1
|
Nikoli A, Orfanidou M, Goulas A, Goulis DG, Polyzos SA. Circulating lipoprotein(a) in patients with nonalcoholic fatty liver disease: a systematic review and meta-analysis. J Gastroenterol Hepatol 2024; 39:2572-2581. [PMID: 39417560 PMCID: PMC11660206 DOI: 10.1111/jgh.16768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/21/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND AND AIM Although nonalcoholic fatty liver disease (NAFLD) and lipoprotein(a) [Lp(a)] are associated with cardiovascular diseases, existing data on Lp(a) in NAFLD are conflicting. The aim of this systematic review and meta-analysis was to summarize and compare data on circulating Lp(a) between NAFLD patients and non-NAFLD controls. METHODS A systematic literature search was performed in PubMed, Scopus, and Cochrane Library. This meta-analysis included 18 studies containing data on 74 691 individuals (20 220 patients with NAFLD and 54 471 controls). RESULTS Circulating Lp(a) was similar between patients with NAFLD and controls (standardized mean difference [SMD] 0.09; 95% confidence interval [95% CI] -0.21, 0.38). The heterogeneity among studies was high (I2 = 100%); no publication bias was detected (Egger's test P = 0.941). However, in subgroup analysis, Lp(a) was lower in NAFLD patients than controls, when Lp(a) was measured with nephelometry (SMD -0.26; 95% CI -0.46, -0.06), but not turbidimetry; this analysis also resulted in mild reduction of heterogeneity within the subgroup of nephelometry (I2 = 87%). The sensitivity analyses, based on the exclusion of studies with Newcastle-Ottawa Scale score ≤6 (n = 5), studies in which liver biopsy was used for NAFLD diagnosis (n = 4) or studies that adopted the criteria of metabolic dysfunction-associated fatty liver disease (n = 2), and meta-regression analysis did not explain the high heterogeneity among studies. CONCLUSIONS Overall, circulating Lp(a) was similar between NAFLD patients and non-NAFLD controls; however, patients with NAFLD had lower circulating Lp(a) compared with controls, when Lp(a) was measured with nephelometry. These results should be cautiously interpreted, because of the high heterogeneity among studies.
Collapse
Affiliation(s)
- Aikaterini Nikoli
- First Laboratory of Pharmacology, School of MedicineAristotle University of ThessalonikiThessalonikiGreece
| | - Myrsini Orfanidou
- First Laboratory of Pharmacology, School of MedicineAristotle University of ThessalonikiThessalonikiGreece
| | - Antonis Goulas
- First Laboratory of Pharmacology, School of MedicineAristotle University of ThessalonikiThessalonikiGreece
| | - Dimitrios G. Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, School of MedicineAristotle University of ThessalonikiThessalonikiGreece
| | - Stergios A. Polyzos
- First Laboratory of Pharmacology, School of MedicineAristotle University of ThessalonikiThessalonikiGreece
| |
Collapse
|
2
|
Kathuria I, Prasad A, Sharma BK, Aithabathula RV, Ofosu-Boateng M, Gyamfi MA, Jiang J, Park F, Singh UP, Singla B. Nidogen 2 Overexpression Promotes Hepatosteatosis and Atherosclerosis. Int J Mol Sci 2024; 25:12782. [PMID: 39684493 DOI: 10.3390/ijms252312782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Clinical and genetic studies strongly support a significant connection between nonalcoholic fatty liver disease (NAFLD) and atherosclerotic cardiovascular disease (ASCVD) and identify ASCVD as the primary cause of death in NAFLD patients. Understanding the molecular factors and mechanisms regulating these diseases is critical for developing novel therapies that target them simultaneously. Our preliminary immunoblotting experiments demonstrated elevated expression of nidogen 2 (NID2), a basement membrane glycoprotein, in human atherosclerotic vascular tissues and murine steatotic livers. Therefore, we investigated the role of NID2 in regulating hepatosteatosis and atherosclerosis utilizing Western diet-fed Apoe-/- mice with/without NID2 overexpression. Quantitative real-time PCR confirmed increased NID2 mRNA expression in multiple organs (liver, heart, kidney, and adipose) of NID2-overexpressing mice. Male mice with NID2 overexpression exhibited higher liver and epididymal white adipose tissue mass, increased hepatic lipid accumulation, and fibrosis. Additionally, these mice developed larger atherosclerotic lesions in the whole aortas and aortic roots, with increased necrotic core formation. Mechanistic studies showed reduced AMPK activation in the livers of NID2-overexpressing mice compared with controls, without any effects on hepatic inflammation. In conclusion, these findings suggest that NID2 plays a deleterious role in both hepatosteatosis and atherosclerosis, making it a potential therapeutic target for these conditions.
