishnan, R.; et al. Effects of the PPARγ agonist pioglitazone on lipoprotein metabolism in patients with type 2 diabetes mellitus. J. Clin. Investig. 2005, 115, 1323–1332. [Google Scholar] [CrossRef] [PubMed]
- Sanyal, A.J.; Chalasani, N.; Kowdley, K.V.; McCullough, A.; Diehl, A.M.; Bass, N.M.; Neuschwander-Tetri, B.A.; Lavine, J.E.; Tonascia, J.; Unalp, A.; et al. Pioglitazone, Vitamin E, or Placebo for Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2010, 362, 1675–1685. [Google Scholar] [CrossRef] [PubMed]
- Aithal, G.P.; Thomas, J.A.; Kaye, P.V.; Lawson, A.; Ryder, S.D.; Spendlove, I.; Austin, A.S.; Freeman, J.G.; Morgan, L.; Webber, J. Randomized, Placebo-Controlled Trial of Pioglitazone in Nondiabetic Subjects With Nonalcoholic Steatohepatitis. Gastroenterology 2008, 135, 1176–1184. [Google Scholar] [CrossRef] [PubMed]
- Belfort, R.; Harrison, S.A.; Brown, K.; Darland, C.; Finch, J.; Hardies, J.; Balas, B.; Gastaldelli, A.; Tio, F.; Pulcini, J.; et al. A Placebo-Controlled Trial of Pioglitazone in Subjects with Nonalcoholic Steatohepatitis. N. Engl. J. Med. 2006, 355, 2297–2307. [Google Scholar] [CrossRef]
- Mohanty, P.; Aljada, A.; Ghanim, H.; Hofmeyer, D.; Tripathy, D.; Syed, T.; Al-Haddad, W.; Dhindsa, S.; Dandona, P. Evidence for a potent antiinflammatory effect of rosiglitazone. J. Clin. Endocrinol. Metab. 2004, 89, 2728–2735. [Google Scholar] [CrossRef]
- Crossno, J.T.; Majka, S.M.; Grazia, T.; Gill, R.G.; Klemm, D.J. Rosiglitazone promotes development of a novel adipocyte population from bone marrow–derived circulating progenitor cells. J. Clin. Investig. 2006, 116, 3220–3228. [Google Scholar] [CrossRef]
- Ratziu, V.; Giral, P.; Jacqueminet, S.; Charlotte, F.; Hartemann-Heurtier, A.; Serfaty, L.; Podevin, P.; Lacorte, J.-M.; Bernhardt, C.; Bruckert, E.; et al. Rosiglitazone for nonalcoholic steatohepatitis: One-year results of the randomized placebo-controlled Fatty Liver Improvement with Rosiglitazone Therapy (FLIRT) Trial. Gastroenterology 2008, 135, 100–110. [Google Scholar] [CrossRef]
- Ratziu, V.; Charlotte, F.; Bernhardt, C.; Giral, P.; Halbron, M.; Lenaour, G.; Hartmann-Heurtier, A.; Bruckert, E.; Poynard, T.; LIDO Study Group. Long-term efficacy of rosiglitazone in nonalcoholic steatohepatitis: Results of the fatty liver improvement by rosiglitazone therapy (FLIRT 2) extension trial. Hepatology 2010, 51, 445–453. [Google Scholar] [CrossRef]
- Nesto, R.W.; Bell, D.; Bonow, R.O.; Fonseca, V.; Grundy, S.M.; Horton, E.S.; Le Winter, M.; Porte, D.; Semenkovich, C.F.; Smith, S.; et al. Thiazolidinedione Use, Fluid Retention, and Congestive Heart Failure. Circulation 2003, 108, 2941–2948. [Google Scholar] [CrossRef]
- Müller, T.D.; Finan, B.; Bloom, S.R.; D’Alessio, D.; Drucker, D.J.; Flatt, P.R.; Fritsche, A.; Gribble, F.; Grill, H.J.; Habener, J.F.; et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 2019, 30, 72–130. [Google Scholar] [CrossRef]
- Bednarz, K.; Kowalczyk, K.; Cwynar, M.; Czapla, D.; Czarkowski, W.; Kmita, D.; Nowak, A.; Madej, P. The Role of Glp-1 Receptor Agonists in Insulin Resistance with Concomitant Obesity Treatment in Polycystic Ovary Syndrome. Int. J. Mol. Sci. 2022, 23, 4334. [Google Scholar] [CrossRef] [PubMed]
- Deacon, C.F. Physiology and Pharmacology of DPP-4 in Glucose Homeostasis and the Treatment of Type 2 Diabetes. Front. Endocrinol. 2019, 10, 80. [Google Scholar] [CrossRef]
- Barchetta, I.; Ceccarelli, V.; Cimini, F.A.; Barone, E.; Sentinelli, F.; Coluzzi, M.; Chiappetta, C.; Bertoccini, L.; Tramutola, A.; Labbadia, G.; et al. Circulating dipeptidyl peptidase-4 is independently associated with the presence and severity of NAFLD/NASH in individuals with and without obesity and metabolic disease. J. Endocrinol. Investig. 2021, 44, 979–988. [Google Scholar] [CrossRef]
- Bae, J.C. DPP-4 Inhibitor in Type 2 Diabetes Mellitus Patient with Non-Alcoholic Fatty Liver Disease: Achieving Two Goals at Once? Endocrinol. Metab. 2022, 37, 858–860. [Google Scholar] [CrossRef]
- Seghieri, M.; Christensen, A.S.; Andersen, A.; Solini, A.; Knop, F.K.; Vilsbøll, T. Future Perspectives on GLP-1 Receptor Agonists and GLP-1/glucagon Receptor Co-agonists in the Treatment of NAFLD. Front. Endocrinol. 2018, 9, 649. [Google Scholar] [CrossRef]
- Hartman, M.L.; Sanyal, A.J.; Loomba, R.; Wilson, J.M.; Nikooienejad, A.; Bray, R.; Karanikas, C.A.; Duffin, K.L.; Robins, D.A.; Haupt, A. Effects of Novel Dual GIP and GLP-1 Receptor Agonist Tirzepatide on Biomarkers of Nonalcoholic Steatohepatitis in Patients With Type 2 Diabetes. Diabetes Care 2020, 43, 1352–1355. [Google Scholar] [CrossRef]
- Loomba, R.; Hartman, M.L.; Lawitz, E.J.; Vuppalanchi, R.; Boursier, J.; Bugianesi, E.; Yoneda, M.; Behling, C.; Cummings, O.W.; Tang, Y.; et al. Tirzepatide for Metabolic Dysfunction-Associated Steatohepatitis with Liver Fibrosis. N. Engl. J. Med. 2024, 391, 299–310. [Google Scholar] [CrossRef]
- Armstrong, M.J.; Gaunt, P.; Aithal, G.P.; Barton, D.; Hull, D.; Parker, R.; Hazlehurst, J.M.; Guo, K.; LEAN Trial Team; Abouda, G.; et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): A multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet Lond. Engl. 2016, 387, 679–690. [Google Scholar] [CrossRef]
- DeFronzo, R.A.; Davidson, J.A.; Del Prato, S. The role of the kidneys in glucose homeostasis: A new path towards normalizing glycaemia. Diabetes Obes. Metab. 2012, 14, 5–14. [Google Scholar] [CrossRef]
- Ghezzi, C.; Loo, D.D.F.; Wright, E.M. Physiology of renal glucose handling via SGLT1, SGLT2 and GLUT2. Diabetologia 2018, 61, 2087–2097. [Google Scholar] [CrossRef]
- Tahrani, A.A.; Barnett, A.H.; Bailey, C.J. SGLT inhibitors in management of diabetes. Lancet Diabetes Endocrinol. 2013, 1, 140–151. [Google Scholar] [CrossRef] [PubMed]
- Tang, J.; Ye, L.; Yan, Q.; Zhang, X.; Wang, L. Effects of Sodium-Glucose Cotransporter 2 Inhibitors on Water and Sodium Metabolism. Front. Pharmacol. 2022, 13, 800490. [Google Scholar] [CrossRef]
- Renal Sodium-Glucose Cotransporter Inhibition in the Management of Type 2 Diabetes Mellitus—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/26354881/ (accessed on 7 February 2025).