Collapse
Affiliation(s)
- Ishita Kathuria
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Aditi Prasad
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Bal Krishan Sharma
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Malvin Ofosu-Boateng
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Maxwell A Gyamfi
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Udai P Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Bhupesh Singla
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
3
|
Balta C, Herman H, Ciceu A, Lepre CC, Mladin B, Rosu M, Oatis D, Russo M, Peteu VE, Gherghiceanu M, Fenyvesi F, Cotoraci C, Trotta MC, D'Amico M, Hermenean A. Chrysin-loaded calixarene-cyclodextrin ternary drug delivery system inhibits TGF-β and galectin-1 mediated pathways in diabetic liver fibrosis. Biochem Pharmacol 2024; 229:116474. [PMID: 39122218 DOI: 10.1016/j.bcp.2024.116474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/12/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
This study investigated the efficacy of a new chrysin-loaded calixarene-cyclodextrin ternary drug delivery system (DDS) in reversing liver fibrosis in a mouse model of chronic diabetes. The system was designed to enhance the solubility and bioavailability of chrysin (CHR) and calixarene 0118 (OTX008). Adult male CD1 mice received streptozotocin (STZ) injections to induce diabetes. After 20 weeks, they underwent intraperitoneal treatments twice weekly for a two-week period. Histological analyses revealed that long-term hyperglycaemia increased liver fibrosis and altered hepatic ultrastructure, characterized by lipid accumulation, hepatic stellate cell activation, and collagen deposition. The treatment with the chrysin-loaded DDS restored liver structure closely to normal levels, as opposed to the minimal impact observed with sulfobutylated β-cyclodextrin (SBECD) alone. The treatment significantly decreased serum activities of alanine /aspartate transaminases and reduced the gene expression of collagen type I (Col-I). It also modulated the transforming growth factor beta 1 (TGF-β1)/Smad signalling pathway, inhibiting the activation and proliferation of hepatic stellate cells. The treatment led to a downregulation of the TGF-β1 gene and its receptors TGFβR1 and TGFβR2, together with a decrease in Smad 2 and 3 mRNA levels. Conversely, Smad 7 mRNA expression was increased by the DDS. Furthermore, it downregulated galectin-1 (Gal-1) gene and protein levels, which correlated with fibrotic markers. In conclusion, the chrysin-loaded calixarene-cyclodextrin ternary DDS presents a promising therapeutic approach for diabetic liver fibrosis, effectively targeting fibrotic pathways and restoring hepatic function and structure.
Collapse
Affiliation(s)
- Cornel Balta
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310025 Arad, Romania
| | - Hildegard Herman
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310025 Arad, Romania
| | - Alina Ciceu
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310025 Arad, Romania
| | - Caterina Claudia Lepre
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy; PhD Course in Translational Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Bianca Mladin
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310025 Arad, Romania
| | - Marcel Rosu
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310025 Arad, Romania
| | - Daniela Oatis
- Doctoral School of Biology, Faculty of Medicine, Vasile Goldis Western University of Arad, Arad, Romania
| | - Marina Russo
- PhD Course in National Interest in Public Administration and Innovation for Disability and Social Inclusion, Department of Mental, Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy; School of Pharmacology and Clinical Toxicology, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | | | - Mihaela Gherghiceanu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; Department of Cell Biology, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ferenc Fenyvesi
- Department of Molecular Pharmaceutics and Nanopharmaceutics, Faculty of Pharmacy, University of Debrecen, H-4032 Debrecen, Hungary
| | - Coralia Cotoraci
- Department of Haematology, Faculty of Medicine, Vasile Goldis Western University of Arad, 310025 Arad, Romania
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Michele D'Amico
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Anca Hermenean
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 310025 Arad, Romania; Department of Histology, Faculty of Medicine, Vasile Goldis Western University of Arad, 310025 Arad, Romania.
| |
Collapse
|
4
|
de Paula D, Feter N, Dos Reis RCP, Griep RH, Duncan BB, Schmidt MI. Associations of activity, sedentary and sleep behaviors with prevalent steatotic liver disease in middle-aged and older adults: the ELSA-Brasil study. JOURNAL OF ACTIVITY, SEDENTARY AND SLEEP BEHAVIORS 2024; 3:16. [PMID: 40217540 PMCID: PMC11960374 DOI: 10.1186/s44167-024-00055-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/11/2024] [Indexed: 04/14/2025]
Abstract
BACKGROUND Steatotic liver disease (SLD) is a prevalent metabolic disease. While single component movement behaviors have been related to its development, comprehensive assessments of their joint associations are scarce. OBJECTIVE To investigate the single-component and multi-component associations of moderate and vigorous physical activity (MVPA), light physical activity (LPA), sedentary behavior (SB), and sleep with prevalent SLD in Brazilian adults. METHODS A cross-sectional analysis using data from the third wave of the ELSA-Brasil cohort (2017-2019). Participants wore an ActiGraph wGT3X-BT in the waist for seven days and completed a sleep diary. SLD was defined by a Fatty Liver Index ≥ 60. To investigate single-component and multi-component associations, we used three exposure modeling approaches based on Poisson models: multivariable-adjusted regression, restricted cubic splines, and compositional data analysis. RESULTS Among 8569 participants (55.7% women, mean age 59.2 ± 8.60), 43.9% had SLD. Total activity volume adjusted for covariates was inversely associated with prevalent SLD. Every 1 mg/day increase in total activity volume was associated with a PR of 0.95 in individuals sleeping < 7 h/day (95% CI 0.94-0.97) and 0.95 (95% CI 0.93-0.96) in those sleeping ≥ 7 h/day. Increasing 30 min/day of MVPA was associated with decreasing prevalence of SLD (sleep ≥ 7 h/day [PR 0.83; 95% CI 0.77-0.89]; sleep ≥ 7 h/day [PR 0.78; 95% CI 0.74-0.83]). Sleep, SB, and LPA were not associated with SLD. Associations of total activity volume and MVPA were more pronounced among females. Adjustment with adiposity markers attenuated the associations. CONCLUSIONS In adults, total activity volume and MVPA were inversely associated with SLD in a dose-response fashion. Substituting lower-intensity behaviors with MVPA was associated with a lower prevalence of SLD independent of sleep duration, sex, and age.