- Pradhan, R.; Yin, H.; Yu, O.; Azoulay, L. Glucagon-Like Peptide 1 Receptor Agonists and Sodium-Glucose Cotransporter 2 Inhibitors and Risk of Nonalcoholic Fatty Liver Disease Among Patients with Type 2 Diabetes. Diabetes Care 2022, 45, 819–829. [Google Scholar] [CrossRef] [PubMed]
- Ding, C.; Tang, Y.; Zhu, W.; Huang, P.; Lian, P.; Ran, J.; Huang, X. Sodium-glucose cotransporter protein-2 inhibitors and glucagon-like peptide-1 receptor agonists versus thiazolidinediones for non-alcoholic fatty liver disease: A network meta-analysis. Acta Diabetol. 2022, 59, 519–533. [Google Scholar] [CrossRef]
- Hamaguchi, E.; Takamura, T.; Sakurai, M.; Mizukoshi, E.; Zen, Y.; Takeshita, Y.; Kurita, S.; Arai, K.; Yamashita, T.; Sasaki, M.; et al. Histological Course of Nonalcoholic Fatty Liver Disease in Japanese Patients. Diabetes Care 2010, 33, 284–286. [Google Scholar] [CrossRef]
- Garvey, W.T.; Van Gaal, L.; Leiter, L.A.; Vijapurkar, U.; List, J.; Cuddihy, R.; Ren, J.; Davies, M.J. Effects of canagliflozin versus glimepiride on adipokines and inflammatory biomarkers in type 2 diabetes. Metabolism 2018, 85, 32–37. [Google Scholar] [CrossRef]
- Kuchay, M.S.; Krishan, S.; Mishra, S.K.; Farooqui, K.J.; Singh, M.K.; Wasir, J.S.; Bansal, B.; Kaur, P.; Jevalikar, G.; Gill, H.K.; et al. Effect of Empagliflozin on Liver Fat in Patients With Type 2 Diabetes and Nonalcoholic Fatty Liver Disease: A Randomized Controlled Trial (E-LIFT Trial). Diabetes Care 2018, 41, 1801–1808. [Google Scholar] [CrossRef]
- Kahl, S.; Gancheva, S.; Straßburger, K.; Herder, C.; Machann, J.; Katsuyama, H.; Kabisch, S.; Henkel, E.; Kopf, S.; Lagerpusch, M.; et al. Empagliflozin Effectively Lowers Liver Fat Content in Well-Controlled Type 2 Diabetes: A Randomized, Double-Blind, Phase 4, Placebo-Controlled Trial. Diabetes Care 2020, 43, 298–305. [Google Scholar] [CrossRef]
- Shimizu, M.; Suzuki, K.; Kato, K.; Jojima, T.; Iijima, T.; Murohisa, T.; Iijima, M.; Takekawa, H.; Usui, I.; Hiraishi, H.; et al. Evaluation of the effects of dapagliflozin, a sodium-glucose co-transporter-2 inhibitor, on hepatic steatosis and fibrosis using transient elastography in patients with type 2 diabetes and non-alcoholic fatty liver disease. Diabetes Obes. Metab. 2019, 21, 285–292. [Google Scholar] [CrossRef]
- Leiter, L.A.; Forst, T.; Polidori, D.; Balis, D.A.; Xie, J.; Sha, S. Effect of canagliflozin on liver function tests in patients with type 2 diabetes. Diabetes Metab. 2016, 42, 25–32. [Google Scholar] [CrossRef]
- Lefer, A.M.; Scalia, R.; Lefer, D.J. Vascular effects of HMG CoA-reductase inhibitors (statins) unrelated to cholesterol lowering: New concepts for cardiovascular disease. Cardiovasc. Res. 2001, 49, 281–287. [Google Scholar] [CrossRef] [PubMed]
- Ali, A.H.; Younis, N.; Abdallah, R.; Shaer, F.; Dakroub, A.; Ayoub, M.A.; Iratni, R.; Yassine, H.M.; Zibara, K.; Orekhov, A.; et al. Lipid-Lowering Therapies for Atherosclerosis: Statins, Fibrates, Ezetimibe and PCSK9 Monoclonal Antibodies. Curr. Med. Chem. 2021, 28, 7427–7445. [Google Scholar] [CrossRef] [PubMed]
- Stein, E.A.; Lane, M.; Laskarzewski, P. Comparison of statins in hypertriglyceridemia. Am. J. Cardiol. 1998, 81, 66B–69B. [Google Scholar] [CrossRef]
- Karlson, B.W.; Palmer, M.K.; Nicholls, S.J.; Lundman, P.; Barter, P.J. A VOYAGER Meta-Analysis of the Impact of Statin Therapy on Low-Density Lipoprotein Cholesterol and Triglyceride Levels in Patients With Hypertriglyceridemia. Am. J. Cardiol. 2016, 117, 1444–1448. [Google Scholar] [CrossRef]
- Yamashita, S.; Tsubakio-Yamamoto, K.; Ohama, T.; Nakagawa-Toyama, Y.; Nishida, M. Molecular mechanisms of HDL-cholesterol elevation by statins and its effects on HDL functions. J. Atheroscler. Thromb. 2010, 17, 436–451. [Google Scholar] [CrossRef]
- Blum, A.; Shamburek, R. The pleiotropic effects of statins on endothelial function, vascular inflammation, immunomodulation and thrombogenesis. Atherosclerosis 2009, 203, 325–330. [Google Scholar] [CrossRef]
- Blanco-Colio, L.M.; Tuñón, J.; Martín-Ventura, J.L.; Egido, J. Anti-inflammatory and immunomodulatory effects of statins. Kidney Int. 2003, 63, 12–23. [Google Scholar] [CrossRef]
- Statin Therapy and Myocardial No-Reflow—Calvert—2006—British JOURNAL of Pharmacology—Wiley Online Library. Available online: https://bpspubs.onlinelibrary.wiley.com/doi/10.1038/sj.bjp.0706863 (accessed on 5 February 2025).
- Horwich, T.B.; MacLellan, W.R.; Fonarow, G.C. Statin therapy is associated with improved survival in ischemic and non-ischemic heart failure. J. Am. Coll. Cardiol. 2004, 43, 642–648. [Google Scholar] [CrossRef]
- Athyros, V.G.; Boutari, C.; Stavropoulos, K.; Anagnostis, P.; Imprialos, K.P.; Doumas, M.; Karagiannis, A. Statins: An Under-Appreciated Asset for the Prevention and the Treatment of NAFLD or NASH and the Related Cardiovascular Risk. Curr. Vasc. Pharmacol. 2018, 16, 246–253. [Google Scholar] [CrossRef]
- Lee, J.I.; Lee, H.W.; Lee, K.S.; Lee, H.S.; Park, J.-Y. Effects of Statin Use on the Development and Progression of Nonalcoholic Fatty Liver Disease: A Nationwide Nested Case-Control Study. Am. J. Gastroenterol. 2021, 116, 116–124. [Google Scholar] [CrossRef]
- Targher, G.; Byrne, C.D.; Tilg, H. NAFLD and increased risk of cardiovascular disease: Clinical associations, pathophysiological mechanisms and pharmacological implications. Gut 2020, 69, 1691–1705. [Google Scholar] [CrossRef] [PubMed]
- Kaplan, D.E.; Serper, M.A.; Mehta, R.; Fox, R.; John, B.; Aytaman, A.; Baytarian, M.; Hunt, K.; Albrecht, J.; Njei, B.; et al. Effects of Hypercholesterolemia and Statin Exposure on Survival in a Large National Cohort of Patients With Cirrhosis. Gastroenterology 2019, 156, 1693–1706.e12. [Google Scholar] [CrossRef] [PubMed]
- Doumas, M.; Imprialos, K.; Dimakopoulou, A.; Stavropoulos, K.; Binas, A.; Athyros, V.G. The Role of Statins in the Management of Nonalcoholic Fatty Liver Disease. Curr. Pharm. Des. 2018, 24, 4587–4592. [Google Scholar] [CrossRef] [PubMed]
- van den Berg, E.H.; Wolters, A.A.B.; Dullaart, R.P.F.; Moshage, H.; Zurakowski, D.; de Meijer, V.E.; Blokzijl, H. Prescription of statins in suspected non-alcoholic fatty liver disease and high cardiovascular risk, a population-based study. Liver Int. Off. J. Int. Assoc. Study Liver 2019, 39, 1343–1354. [Google Scholar] [CrossRef]
- Pastori, D.; Polimeni, L.; Baratta, F.; Pani, A.; Del Ben, M.; Angelico, F. The efficacy and safety of statins for the treatment of non-alcoholic fatty liver disease. Dig. Liver Dis. Off. J. Ital. Soc. Gastroenterol. Ital. Assoc. Study Liver 2015, 47, 4–11. [Google Scholar] [CrossRef]
- Abdallah, M.; Brown, L.; Provenza, J.; Tariq, R.; Gowda, S.; Singal, A.K. Safety and efficacy of dyslipidemia treatment in NAFLD patients: A meta-analysis of randomized controlled trials. Ann. Hepatol. 2022, 27, 100738. [Google Scholar] [CrossRef]
- Golomb, B.A.; Evans, M.A. Statin adverse effects: A review of the literature and evidence for a mitochondrial mechanism. Am. J. Cardiovasc. Drugs Drugs Devices Interv. 2008, 8, 373–418. [Google Scholar] [CrossRef]
- Simon, T.G.; Bonilla, H.; Yan, P.; Chung, R.T.; Butt, A.A. Atorvastatin and fluvastatin are associated with dose-dependent reductions in cirrhosis and hepatocellular carcinoma, among patients with hepatitis C virus: Results from ERCHIVES. Hepatology 2016, 64, 47–57. [Google Scholar] [CrossRef]
- Zou, B.; Odden, M.C.; Nguyen, M.H. Statin Use and Reduced Hepatocellular Carcinoma Risk in Patients with Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2023, 21, 435–444.e6. [Google Scholar] [CrossRef]
- Kim, G.; Jang, S.-Y.; Nam, C.M.; Kang, E.S. Statin use and the risk of hepatocellular carcinoma in patients at high risk: A nationwide nested case-control study. J. Hepatol. 2018, 68, 476–484. [Google Scholar] [CrossRef]
- Sebastiani, G.; Patel, K.; Ratziu, V.; Feld, J.J.; Neuschwander-Tetri, B.A.; Pinzani, M.; Petta, S.; Berzigotti, A.; Metrakos, P.; Shoukry, N.; et al. Current considerations for clinical management and care of non-alcoholic fatty liver disease: Insights from the 1st International Workshop of the Canadian NASH Network (CanNASH). Can. Liver J. 2022, 5, 61–90. [Google Scholar] [CrossRef] [PubMed]
- Mancini, G.B.J.; Baker, S.; Bergeron, J.; Fitchett, D.; Frohlich, J.; Genest, J.; Gupta, M.; Hegele, R.A.; Ng, D.; Pearson, G.J.; et al. Diagnosis, Prevention, and Management of Statin Adverse Effects and Intolerance: Canadian Consensus Working Group Update (2016). Can. J. Cardiol. 2016, 32, S35–S65. [Google Scholar] [CrossRef] [PubMed]
- Arnaud, C.; Burger, F.; Steffens, S.; Veillard, N.R.; Nguyen, T.H.; Trono, D.; Mach, F. Statins reduce interleukin-6-induced C-reactive protein in human hepatocytes: New evidence for direct antiinflammatory effects of statins. Arterioscler. Thromb. Vasc. Biol. 2005, 25, 1231–1236. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Ren, X.; Zhang, B.; Lan, T.; Liu, B. A Systematic Review of Statins for the Treatment of Nonalcoholic Steatohepatitis: Safety, Efficacy, and Mechanism of Action. Molecules 2024, 29, 1859. [Google Scholar] [CrossRef]
- Chen, D.; Sui, L.; Chen, C.; Liu, S.; Sun, X.; Guan, J. Atorvastatin suppresses NLRP3 inflammasome activation in intracerebral hemorrhage via TLR4- and MyD88-dependent pathways. Aging 2022, 14, 462–476. [Google Scholar] [CrossRef]
- Vargas, J.I.; Arrese, M.; Shah, V.H.; Arab, J.P. Use of Statins in Patients with Chronic Liver Disease and Cirrhosis: Current Views and Prospects. Curr. Gastroenterol. Rep. 2017, 19, 43. [Google Scholar] [CrossRef]
- Islam, M.M.; Poly, T.N.; Walther, B.A.; Yang, H.-C.; Jack Li, Y.-C. Statin Use and the Risk of Hepatocellular Carcinoma: A Meta-Analysis of Observational Studies. Cancers 2020, 12, 671. [Google Scholar] [CrossRef]
- McConnell, M.J.; Kostallari, E.; Ibrahim, S.H.; Iwakiri, Y. The evolving role of liver sinusoidal endothelial cells in liver health and disease. Hepatology 2023, 78, 649–669. [Google Scholar] [CrossRef]
- Fujita, T.; Narumiya, S. Roles of hepatic stellate cells in liver inflammation: A new perspective. Inflamm. Regen. 2016, 36, 1. [Google Scholar] [CrossRef]
- Torres-Peña, J.D.; Martín-Piedra, L.; Fuentes-Jiménez, F. Statins in Non-alcoholic Steatohepatitis. Front. Cardiovasc. Med. 2021, 8, 777131. [Google Scholar] [CrossRef]
- da Silva Pereira, E.N.G.; Franco, R.L.C.; Santos, R.D.C.D.; Daliry, A. Statins and non-alcoholic fatty liver disease: A concise review. Biomed. Pharmacother. 2025, 183, 117805. [Google Scholar] [CrossRef]
- Yano, M.; Matsumura, T.; Senokuchi, T.; Ishii, N.; Murata, Y.; Taketa, K.; Motoshima, H.; Taguchi, T.; Sonoda, K.; Kukidome, D.; et al. Statins activate peroxisome proliferator-activated receptor gamma through extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase-dependent cyclooxygenase-2 expression in macrophages. Circ. Res. 2007, 100, 1442–1451. [Google Scholar] [CrossRef] [PubMed]
- Vieira-Silva, S.; Falony, G.; Belda, E.; Nielsen, T.; Aron-Wisnewsky, J.; Chakaroun, R.; Forslund, S.K.; Assmann, K.; Valles-Colomer, M.; Nguyen, T.T.D.; et al. Statin therapy is associated with lower prevalence of gut microbiota dysbiosis. Nature 2020, 581, 310–315. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Xu, H.; Zhou, X.; Chen, W.; Zhou, H. Dysregulated bile acid homeostasis: Unveiling its role in metabolic diseases. Med. Rev. 2024, 4, 262–283. [Google Scholar] [CrossRef]
- Xiang, D.; Yang, J.; Liu, L.; Yu, H.; Gong, X.; Liu, D. The regulation of tissue-specific farnesoid X receptor on genes and diseases involved in bile acid homeostasis. Biomed. Pharmacother. 2023, 168, 115606. [Google Scholar] [CrossRef]
- Farnesoid X Receptor Activation by Bile Acids Suppresses Lipid Peroxidation and Ferroptosis|Nature Communications. Available online: https://www.nature.com/articles/s41467-023-42702-8 (accessed on 7 February 2025).