Collapse
Affiliation(s)
- Danilo de Paula
- Department of Epidemiology, Universidade Federal do Rio Grande do Sul, 2400 Ramiro Barcelos St.-2nd floor, Porto Alegre, Rio Grande Do Sul, 90035-003, Brazil.
- Center for Clinical Research, Hospital de Clínicas de Porto Alegre, 2350 Ramiro Barcelos St.-Building 21, 4 Floor, Porto Alegre, Rio Grande do sul, 90035-003, Brazil.
| | - Natan Feter
- Department of Epidemiology, Universidade Federal do Rio Grande do Sul, 2400 Ramiro Barcelos St.-2nd floor, Porto Alegre, Rio Grande Do Sul, 90035-003, Brazil
| | - Rodrigo Citton Padilha Dos Reis
- Department of Epidemiology, Universidade Federal do Rio Grande do Sul, 2400 Ramiro Barcelos St.-2nd floor, Porto Alegre, Rio Grande Do Sul, 90035-003, Brazil
- Department of Statistics, Universidade Federal do Rio Grande do Sul, 9500 Bento Gonçalves Av Building 43, Porto Alegre, Rio Grande Do Sul, 91509-900, Brazil
| | - Rosane Harter Griep
- Laboratório de Educação em Ambiente e Saúde, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 1480 Leopoldo Bulhões St., Rio de Janeiro, 21041210, Brazil
| | - Bruce Bartholow Duncan
- Department of Epidemiology, Universidade Federal do Rio Grande do Sul, 2400 Ramiro Barcelos St.-2nd floor, Porto Alegre, Rio Grande Do Sul, 90035-003, Brazil
- Center for Clinical Research, Hospital de Clínicas de Porto Alegre, 2350 Ramiro Barcelos St.-Building 21, 4 Floor, Porto Alegre, Rio Grande do sul, 90035-003, Brazil
| | - Maria Inês Schmidt
- Department of Epidemiology, Universidade Federal do Rio Grande do Sul, 2400 Ramiro Barcelos St.-2nd floor, Porto Alegre, Rio Grande Do Sul, 90035-003, Brazil
- Center for Clinical Research, Hospital de Clínicas de Porto Alegre, 2350 Ramiro Barcelos St.-Building 21, 4 Floor, Porto Alegre, Rio Grande do sul, 90035-003, Brazil
| |
Collapse
|
5
|
Semertzidis A, Mouskeftara T, Gika H, Pousinis P, Makedou K, Goulas A, Kountouras J, Polyzos SA. Effects of Combined Low-Dose Spironolactone Plus Vitamin E versus Vitamin E Monotherapy on Lipidomic Profile in Non-Alcoholic Fatty Liver Disease: A Post Hoc Analysis of a Randomized Controlled Trial. J Clin Med 2024; 13:3798. [PMID: 38999363 PMCID: PMC11242225 DOI: 10.3390/jcm13133798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/13/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Background/Objectives: Lipid dysmetabolism seems to contribute to the development and progression of nonalcoholic fatty liver disease (NAFLD). Our aim was to compare serum lipidomic profile between patients with NAFLD having received monotherapy with vitamin E (400 IU/d) and those having received combination therapy with vitamin E (400 IU/d) and low-dose spironolactone (25 mg/d) for 52 weeks. Methods: This was a post hoc study of a randomized controlled trial (NCT01147523). Serum lipidomic analysis was performed in vitamin E monotherapy group (n = 15) and spironolactone plus vitamin E combination therapy group (n = 12). We employed an untargeted liquid chromatography-mass spectrometry lipid profiling approach in positive and negative ionization mode. Results: Univariate analysis revealed 36 lipid molecules statistically different between groups in positive mode and seven molecules in negative mode. Multivariate analysis in negative mode identified six lipid molecules that remained robustly different between groups. After adjustment for potential confounders, including gender, omega-3 supplementation, leptin concentration and homeostasis model assessment-insulin resistance (HOMA-IR), four lipid molecules remained significant between groups: FA 20:5, SM 34:2;O2, SM 42:3;O2 and CE 22:6, all being higher in the combination treatment group. Conclusions: The combination of spironolactone with vitamin E led to higher circulating levels of four lipid molecules than vitamin E monotherapy, after adjustment for potential confounders. Owing to very limited relevant data, we could not support that these changes in lipid molecules may be beneficial or not for the progression of NAFLD. Thus, mechanistic studies are warranted to clarify the potential clinical significance of these findings.