- Han, S.Y.; Song, H.K.; Cha, J.J.; Han, J.Y.; Kang, Y.S.; Cha, D.R. Farnesoid X receptor (FXR) agonist ameliorates systemic insulin resistance, dysregulation of lipid metabolism, and alterations of various organs in a type 2 diabetic kidney animal model. Acta Diabetol. 2021, 58, 495–503. [Google Scholar] [CrossRef]
- Clifford, B.L.; Sedgeman, L.R.; Williams, K.J.; Morand, P.; Cheng, A.; Jarrett, K.E.; Chan, A.P.; Brearley-Sholto, M.C.; Wahlström, A.; Ashby, J.W.; et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab. 2021, 33, 1671–1684.e4. [Google Scholar] [CrossRef]
- Pelton, P.D.; Patel, M.; Demarest, K.T. Nuclear receptors as potential targets for modulating reverse cholesterol transport. Curr. Top. Med. Chem. 2005, 5, 265–282. [Google Scholar] [CrossRef]
- Goodwin, B.; Jones, S.A.; Price, R.R.; Watson, M.A.; McKee, D.D.; Moore, L.B.; Galardi, C.; Wilson, J.G.; Lewis, M.C.; Roth, M.E.; et al. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol. Cell 2000, 6, 517–526. [Google Scholar] [CrossRef]
- Aguilar-Olivos, N.E.; Carrillo-Córdova, D.; Oria-Hernández, J.; Sánchez-Valle, V.; Ponciano-Rodríguez, G.; Ramírez-Jaramillo, M.; Chablé-Montero, F.; Chávez-Tapia, N.C.; Uribe, M.; Méndez-Sánchez, N. The nuclear receptor FXR, but not LXR, up-regulates bile acid transporter expression in non-alcoholic fatty liver disease. Ann. Hepatol. 2015, 14, 487–493. [Google Scholar]
- Malagnino, V.; Hussner, J.; Issa, A.; Midzic, A.; Meyer Zu Schwabedissen, H.E. OATP1B3-1B7, a novel organic anion transporting polypeptide, is modulated by FXR ligands and transports bile acids. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 317, G751–G762. [Google Scholar] [CrossRef] [PubMed]
- Ren, T.; Pang, L.; Dai, W.; Wu, S.; Kong, J. Regulatory mechanisms of the bile salt export pump (BSEP/ABCB11) and its role in related diseases. Clin. Res. Hepatol. Gastroenterol. 2021, 45, 101641. [Google Scholar] [CrossRef] [PubMed]
- Tang, Y.; Fan, Y.; Wang, Y.; Wang, D.; Huang, Q.; Chen, T.; Cao, X.; Wen, C.; Shen, X.; Li, J.; et al. A Current Understanding of FXR in NAFLD: The multifaceted regulatory role of FXR and novel lead discovery for drug development. Biomed. Pharmacother. 2024, 175, 116658. [Google Scholar] [CrossRef]
- Wei, M.; Cao, W.-B.; Zhao, R.-D.; Sun, D.-P.; Liang, Y.-Z.; Huang, Y.-D.; Cheng, Z.-W.; Ouyang, J.; Yang, W.-S.; Yu, W.-B. Fibroblast growth factor 15, induced by elevated bile acids, mediates the improvement of hepatic glucose metabolism after sleeve gastrectomy. World J. Gastroenterol. 2023, 29, 3280–3291. [Google Scholar] [CrossRef]
- Kliewer, S.A.; Mangelsdorf, D.J. Bile Acids as Hormones: The FXR-FGF15/19 Pathway. Dig. Dis. 2015, 33, 327–331. [Google Scholar] [CrossRef]
- Song, L.; Wang, L.; Hou, Y.; Zhou, J.; Chen, C.; Ye, X.; Dong, W.; Gao, H.; Liu, Y.; Qiao, G.; et al. FGF4 protects the liver from nonalcoholic fatty liver disease by activating the AMP-activated protein kinase-Caspase 6 signal axis. Hepatology 2022, 76, 1105–1120. [Google Scholar] [CrossRef]
- Song, L.; Hou, Y.; Xu, D.; Dai, X.; Luo, J.; Liu, Y.; Huang, Z.; Yang, M.; Chen, J.; Hu, Y.; et al. Hepatic FXR-FGF4 is required for bile acid homeostasis via an FGFR4-LRH-1 signal node under cholestatic stress. Cell Metab. 2025, 37, 104–120.e9. [Google Scholar] [CrossRef]
- Meng, D.; Zhang, F.; Yu, W.; Zhang, X.; Yin, G.; Liang, P.; Feng, Y.; Chen, S.; Liu, H. Biological Role and Related Natural Products of SIRT1 in Nonalcoholic Fatty Liver. Diabetes Metab. Syndr. Obes. 2023, 16, 4043–4064. [Google Scholar] [CrossRef]
- Schramm, C.; Wedemeyer, H.; Mason, A.; Hirschfield, G.M.; Levy, C.; Kowdley, K.V.; Milkiewicz, P.; Janczewska, E.; Malova, E.S.; Sanni, J.; et al. Farnesoid X receptor agonist tropifexor attenuates cholestasis in a randomised trial in patients with primary biliary cholangitis. JHEP Rep. Innov. Hepatol. 2022, 4, 100544. [Google Scholar] [CrossRef]
- Ratziu, V.; Harrison, S.A.; Loustaud-Ratti, V.; Bureau, C.; Lawitz, E.; Abdelmalek, M.; Alkhouri, N.; Francque, S.; Girma, H.; Darteil, R.; et al. Hepatic and renal improvements with FXR agonist vonafexor in individuals with suspected fibrotic NASH. J. Hepatol. 2023, 78, 479–492. [Google Scholar] [CrossRef]
- Xu, J.; Wang, Y.; Khoshdeli, M.; Peach, M.; Chuang, J.-C.; Lin, J.; Tsai, W.-W.; Mahadevan, S.; Minto, W.; Diehl, L.; et al. IL-31 levels correlate with pruritus in patients with cholestatic and metabolic liver diseases and is farnesoid X receptor responsive in NASH. Hepatology 2023, 77, 20–32. [Google Scholar] [CrossRef] [PubMed]
- Abenavoli, L.; Falalyeyeva, T.; Boccuto, L.; Tsyryuk, O.; Kobyliak, N. Obeticholic Acid: A New Era in the Treatment of Nonalcoholic Fatty Liver Disease. Pharmacy 2018, 11, 104. [Google Scholar] [CrossRef]
- Makri, E.; Cholongitas, E.; Tziomalos, K. Emerging role of obeticholic acid in the management of nonalcoholic fatty liver disease. World J. Gastroenterol. 2016, 22, 9039–9043. [Google Scholar] [CrossRef] [PubMed]
- Neuschwander-Tetri, B.A.; Loomba, R.; Sanyal, A.J.; Lavine, J.E.; Natta, M.L.V.; Abdelmalek, M.F.; Chalasani, N.; Dasarathy, S.; Diehl, A.M.; Hameed, B.; et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): A multicentre, randomised, placebo-controlled trial. Lancet 2015, 385, 956–965. [Google Scholar] [CrossRef]
- Younossi, Z.M.; Ratziu, V.; Loomba, R.; Rinella, M.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; Geier, A.; Beckebaum, S.; Newsome, P.N.; et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: Interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet Lond. Engl. 2019, 394, 2184–2196. [Google Scholar] [CrossRef]
- Nathani, R.R.; Bansal, M.B. Update on Clinical Trials for Nonalcoholic Steatohepatitis. Gastroenterol. Hepatol. 2023, 19, 371–381. [Google Scholar]
- Jiang, J.; Ma, Y.; Liu, Y.; Lu, D.; Gao, X.; Krausz, K.W.; Desai, D.; Amin, S.G.; Patterson, A.D.; Gonzalez, F.J.; et al. Glycine-β-muricholic acid antagonizes the intestinal farnesoid X receptor-ceramide axis and ameliorates NASH in mice. Hepatol. Commun. 2022, 6, 3363–3378. [Google Scholar] [CrossRef]
- Fleishman, J.S.; Kumar, S. Bile acid metabolism and signaling in health and disease: Molecular mechanisms and therapeutic targets. Signal Transduct. Target. Ther. 2024, 9, 97. [Google Scholar] [CrossRef]
- Bezerra, R.M.N.; Veiga, L.F.; Caetano, A.C.; Rosalen, P.L.; Amaral, M.E.C.; Palanch, A.C.; de Alencar, S.M. Caffeic acid phenethyl ester reduces the activation of the nuclear factor κB pathway by high-fat diet-induced obesity in mice. Metabolism 2012, 61, 1606–1614. [Google Scholar] [CrossRef]
- Shen, C.; Pan, Z.; Wu, S.; Zheng, M.; Zhong, C.; Xin, X.; Lan, S.; Zhu, Z.; Liu, M.; Wu, H.; et al. Emodin palliates high-fat diet-induced nonalcoholic fatty liver disease in mice via activating the farnesoid X receptor pathway. J. Ethnopharmacol. 2021, 279, 114340. [Google Scholar] [CrossRef]
- Lei, Y.; Tang, L.; Chen, Q.; Wu, L.; He, W.; Tu, D.; Wang, S.; Chen, Y.; Liu, S.; Xie, Z.; et al. Disulfiram ameliorates nonalcoholic steatohepatitis by modulating the gut microbiota and bile acid metabolism. Nat. Commun. 2022, 13, 6862. [Google Scholar] [CrossRef] [PubMed]
- Renga, B.; Mencarelli, A.; Migliorati, M.; Distrutti, E.; Fiorucci, S. Bile-acid-activated farnesoid X receptor regulates hydrogen sulfide production and hepatic microcirculation. World J. Gastroenterol. WJG 2009, 15, 2097–2108. [Google Scholar] [CrossRef] [PubMed]
- Nian, F.; Wu, L.; Xia, Q.; Tian, P.; Ding, C.; Lu, X. Akkermansia muciniphila and Bifidobacterium bifidum Prevent NAFLD by Regulating FXR Expression and Gut Microbiota. J. Clin. Transl. Hepatol. 2023, 11, 763–776. [Google Scholar] [CrossRef] [PubMed]
- Paglialunga, S.; Dehn, C.A. Clinical assessment of hepatic de novo lipogenesis in non-alcoholic fatty liver disease. Lipids Health Dis. 2016, 15, 159. [Google Scholar] [CrossRef]
- Lambrecht, J.; Tacke, F. Acetyl-CoA Carboxylase Inhibition as a Therapeutic Tool in the Battle Against NASH: Hitting More Than Just One Mechanism? Cell. Mol. Gastroenterol. Hepatol. 2020, 10, 859–861. [Google Scholar] [CrossRef]
- Ross, T.T.; Crowley, C.; Kelly, K.L.; Rinaldi, A.; Beebe, D.A.; Lech, M.P.; Martinez, R.V.; Carvajal-Gonzalez, S.; Boucher, M.; Hirenallur-Shanthappa, D.; et al. Acetyl-CoA Carboxylase Inhibition Improves Multiple Dimensions of NASH Pathogenesis in Model Systems. Cell. Mol. Gastroenterol. Hepatol. 2020, 10, 829–851. [Google Scholar] [CrossRef]
- Mateo-Marín, M.A.; Alves-Bezerra, M. Targeting acetyl-CoA carboxylases for the treatment of MASLD. J. Lipid Res. 2024, 65, 100676. [Google Scholar] [CrossRef]
- Wang, Y.; Viscarra, J.; Kim, S.-J.; Sul, H.S. Transcriptional regulation of hepatic lipogenesis. Nat. Rev. Mol. Cell Biol. 2015, 16, 678–689. [Google Scholar] [CrossRef]
- Goedeke, L.; Bates, J.; Vatner, D.F.; Perry, R.J.; Wang, T.; Ramirez, R.; Li, L.; Ellis, M.W.; Zhang, D.; Wong, K.E.; et al. Acetyl-CoA Carboxylase Inhibition Reverses NAFLD and Hepatic Insulin Resistance but Promotes Hypertriglyceridemia in Rodents. Hepatology 2018, 68, 2197–2211. [Google Scholar] [CrossRef]
- Zhao, S.; Jang, C.; Liu, J.; Uehara, K.; Gilbert, M.; Izzo, L.; Zeng, X.; Trefely, S.; Fernandez, S.; Carrer, A.; et al. Dietary fructose feeds hepatic lipogenesis via microbiota-derived acetate. Nature 2020, 579, 586–591. [Google Scholar] [CrossRef]
- DiNicolantonio, J.J.; Subramonian, A.M.; O’Keefe, J.H. Added fructose as a principal driver of non-alcoholic fatty liver disease: A public health crisis. Open Heart 2017, 4, e000631. [Google Scholar] [CrossRef] [PubMed]
- Bergman, A.; Carvajal-Gonzalez, S.; Tarabar, S.; Saxena, A.R.; Esler, W.P.; Amin, N.B. Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of a Liver-Targeting Acetyl-CoA Carboxylase Inhibitor (PF-05221304): A Three-Part Randomized Phase 1 Study. Clin. Pharmacol. Drug Dev. 2020, 9, 514–526. [Google Scholar] [CrossRef] [PubMed]
- Kim, C.-W.; Addy, C.; Kusunoki, J.; Anderson, N.N.; Deja, S.; Fu, X.; Burgess, S.C.; Li, C.; Ruddy, M.; Chakravarthy, M.; et al. Acetyl CoA Carboxylase Inhibition Reduces Hepatic Steatosis but Elevates Plasma Triglycerides in Mice and Humans: A Bedside to Bench Investigation. Cell Metab. 2017, 26, 394–406.e6. [Google Scholar] [CrossRef] [PubMed]
- Matsumoto, M.; Yashiro, H.; Ogino, H.; Aoyama, K.; Nambu, T.; Nakamura, S.; Nishida, M.; Wang, X.; Erion, D.M.; Kaneko, M. Acetyl-CoA carboxylase 1 and 2 inhibition ameliorates steatosis and hepatic fibrosis in a MC4R knockout murine model of nonalcoholic steatohepatitis. PLoS ONE 2020, 15, e0228212. [Google Scholar] [CrossRef]
- Stiede, K.; Miao, W.; Blanchette, H.S.; Beysen, C.; Harriman, G.; Harwood, H.J.; Kelley, H.; Kapeller, R.; Schmalbach, T.; Westlin, W.F. Acetyl-coenzyme A carboxylase inhibition reduces de novo lipogenesis in overweight male subjects: A randomized, double-blind, crossover study. Hepatology 2017, 66, 324–334. [Google Scholar] [CrossRef]
- Kelly, K.L.; Reagan, W.J.; Sonnenberg, G.E.; Clasquin, M.; Hales, K.; Asano, S.; Amor, P.A.; Carvajal-Gonzalez, S.; Shirai, N.; Matthews, M.D.; et al. De novo lipogenesis is essential for platelet production in humans. Nat. Metab. 2020, 2, 1163–1178. [Google Scholar] [CrossRef]
- Alkhouri, N.; Lawitz, E.; Noureddin, M.; DeFronzo, R.; Shulman, G.I. GS-0976 (Firsocostat): An investigational liver-directed acetyl-CoA carboxylase (ACC) inhibitor for the treatment of non-alcoholic steatohepatitis (NASH). Expert Opin. Investig. Drugs 2020, 29, 135–141. [Google Scholar] [CrossRef]
- Calle, R.A.; Amin, N.B.; Carvajal-Gonzalez, S.; Ross, T.T.; Bergman, A.; Aggarwal, S.; Crowley, C.; Rinaldi, A.; Mancuso, J.; Aggarwal, N.; et al. ACC inhibitor alone or co-administered with a DGAT2 inhibitor in patients with non-alcoholic fatty liver disease: Two parallel, placebo-controlled, randomized phase 2a trials. Nat. Med. 2021, 27, 1836–1848. [Google Scholar] [CrossRef]
- Sinha, R.A.; Singh, B.K.; Yen, P.M. Direct effects of thyroid hormones on hepatic lipid metabolism. Nat. Rev. Endocrinol. 2018, 14, 259–269. [Google Scholar] [CrossRef]
- Chung, G.E.; Kim, D.; Kim, W.; Yim, J.Y.; Park, M.J.; Kim, Y.J.; Yoon, J.-H.; Lee, H.-S. Non-alcoholic fatty liver disease across the spectrum of hypothyroidism. J. Hepatol. 2012, 57, 150–156. [Google Scholar] [CrossRef]
- Ritter, M.J.; Amano, I.; Hollenberg, A.N. Thyroid Hormone Signaling and the Liver. Hepatology 2020, 72, 742–752. [Google Scholar] [CrossRef] [PubMed]
- The Roles of Autophagy and Thyroid Hormone in the Pathogenesis and Treatment of NAFLD. Available online: https://www.oaepublish.com/articles/2394-5079.2021.82 (accessed on 7 February 2025).