Collapse
Affiliation(s)
- Anastasios Semertzidis
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Thomai Mouskeftara
- Laboratory of Forensic Medicine & Toxicology, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Helen Gika
- Laboratory of Forensic Medicine & Toxicology, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
- BIOMIC AUTh, Center for Interdisciplinary Research and Innovation, Aristotle University of Thessaloniki, 570 01 Thessaloniki, Greece
| | - Petros Pousinis
- BIOMIC AUTh, Center for Interdisciplinary Research and Innovation, Aristotle University of Thessaloniki, 570 01 Thessaloniki, Greece
| | - Kali Makedou
- Laboratory of Biochemistry, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Antonis Goulas
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Jannis Kountouras
- Second Medical Clinic, Ippokration General Hospital, School of Medicine, Aristotle University of Thessaloniki, 546 42 Thessaloniki, Greece
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| |
Collapse
|
6
|
Knuth MM, Xue J, Elnagheeb M, Gharaibeh RZ, Schoenrock SA, McRitchie S, Brouwer C, Sumner SJ, Tarantino L, Valdar W, Rector RS, Simon JM, Ideraabdullah F. Early life exposure to vitamin D deficiency impairs molecular mechanisms that regulate liver cholesterol biosynthesis, energy metabolism, inflammation, and detoxification. Front Endocrinol (Lausanne) 2024; 15:1335855. [PMID: 38800476 PMCID: PMC11116800 DOI: 10.3389/fendo.2024.1335855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Emerging data suggests liver disease may be initiated during development when there is high genome plasticity and the molecular pathways supporting liver function are being developed. Methods Here, we leveraged our Collaborative Cross mouse model of developmental vitamin D deficiency (DVD) to investigate the role of DVD in dysregulating the molecular mechanisms underlying liver disease. We defined the effects on the adult liver transcriptome and metabolome and examined the role of epigenetic dysregulation. Given that the parental origin of the genome (POG) influences response to DVD, we used our established POG model [POG1-(CC011xCC001)F1 and POG2-(CC001xCC011)F1] to identify interindividual differences. Results We found that DVD altered the adult liver transcriptome, primarily downregulating genes controlling liver development, response to injury/infection (detoxification & inflammation), cholesterol biosynthesis, and energy production. In concordance with these transcriptional changes, we found that DVD decreased liver cell membrane-associated lipids (including cholesterol) and pentose phosphate pathway metabolites. Each POG also exhibited distinct responses. POG1 exhibited almost 2X more differentially expressed genes (DEGs) with effects indicative of increased energy utilization. This included upregulation of lipid and amino acid metabolism genes and increased intermediate lipid and amino acid metabolites, increased energy cofactors, and decreased energy substrates. POG2 exhibited broader downregulation of cholesterol biosynthesis genes with a metabolomics profile indicative of decreased energy utilization. Although DVD primarily caused loss of liver DNA methylation for both POGs, only one epimutation was shared, and POG2 had 6.5X more differentially methylated genes. Differential methylation was detected at DEGs regulating developmental processes such as amino acid transport (POG1) and cell growth & differentiation (e.g., Wnt & cadherin signaling, POG2). Conclusions These findings implicate a novel role for maternal vitamin D in programming essential offspring liver functions that are dysregulated in liver disease. Importantly, impairment of these processes was not rescued by vitamin D treatment at weaning, suggesting these effects require preventative measures. Substantial differences in POG response to DVD demonstrate that the parental genomic context of exposure determines offspring susceptibility.
Collapse
Affiliation(s)
- Megan M. Knuth
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jing Xue
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Marwa Elnagheeb
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Raad Z. Gharaibeh
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, United States
| | - Sarah A. Schoenrock
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susan McRitchie
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
| | - Cory Brouwer
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, United States
- University of North Carolina at Charlotte Bioinformatics Service Division, North Carolina Research Campus, Kannapolis, NC, United States
| | - Susan J. Sumner
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Lisa Tarantino
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - William Valdar
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - R. Scott Rector
- Research Service, Harry S. Truman Memorial Veterans Medical Center, Columbia, MO, United States
- NextGen Precision Health, University of Missouri, Columbia, MO, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Jeremy M. Simon
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Neuroscience Center Bioinformatics Core, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Folami Ideraabdullah
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC, United States
- Department of Nutrition, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
7
|
Kokkorakis M, Muzurović E, Volčanšek Š, Chakhtoura M, Hill MA, Mikhailidis DP, Mantzoros CS. Steatotic Liver Disease: Pathophysiology and Emerging Pharmacotherapies. Pharmacol Rev 2024; 76:454-499. [PMID: 38697855 DOI: 10.1124/pharmrev.123.001087] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 05/05/2024] Open
Abstract
Steatotic liver disease (SLD) displays a dynamic and complex disease phenotype. Consequently, the metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH) therapeutic pipeline is expanding rapidly and in multiple directions. In parallel, noninvasive tools for diagnosing and monitoring responses to therapeutic interventions are being studied, and clinically feasible findings are being explored as primary outcomes in interventional trials. The realization that distinct subgroups exist under the umbrella of SLD should guide more precise and personalized treatment recommendations and facilitate advancements in pharmacotherapeutics. This review summarizes recent updates of pathophysiology-based nomenclature and outlines both effective pharmacotherapeutics and those in the pipeline for MASLD/MASH, detailing their mode of action and the current status of phase 2 and 3 clinical trials. Of the extensive arsenal of pharmacotherapeutics in the MASLD/MASH pipeline, several have been rejected, whereas other, mainly monotherapy options, have shown only marginal benefits and are now being tested as part of combination therapies, yet others are still in development as monotherapies. Although the Food and Drug Administration (FDA) has recently approved resmetirom, additional therapeutic approaches in development will ideally target MASH and fibrosis while improving cardiometabolic risk factors. Due to the urgent need for the development of novel therapeutic strategies and the potential availability of safety and tolerability data, repurposing existing and approved drugs is an appealing option. Finally, it is essential to highlight that SLD and, by extension, MASLD should be recognized and approached as a systemic disease affecting multiple organs, with the vigorous implementation of interdisciplinary and coordinated action plans. SIGNIFICANCE STATEMENT: Steatotic liver disease (SLD), including metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis, is the most prevalent chronic liver condition, affecting more than one-fourth of the global population. This review aims to provide the most recent information regarding SLD pathophysiology, diagnosis, and management according to the latest advancements in the guidelines and clinical trials. Collectively, it is hoped that the information provided furthers the understanding of the current state of SLD with direct clinical implications and stimulates research initiatives.
Collapse
Affiliation(s)
- Michail Kokkorakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Emir Muzurović
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Špela Volčanšek
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Marlene Chakhtoura
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Michael A Hill
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Dimitri P Mikhailidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (M.K., C.S.M.); Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands (M.K.); Endocrinology Section, Department of Internal Medicine, Clinical Center of Montenegro, Podgorica, Montenegro (E.M.); Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.); Department of Endocrinology, Diabetes, and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia (Š.V.); Medical Faculty Ljubljana, Ljubljana, Slovenia (Š.V.); Division of Endocrinology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon (M.C.); Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri (M.A.H.); Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri (M.A.H.); Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom (D.P.M.); Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates (D.P.M.); and Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts (C.S.M.)
| |
Collapse
|
8
|
Makri ES, Makri E, Goulas A, Xanthopoulos K, Polyzos SA. Animal studies of sodium-glucose co-transporter 2 inhibitors in nonalcoholic fatty liver disease. Ann Gastroenterol 2024; 37:280-290. [PMID: 38779641 PMCID: PMC11107411 DOI: 10.20524/aog.2024.0884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/10/2024] [Indexed: 05/25/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered one of the most common chronic liver diseases. Modern lifestyle, characterized by increasing rates of obesity and type 2 diabetes mellitus (T2DM), has led to a "pandemic" of NAFLD that imposes a personal health and socioeconomic burden. Apart from overnutrition and insulin resistance, various metabolic aberrations, gut microbiota and genetic predispositions are involved in the pathogenesis of the disease. The multifactorial nature of NAFLD's pathogenesis makes the development of pharmacological therapies for patients with this disease challenging. Sodium-glucose co-transporter 2 inhibitors (SGLT-2i) are antidiabetic agents that reduce blood glucose mainly by increasing its renal excretion. As T2DM is one of the major contributors to NAFLD, SGLT-2i have emerged as promising agents for the management of NAFLD. In this review, we summarize the main animal studies on SGLT-2i in models of NAFLD.