- Araki, O.; Ying, H.; Zhu, X.G.; Willingham, M.C.; Cheng, S.Y. Distinct Dysregulation of Lipid Metabolism by Unliganded Thyroid Hormone Receptor Isoforms. Mol. Endocrinol. 2009, 23, 308–315. [Google Scholar] [CrossRef] [PubMed]
- Jornayvaz, F.R.; Lee, H.-Y.; Jurczak, M.J.; Alves, T.C.; Guebre-Egziabher, F.; Guigni, B.A.; Zhang, D.; Samuel, V.T.; Silva, J.E.; Shulman, G.I. Thyroid Hormone Receptor-α Gene Knockout Mice Are Protected from Diet-Induced Hepatic Insulin Resistance. Endocrinology 2012, 153, 583–591. [Google Scholar] [CrossRef] [PubMed]
- Karim, G.; Bansal, M.B. Resmetirom: An Orally Administered, Smallmolecule, Liver-directed, β-selective THR Agonist for the Treatment of Non-alcoholic Fatty Liver Disease and Non-alcoholic Steatohepatitis. TouchREVIEWS Endocrinol. 2023, 19, 60–70. [Google Scholar] [CrossRef]
- Hönes, G.S.; Sivakumar, R.G.; Hoppe, C.; König, J.; Führer, D.; Moeller, L.C. Cell-Specific Transport and Thyroid Hormone Receptor Isoform Selectivity Account for Hepatocyte-Targeted Thyromimetic Action of MGL-3196. Int. J. Mol. Sci. 2022, 23, 13714. [Google Scholar] [CrossRef]
- Luong, X.G.; Stevens, S.K.; Jekle, A.; Lin, T.-I.; Gupta, K.; Misner, D.; Chanda, S.; Mukherjee, S.; Williams, C.; Stoycheva, A.; et al. Regulation of gene transcription by thyroid hormone receptor β agonists in clinical development for the treatment of non-alcoholic steatohepatitis (NASH). PLoS ONE 2020, 15, e0240338. [Google Scholar] [CrossRef]
- Bianco, A.C.; Kim, B.W. Deiodinases: Implications of the local control of thyroid hormone action. J. Clin. Investig. 2006, 116, 2571–2579. [Google Scholar] [CrossRef]
- FDA. FDA Approves First Treatment for Patients with Liver Scarring Due to Fatty Liver Disease. 2024. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-first-treatment-patients-liver-scarring-due-fatty-liver-disease (accessed on 7 February 2025).
- Harrison, S.A.; Bedossa, P.; Guy, C.D.; Schattenberg, J.M.; Loomba, R.; Taub, R.; Labriola, D.; Moussa, S.E.; Neff, G.W.; Rinella, M.E.; et al. A Phase 3, Randomized, Controlled Trial of Resmetirom in NASH with Liver Fibrosis. N. Engl. J. Med. 2024, 390, 497–509. [Google Scholar] [CrossRef]
- Cusi, K. Selective Agonists of Thyroid Hormone Receptor Beta for the Treatment of NASH. N. Engl. J. Med. 2024, 390, 559–561. [Google Scholar] [CrossRef]
- FDA Grants Tentative Approval to Generic Version of Rifaximin for IBS-D. Available online: https://www.ajmc.com/view/fda-grants-tentative-approval-to-generic-version-of-rifaximin-for-ibs-d (accessed on 7 February 2025).
- Lacy, B.E.; Chang, L.; Rao, S.S.C.; Heimanson, Z.; Sayuk, G.S. Rifaximin Treatment for Individual and Multiple Symptoms of Irritable Bowel Syndrome With Diarrhea: An Analysis Using New End Points. Clin. Ther. 2023, 45, 198–209. [Google Scholar] [CrossRef]
- Lembo, A.; Pimentel, M.; Rao, S.S.; Schoenfeld, P.; Cash, B.; Weinstock, L.B.; Paterson, C.; Bortey, E.; Forbes, W.P. Repeat Treatment With Rifaximin Is Safe and Effective in Patients With Diarrhea-Predominant Irritable Bowel Syndrome. Gastroenterology 2016, 151, 1113–1121. [Google Scholar] [CrossRef] [PubMed]
- Pimentel, M.; Lembo, A.; Chey, W.D.; Zakko, S.; Ringel, Y.; Yu, J.; Mareya, S.M.; Shaw, A.L.; Bortey, E.; Forbes, W.P. Rifaximin Therapy for Patients with Irritable Bowel Syndrome without Constipation. N. Engl. J. Med. 2011, 364, 22–32. [Google Scholar] [CrossRef] [PubMed]
- Kimer, N.; Krag, A.; Møller, S.; Bendtsen, F.; Gluud, L.L. Systematic review with meta-analysis: The effects of rifaximin in hepatic encephalopathy. Aliment. Pharmacol. Ther. 2014, 40, 123–132. [Google Scholar] [CrossRef] [PubMed]
- Scott, L.J. Rifaximin: A Review of Its Use in Reducing Recurrence of Overt Hepatic Encephalopathy Episodes. Drugs 2014, 74, 2153–2160. [Google Scholar] [CrossRef]
- Gastrointestinal Drugs Advisory Committee; Notice of Meeting. Available online: https://www.federalregister.gov/documents/2010/12/02/2010-30274/gastrointestinal-drugs-advisory-committee-notice-of-meeting (accessed on 7 February 2025).
- Patidar, K.R.; Bajaj, J.S. Antibiotics for the treatment of hepatic encephalopathy. Metab. Brain Dis. 2013, 28, 307–312. [Google Scholar] [CrossRef]
- Commissioner, O. of the FDA Approves New Drug to Treat Travelers’ Diarrhea. Available online: https://www.fda.gov/news-events/press-announcements/fda-approves-new-drug-treat-travelers-diarrhea (accessed on 7 February 2025).
- Koo, H.L.; DuPont, H.L. Rifaximin: A unique gastrointestinal-selective antibiotic for enteric diseases. Curr. Opin. Gastroenterol. 2010, 26, 17. [Google Scholar] [CrossRef]
- Hu, Y.; Ren, J.; Zhan, M.; Li, W.; Dai, H. Efficacy of Rifaximin in Prevention of Travelers’ Diarrhea: A Meta-Analysis of Randomized, Double-Blind, Placebo-Controlled Trials. J. Travel Med. 2012, 19, 352–356. [Google Scholar] [CrossRef]
- Dupont, H.L.; Jiang, Z.-D.; Belkind–Gerson, J.; Okhuysen, P.C.; Ericsson, C.D.; Ke, S.; Huang, D.B.; Dupont, M.W.; Adachi, J.A.; Cabada, F.J.D.L.; et al. Treatment of Travelers’ Diarrhea: Randomized Trial Comparing Rifaximin, Rifaximin Plus Loperamide, and Loperamide Alone. Clin. Gastroenterol. Hepatol. 2007, 5, 451–456. [Google Scholar] [CrossRef]
- Abdel-Razik, A.; Mousa, N.; Shabana, W.; Refaey, M.; Elzehery, R.; Elhelaly, R.; Zalata, K.; Abdelsalam, M.; Eldeeb, A.A.; Awad, M.; et al. Rifaximin in nonalcoholic fatty liver disease: Hit multiple targets with a single shot. Eur. J. Gastroenterol. Hepatol. 2018, 30, 1237. [Google Scholar] [CrossRef]
- Zhang, L.; Huang, Y.; Zhou, Y.; Buckley, T.; Wang, H.H. Antibiotic Administration Routes Significantly Influence the Levels of Antibiotic Resistance in Gut Microbiota. Antimicrob. Agents Chemother. 2013, 57, 3659–3666. [Google Scholar] [CrossRef]
- Iorio, N.; Malik, Z.; Schey, R. Profile of rifaximin and its potential in the treatment of irritable bowel syndrome. Clin. Exp. Gastroenterol. 2015, 8, 159–167. [Google Scholar] [CrossRef] [PubMed]
- Combined Therapy with a CCR2/CCR5 Antagonist and FGF21 Analogue Synergizes in Ameliorating Steatohepatitis and Fibrosis. Available online: https://www.mdpi.com/1422-0067/23/12/6696 (accessed on 7 February 2025).