Collapse
Affiliation(s)
- Evangelia S. Makri
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki (Evangelia S. Makri, Eleftheria Makri, Antonis Goulas, Stergios A. Polyzos)
| | - Eleftheria Makri
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki (Evangelia S. Makri, Eleftheria Makri, Antonis Goulas, Stergios A. Polyzos)
| | - Antonis Goulas
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki (Evangelia S. Makri, Eleftheria Makri, Antonis Goulas, Stergios A. Polyzos)
| | - Konstantinos Xanthopoulos
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki (Konstantinos Xanthopoulos)
- Institute of Applied Biosciences, Centre for Research and Technology, Thessaloniki (Konstantinos Xanthopoulos), Greece
| | - Stergios A. Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki (Evangelia S. Makri, Eleftheria Makri, Antonis Goulas, Stergios A. Polyzos)
| |
Collapse
|
9
|
Valenzuela-Vallejo L, Chrysafi P, Kouvari M, Guatibonza-Garcia V, Mylonakis SC, Katsarou A, Verrastro O, Markakis G, Eslam M, Papatheodoridis G, Mingrone G, George J, Mantzoros CS. Circulating hormones in biopsy-proven steatotic liver disease and steatohepatitis: A Multicenter Observational Study. Metabolism 2023; 148:155694. [PMID: 37757973 DOI: 10.1016/j.metabol.2023.155694] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/06/2023] [Accepted: 09/24/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND The role of metabolic/inflammatory hormonal systems in metabolic dysfunction associated steatotic liver disease (MASLD) remains to be fully elucidated. PURPOSE To report the levels of the novel total and H-specific growth differentiation factor-15 (GDF-15) and other established hormonal systems and to describe hormonal patterns in controls and patients with MASLD and its stages. METHODS This is a multicenter study from two Gastroenterology-Hepatology Departments (Greece and Australia) and one Bariatric-Metabolic Surgery Department (Italy). Overall, n = 455 serum samples of patients with biopsy-proven MASLD (n = 374) and Controls (n = 81) were recruited. RESULTS We report for the first time that total and H-specific GDF-15 levels are higher in MASLD, at-risk metabolic dysfunction associated steatohepatitis (MASH), and severe fibrosis than in Controls. In addition, follistatin-like-3 (FSTL-3), free insulin-like growth factor-1 (IGF-1), leptin, and insulin levels were higher in MASLD patients than in Controls, while adiponectin levels were lower in MASLD subjects than in Controls. Activin-A, follistatin (FST), FSTL-3, and insulin levels significantly increased in severe fibrosis compared to no/mild fibrosis, while free IGF-1 decreased. In addition, adiponectin levels were lower in subjects without fibrosis vs. any fibrosis. Moreover, GDF-15 presented a strong positive association for the likelihood of having MASLD and at-risk MASH, while in adjusted analyses, FST and adiponectin showed inverse associations. Two different patterns of at-risk MASH were revealed through unsupervised analysis (total variation explained=54%). The most frequent pattern met in our sample (34.3%) was characterized by higher levels of total and H-specific GDF-15, follistatins, and activins, as well as low adiponectin levels. The second pattern revealed was characterized by high levels of free IGF-1, insulin, and leptin, with low levels of activin-A and adiponectin. Similar patterns were also generated in the case of overall MASLD. CONCLUSIONS Total and H-specific GDF-15 levels increase as MASLD severity progresses. FSTL-3, free IGF-1, leptin, and insulin are also higher, whereas adiponectin and activin-A levels are lower in the MASLD group than in Controls. Hormonal systems, including GDF-15, may not only be involved in the pathophysiology but could also prove useful for the diagnostic workup of MASLD and its stages and may potentially be of therapeutic value.
Collapse
Affiliation(s)
- Laura Valenzuela-Vallejo
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Pavlina Chrysafi
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Matina Kouvari
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Valentina Guatibonza-Garcia
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Sophia C Mylonakis
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Angeliki Katsarou
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Ornella Verrastro
- Department of Gastroenterology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Georgios Markakis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens, Greece
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Georgios Papatheodoridis
- Department of Gastroenterology, Medical School of National and Kapodistrian University of Athens, General Hospital of Athens "Laiko", Athens, Greece
| | - Geltrude Mingrone
- Department of Gastroenterology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
| | - Christos S Mantzoros
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Boston VA Healthcare System, Boston, MA 02130, United States of America
| |
Collapse
|
10
|
Kokkorakis M, Boutari C, Katsiki N, Mantzoros CS. From non-alcoholic fatty liver disease (NAFLD) to steatotic liver disease (SLD): an ongoing journey towards refining the terminology for this prevalent metabolic condition and unmet clinical need. Metabolism 2023; 147:155664. [PMID: 37517792 DOI: 10.1016/j.metabol.2023.155664] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Affiliation(s)
- Michail Kokkorakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Chrysoula Boutari
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
11
|
Koureta E, Cholongitas E. Combination therapies in nonalcoholic fatty liver disease using antidiabetic and disease-specific drugs. Ann Gastroenterol 2023; 36:378-391. [PMID: 37396007 PMCID: PMC10304532 DOI: 10.20524/aog.2023.0806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/19/2023] [Indexed: 07/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common diseases in the world, affecting approximately one fourth of the worldwide population. Glucose metabolism dysregulation and type 2 diabetes mellitus (T2DM), as part of the metabolic syndrome, are important factors implicated in the pathogenesis and progression of NAFLD to nonalcoholic steatohepatitis (NASH) and cirrhosis. Although a great deal of research has already been conducted regarding possible therapeutic medications for NAFLD/NASH, no drugs have been approved until now. Combination therapies in NAFLD seem to represent an attractive approach concerning treatment of the disease, as multiple pathophysiologic pathways contribute to the development and advance of NAFLD. In this review we discuss the impact of combining antidiabetic drugs, focusing on pioglitazone, sodium glucose cotransporter 2 inhibitors and glucagon-like peptide-1 receptor agonists. We also include data from the literature concerning combinations of newer "NAFLD-specific" drugs.