- Harrison, S.A.; Ruane, P.J.; Freilich, B.; Neff, G.; Patil, R.; Behling, C.; Hu, C.; Shringarpure, R.; de Temple, B.; Fong, E.; et al. A randomized, double-blind, placebo-controlled phase IIa trial of efruxifermin for patients with compensated NASH cirrhosis. JHEP Rep. 2023, 5, 100563. [Google Scholar] [CrossRef] [PubMed]
- Harrison, S.A.; Frias, J.P.; Neff, G.; Abrams, G.A.; Lucas, K.J.; Sanchez, W.; Gogia, S.; Sheikh, M.Y.; Behling, C.; Bedossa, P.; et al. Safety and efficacy of once-weekly efruxifermin versus placebo in non-alcoholic steatohepatitis (HARMONY): A multicentre, randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Gastroenterol. Hepatol. 2023, 8, 1080–1093. [Google Scholar] [CrossRef] [PubMed]
- Akero Therapeutics’ Phase 2b SYMMETRY Cohort D Study Met Safety & Tolerability Endpoints and Showed Adding EFX to GLP-1 Therapy Significantly Improved Non-Invasive Markers of NASH-Related Disease—Akero Therapeutics, Inc. Available online: https://ir.akerotx.com/news-releases/news-release-details/akero-therapeutics-phase-2b-symmetry-cohort-d-study-met-safety/ (accessed on 7 February 2025).
- Rinella, M.E.; Lieu, H.D.; Kowdley, K.V.; Goodman, Z.D.; Alkhouri, N.; Lawitz, E.; Ratziu, V.; Abdelmalek, M.F.; Wong, V.W.-S.; Younes, Z.H.; et al. A randomized, double-blind, placebo-controlled trial of aldafermin in patients with NASH and compensated cirrhosis. Hepatology 2024, 79, 674–689. [Google Scholar] [CrossRef]
- Lonardo, A.; Arab, J.P.; Arrese, M. Perspectives on Precision Medicine Approaches to NAFLD Diagnosis and Management. Adv. Ther. 2021, 38, 2130–2158. [Google Scholar] [CrossRef]
- Sveinbjornsson, G.; Ulfarsson, M.O.; Thorolfsdottir, R.B.; Jonsson, B.A.; Einarsson, E.; Gunnlaugsson, G.; Rognvaldsson, S.; Arnar, D.O.; Baldvinsson, M.; Bjarnason, R.G.; et al. Multiomics study of nonalcoholic fatty liver disease. Nat. Genet. 2022, 54, 1652–1663. [Google Scholar] [CrossRef]
- Lin, J.; Zhang, R.; Liu, H.; Zhu, Y.; Dong, N.; Qu, Q.; Bi, H.; Zhang, L.; Luo, O.; Sun, L.; et al. Multi-omics analysis of the biological mechanism of the pathogenesis of non-alcoholic fatty liver disease. Front. Microbiol. 2024, 15, 1379064. [Google Scholar] [CrossRef]
- Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic Species in the Modulation of Gut Microbiota: An Overview. BioMed Res. Int. 2018, 2018, 9478630. [Google Scholar] [CrossRef]
- Cheng, D.; Song, J.; Xie, M.; Song, D. The bidirectional relationship between host physiology and microbiota and health benefits of probiotics: A review. Trends Food Sci. Technol. 2019, 91, 426–435. [Google Scholar] [CrossRef]
- Mennigen, R.; Bruewer, M. Effect of probiotics on intestinal barrier function. Ann. N. Y. Acad. Sci. 2009, 1165, 183–189. [Google Scholar] [CrossRef]
- Yousefi, B.; Eslami, M.; Ghasemian, A.; Kokhaei, P.; Salek Farrokhi, A.; Darabi, N. Probiotics importance and their immunomodulatory properties. J. Cell. Physiol. 2019, 234, 8008–8018. [Google Scholar] [CrossRef] [PubMed]
- Xue, L.; He, J.; Gao, N.; Lu, X.; Li, M.; Wu, X.; Liu, Z.; Jin, Y.; Liu, J.; Xu, J.; et al. Probiotics may delay the progression of nonalcoholic fatty liver disease by restoring the gut microbiota structure and improving intestinal endotoxemia. Sci. Rep. 2017, 7, 45176. [Google Scholar] [CrossRef]
- Kobyliak, N.; Abenavoli, L.; Mykhalchyshyn, G.; Kononenko, L.; Boccuto, L.; Kyriienko, D.; Dynnyk, O. A Multi-strain Probiotic Reduces the Fatty Liver Index, Cytokines and Aminotransferase levels in NAFLD Patients: Evidence from a Randomized Clinical Trial. J. Gastrointest. Liver Dis. JGLD 2018, 27, 41–49. [Google Scholar] [CrossRef]
- Iacono, A.; Raso, G.M.; Canani, R.B.; Calignano, A.; Meli, R. Probiotics as an emerging therapeutic strategy to treat NAFLD: Focus on molecular and biochemical mechanisms. J. Nutr. Biochem. 2011, 22, 699–711. [Google Scholar] [CrossRef]
- Behrouz, V.; Aryaeian, N.; Zahedi, M.J.; Jazayeri, S. Effects of probiotic and prebiotic supplementation on metabolic parameters, liver aminotransferases, and systemic inflammation in nonalcoholic fatty liver disease: A randomized clinical trial. J. Food Sci. 2020, 85, 3611–3617. [Google Scholar] [CrossRef]
- Parnell, J.A.; Raman, M.; Rioux, K.P.; Reimer, R.A. The potential role of prebiotic fibre for treatment and management of non-alcoholic fatty liver disease and associated obesity and insulin resistance. Liver Int. Off. J. Int. Assoc. Study Liver 2012, 32, 701–711. [Google Scholar] [CrossRef]
- Slavin, J. Fiber and prebiotics: Mechanisms and health benefits. Nutrients 2013, 5, 1417–1435. [Google Scholar] [CrossRef]
- Holscher, H.D. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes 2017, 8, 172–184. [Google Scholar] [CrossRef]
- Guo, Q.; Li, Y.; Dai, X.; Wang, B.; Zhang, J.; Cao, H. Polysaccharides: The Potential Prebiotics for Metabolic Associated Fatty Liver Disease (MAFLD). Nutrients 2023, 15, 3722. [Google Scholar] [CrossRef]
- Wang, S.; Sun, W.; Swallah, M.S.; Amin, K.; Lyu, B.; Fan, H.; Zhang, Z.; Yu, H. Preparation and characterization of soybean insoluble dietary fiber and its prebiotic effect on dyslipidemia and hepatic steatosis in high fat-fed C57BL/6J mice. Food Funct. 2021, 12, 8760–8773. [Google Scholar] [CrossRef]
- Hadi, A.; Mohammadi, H.; Miraghajani, M.; Ghaedi, E. Efficacy of synbiotic supplementation in patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis of clinical trials: Synbiotic supplementation and NAFLD. Crit. Rev. Food Sci. Nutr. 2019, 59, 2494–2505. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Fang, T.; Shi, L.; Wang, Y.; Deng, X.; Wang, J.; Zhou, Y. The synbiotic combination of probiotics and inulin improves NAFLD though modulating gut microbiota. J. Nutr. Biochem. 2024, 125, 109546. [Google Scholar] [CrossRef]
- Khan, M.Y.; Mihali, A.B.; Rawala, M.S.; Aslam, A.; Siddiqui, W.J. The promising role of probiotic and synbiotic therapy in aminotransferase levels and inflammatory markers in patients with nonalcoholic fatty liver disease—A systematic review and meta-analysis. Eur. J. Gastroenterol. Hepatol. 2019, 31, 703–715. [Google Scholar] [CrossRef] [PubMed]
- Vyas, U.; Ranganathan, N. Probiotics, prebiotics, and synbiotics: Gut and beyond. Gastroenterol. Res. Pract. 2012, 2012, 872716. [Google Scholar] [CrossRef]
- Allegretti, J.R.; Mullish, B.H.; Kelly, C.; Fischer, M. The evolution of the use of faecal microbiota transplantation and emerging therapeutic indications. Lancet 2019, 394, 420–431. [Google Scholar] [CrossRef]
- Non-Alcoholic Fatty Liver Disease and Gut Microbial Dysbiosis—Underlying Mechanisms and Gut Microbiota Mediated Treatment Strategies. Available online: http://ouci.dntb.gov.ua/en/works/4bwZWedl/ (accessed on 7 February 2025).