Collapse
Affiliation(s)
- Evgenia Koureta
- First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Cholongitas
- First Department of Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Portincasa P. NAFLD, MAFLD, and beyond: one or several acronyms for better comprehension and patient care. Intern Emerg Med 2023; 18:993-1006. [PMID: 36807050 PMCID: PMC10326150 DOI: 10.1007/s11739-023-03203-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 02/19/2023]
Abstract
The term non-alcoholic fatty liver disease (NAFLD) has rapidly become the most common type of chronic liver disease. NAFLD points to excessive hepatic fat storage and no evidence of secondary hepatic fat accumulation in patients with "no or little alcohol consumption". Both the etiology and pathogenesis of NAFLD are largely unknown, and a definitive therapy is lacking. Since NAFLD is very often and closely associated with metabolic dysfunctions, a consensus process is ongoing to shift the acronym NAFLD to MAFLD, i.e., metabolic-associated fatty liver disease. The change in terminology is likely to improve the classification of affected individuals, the disease awareness, the comprehension of the terminology and pathophysiological aspects involved, and the choice of more personalized therapeutic approaches while avoiding the intrinsic stigmatization due to the term "non-alcoholic". Even more recently, other sub-classifications have been proposed to concentrate the heterogeneous causes of fatty liver disease under one umbrella. While awaiting additional validation studies in this field, we discuss the main reasons underlying this important shift of paradigm.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica "A. Murri", Department of Preventive and Regenerative Medicine and Ionian Area (DiMePrev-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
13
|
Valenzuela-Vallejo L, Sanoudou D, Mantzoros CS. Precision Medicine in Fatty Liver Disease/Non-Alcoholic Fatty Liver Disease. J Pers Med 2023; 13:830. [PMID: 37241000 PMCID: PMC10224312 DOI: 10.3390/jpm13050830] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease, and is related to fatal and non-fatal liver, metabolic, and cardiovascular complications. Its non-invasive diagnosis and effective treatment remain an unmet clinical need. NAFLD is a heterogeneous disease that is most commonly present in the context of metabolic syndrome and obesity, but not uncommonly, may also be present without metabolic abnormalities and in subjects with normal body mass index. Therefore, a more specific pathophysiology-based subcategorization of fatty liver disease (FLD) is needed to better understand, diagnose, and treat patients with FLD. A precision medicine approach for FLD is expected to improve patient care, decrease long-term disease outcomes, and develop better-targeted, more effective treatments. We present herein a precision medicine approach for FLD based on our recently proposed subcategorization, which includes the metabolic-associated FLD (MAFLD) (i.e., obesity-associated FLD (OAFLD), sarcopenia-associated FLD (SAFLD, and lipodystrophy-associated FLD (LAFLD)), genetics-associated FLD (GAFLD), FLD of multiple/unknown causes (XAFLD), and combined causes of FLD (CAFLD) as well as advanced stage fibrotic FLD (FAFLD) and end-stage FLD (ESFLD) subcategories. These and other related advances, as a whole, are expected to enable not only improved patient care, quality of life, and long-term disease outcomes, but also a considerable reduction in healthcare system costs associated with FLD, along with more options for better-targeted, more effective treatments in the near future.
Collapse
Affiliation(s)
- Laura Valenzuela-Vallejo
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4(th) Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Christos S. Mantzoros
- Department of Medicine, Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA;
- Department of Medicine, Boston VA Healthcare System, Boston, MA 02130, USA
| |
Collapse
|
14
|
Ntona S, Papaefthymiou A, Kountouras J, Gialamprinou D, Kotronis G, Boziki M, Polyzos SA, Tzitiridou M, Chatzopoulos D, Thavayogarajah T, Gkolia I, Ntonas G, Vardaka E, Doulberis M. Impact of nonalcoholic fatty liver disease-related metabolic state on depression. Neurochem Int 2023; 163:105484. [PMID: 36634820 DOI: 10.1016/j.neuint.2023.105484] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/15/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), also recently referred as metabolic (dysfunction)-associated fatty liver disease (MAFLD), is characterized by hepatocyte steatosis in the setting of metabolic risk conditions and in the absence of an underlying precursor, for instance alcohol consumption, hepatotropic viruses and hepatotoxic drugs. A possible association between NAFLD and depression has been proposed, owing to intersecting pathophysiological pathways. This narrative review aimed to summarize the current evidence that illustrate the potential pathophysiological and clinical linkage between NAFLD-related metabolic state and depression. Prefrontal cortex lesions are suggested to be a consequence of liver steatosis-associated systematic hyperinflammatory state, a phenomenon also occurring in depression. In addition, depressive symptoms are present in neurotransmitter imbalances. These abnormalities seem to be correlated with NAFLD/MAFLD, in terms of insulin resistance (IR), ammonia and gut dysbiosis' impact on serotonin, dopamine, noradrenaline levels and gamma aminobutyric acid receptors. Furthermore, reduced levels of nesfatin-1 and copine-6-associated BDNF (brain-derived neurotrophic factor) levels have been considered as a probable link between NAFLD and depression. Regarding NAFLD-related gut dysbiosis, it stimulates mediators including lipopolysaccharides, short-chain fatty acids and bile acids, which play significant role in depression. Finally, western diet and IR, which are mainstay components of NAFLD/MAFLD, are, also, substantiated to affect neurotransmitters in hippocampus and produce neurotoxic lipids that contribute to neurologic dysfunction, and thus trigger emotional disturbances, mainly depressive symptoms.