- Craven, L.; Rahman, A.; Nair Parvathy, S.; Beaton, M.; Silverman, J.; Qumosani, K.; Hramiak, I.; Hegele, R.; Joy, T.; Meddings, J.; et al. Allogenic Fecal Microbiota Transplantation in Patients With Nonalcoholic Fatty Liver Disease Improves Abnormal Small Intestinal Permeability: A Randomized Control Trial. Am. J. Gastroenterol. 2020, 115, 1055–1065. [Google Scholar] [CrossRef]
- Del Barrio, M.; Lavín, L.; Santos-Laso, Á.; Arias-Loste, M.T.; Odriozola, A.; Rodriguez-Duque, J.C.; Rivas, C.; Iruzubieta, P.; Crespo, J. Faecal Microbiota Transplantation, Paving the Way to Treat Non-Alcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2023, 24, 6123. [Google Scholar] [CrossRef]
- Popa, S.L.; Ismaiel, A.; Cristina, P.; Cristina, M.; Chiarioni, G.; David, L.; Dumitrascu, D.L. Non-Alcoholic Fatty Liver Disease: Implementing Complete Automated Diagnosis and Staging. A Systematic Review. Diagnostics 2021, 11, 1078. [Google Scholar] [CrossRef]
- Yang, B.; Lu, H.; Ran, Y. Advancing non-alcoholic fatty liver disease prediction: A comprehensive machine learning approach integrating SHAP interpretability and multi-cohort validation. Front. Endocrinol. 2024, 15, 1450317. [Google Scholar] [CrossRef]
- Decharatanachart, P.; Chaiteerakij, R.; Tiyarattanachai, T.; Treeprasertsuk, S. Application of artificial intelligence in non-alcoholic fatty liver disease and liver fibrosis: A systematic review and meta-analysis. Ther. Adv. Gastroenterol. 2021, 14, 17562848211062807. [Google Scholar] [CrossRef]
- Ma, X.; Yang, C.; Liang, K.; Sun, B.; Jin, W.; Chen, L.; Dong, M.; Liu, S.; Xin, Y.; Zhuang, L. A predictive model for the diagnosis of non-alcoholic fatty liver disease based on an integrated machine learning method. Am. J. Transl. Res. 2021, 13, 12704–12713. [Google Scholar] [PubMed]
- Sghaireen, M.G.; Al-Smadi, Y.; Al-Qerem, A.; Srivastava, K.C.; Ganji, K.K.; Alam, M.K.; Nashwan, S.; Khader, Y. Machine Learning Approach for Metabolic Syndrome Diagnosis Using Explainable Data-Augmentation-Based Classification. Diagnostics 2022, 12, 3117. [Google Scholar] [CrossRef] [PubMed]
- Cao, W.; An, X.; Cong, L.; Lyu, C.; Zhou, Q.; Guo, R. Application of Deep Learning in Quantitative Analysis of 2-Dimensional Ultrasound Imaging of Nonalcoholic Fatty Liver Disease. J. Ultrasound Med. Off. J. Am. Inst. Ultrasound Med. 2020, 39, 51–59. [Google Scholar] [CrossRef] [PubMed]
- Pugliese, N.; Bertazzoni, A.; Hassan, C.; Schattenberg, J.M.; Aghemo, A. Revolutionizing MASLD: How Artificial Intelligence Is Shaping the Future of Liver Care. Cancers 2025, 17, 722. [Google Scholar] [CrossRef]
- Aggarwal, P.; Alkhouri, N. Artificial Intelligence in Nonalcoholic Fatty Liver Disease: A New Frontier in Diagnosis and Treatment. Clin. Liver Dis. 2021, 17, 392–397. [Google Scholar] [CrossRef]
- Xia, J.; Wang, Z.; Huan, Y.; Xue, W.; Wang, X.; Wang, Y.; Liu, Z.; Hsieh, J.-H.; Zhang, L.; Wu, S.; et al. Pose Filter-Based Ensemble Learning Enables Discovery of Orally Active, Nonsteroidal Farnesoid X Receptor Agonists. J. Chem. Inf. Model. 2020, 60, 1202–1214. [Google Scholar] [CrossRef]
- Velliou, R.-I.; Giannousi, E.; Ralliou, C.; Kassi, E.; Chatzigeorgiou, A. Ex Vivo Tools and Models in MASLD Research. Cells 2024, 13, 1827. [Google Scholar] [CrossRef]
- De Chiara, F.; Ferret-Miñana, A.; Ramón-Azcón, J. The Synergy between Organ-on-a-Chip and Artificial Intelligence for the Study of NAFLD: From Basic Science to Clinical Research. Biomedicines 2021, 9, 248. [Google Scholar] [CrossRef]
- Chu, A.; Nguyen, D.; Talathi, S.S.; Wilson, A.C.; Ye, C.; Smith, W.L.; Kaplan, A.D.; Duoss, E.B.; Stolaroff, J.K.; Giera, B. Automated detection and sorting of microencapsulation via machine learning. Lab. Chip 2019, 19, 1808–1817. [Google Scholar] [CrossRef]
- Kalapala, R.; Rughwani, H.; Reddy, D.N. Artificial Intelligence in Hepatology—Ready for the Primetime. J. Clin. Exp. Hepatol. 2023, 13, 149–161. [Google Scholar] [CrossRef]
- Reiniš, J.; Petrenko, O.; Simbrunner, B.; Hofer, B.S.; Schepis, F.; Scoppettuolo, M.; Saltini, D.; Indulti, F.; Guasconi, T.; Albillos, A.; et al. Assessment of portal hypertension severity using machine learning models in patients with compensated cirrhosis. J. Hepatol. 2023, 78, 390–400. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Wang, X.; Zhang, J.; Zhang, S.; Jiao, J. Applications of artificial intelligence (AI) in researches on non-alcoholic fatty liver disease(NAFLD): A systematic review. Rev. Endocr. Metab. Disord. 2022, 23, 387–400. [Google Scholar] [CrossRef] [PubMed]
- Baser, O.; Samayoa, G.; Yapar, N.; Baser, E. Artificial Intelligence in Identifying Patients With Undiagnosed Nonalcoholic Steatohepatitis. J. Health Econ. Outcomes Res. 2024, 11, 86–94. [Google Scholar] [CrossRef]
- Hagström, H.; Vessby, J.; Ekstedt, M.; Shang, Y. 99% of patients with NAFLD meet MASLD criteria and natural history is therefore identical. J. Hepatol. 2024, 80, e76–e77. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Boulos, M.; Mousa, R.S.; Jeries, N.; Simaan, E.; Alam, K.; Bulus, B.; Assy, N. Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome. Int. J. Mol. Sci. 2025, 26, 3448. https://doi.org/10.3390/ijms26073448
Boulos M, Mousa RS, Jeries N, Simaan E, Alam K, Bulus B, Assy N. Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome. International Journal of Molecular Sciences. 2025; 26(7):3448. https://doi.org/10.3390/ijms26073448
Chicago/Turabian StyleBoulos, Mariana, Rabia S. Mousa, Nizar Jeries, Elias Simaan, Klode Alam, Bulus Bulus, and Nimer Assy. 2025. "Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome" International Journal of Molecular Sciences 26, no. 7: 3448. https://doi.org/10.3390/ijms26073448
APA StyleBoulos, M., Mousa, R. S., Jeries, N., Simaan, E., Alam, K., Bulus, B., & Assy, N. (2025). Hidden in the Fat: Unpacking the Metabolic Tango Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Metabolic Syndrome. International Journal of Molecular Sciences, 26(7), 3448. https://doi.org/10.3390/ijms26073448