Collapse
Affiliation(s)
- Smaragda Ntona
- Alexandrovska University Hospital, Medical University Sofia, 1431, Sofia, Bulgaria
| | - Apostolis Papaefthymiou
- Department of Gastroenterology, University Hospital of Larisa, 41110, Mezourlo, Larissa, Thessaly, Greece; First Laboratory of Pharmacology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Macedonia, Greece; Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece
| | - Jannis Kountouras
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece.
| | - Dimitra Gialamprinou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Second Neonatal Department and NICU, Papageorgiou General Hospital, Aristotle University of Thessaloniki, 56403, Thessaloniki, Macedonia, Greece
| | - Georgios Kotronis
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Department of Internal Medicine, General Hospital Aghios Pavlos of Thessaloniki, 55134, Thessaloniki, Macedonia, Greece
| | - Marina Boziki
- Second Neurological Department, Aristotle University of Thessaloniki, AHEPA University General Hospital of Thessaloniki, Thessaloniki, 54636, Macedonia, Greece
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, Aristotle University of Thessaloniki, 54124, Thessaloniki, Macedonia, Greece
| | - Maria Tzitiridou
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; School of Healthcare Sciences, Midwifery Department, University of West Macedonia, Koila, Kozani, 50100, Macedonia, Greece
| | - Dimitrios Chatzopoulos
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece
| | - Tharshika Thavayogarajah
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091, Zurich, Switzerland
| | - Ioanna Gkolia
- Psychiatric Hospital of Thessaloniki, 54634, Stavroupoli, Macedonia, Greece
| | - Georgios Ntonas
- Department of Anesthesiology, Agios Dimitrios General Hospital, 54635, Thessaloniki, Macedonia, Greece
| | - Elisabeth Vardaka
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400, Thessaloniki, Greece
| | - Michael Doulberis
- Second Medical Clinic, School of Medicine, Aristotle University of Thessaloniki, Ippokration Hospital, 54642, Thessaloniki, Macedonia, Greece; Department of Gastroenterology and Hepatology, University of Zurich, 8091, Zurich, Switzerland; Division of Gastroenterology and Hepatology, Medical University Department, Kantonsspital Aarau, 5001, Aarau, Switzerland
| |
Collapse
|
15
|
Role of B Lymphocytes in the Pathogenesis of NAFLD: A 2022 Update. Int J Mol Sci 2022; 23:ijms232012376. [PMID: 36293233 PMCID: PMC9603875 DOI: 10.3390/ijms232012376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Non-alcoholic fatty liver disease and its related complications are becoming one of the most important health problems globally. The liver functions as both a metabolic and an immune organ. The crosstalk between hepatocytes and intrahepatic immune cells plays a key role in coordinating a dual function of the liver in terms of the protection of the host from antigenic overload as a result of receiving nutrients and gut microbiota antigenic stimulation via facilitating immunologic tolerance. B cells are the most abundant lymphocytes in the liver. The crucial role of intrahepatic B cells in energy metabolism under different immune conditions is now emerging in the literature. The accumulating evidence has demonstrated that the antibodies and cytokines produced by B cells in the microenvironment play key and distinct roles in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Herein, we have aimed to consolidate and update the current knowledge about the pathophysiological roles of B cells as well as the underlying mechanisms in energy metabolism. Understanding how B cells can exacerbate and suppress liver damage by exploiting the antibodies and cytokines they produce will be of great importance for designing B-cell targeting therapies to treat various liver diseases.
Collapse
|
16
|
Ran X, Hu G, He F, Li K, Li F, Xu D, Liu J, Fu S. Phytic Acid Improves Hepatic Steatosis, Inflammation, and Oxidative Stress in High-Fat Diet (HFD)-Fed Mice by Modulating the Gut-Liver Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:11401-11411. [PMID: 36040330 DOI: 10.1021/acs.jafc.2c04406] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) induced by obesity is a grave threat to human health. Phytic acid (PA) is a natural compound found in high-fiber diets, such as soybeans. This study investigated the effects and mechanisms of PA on obesity, hepatic lipid metabolism, and gut-liver axis homeostasis in high-fat diet (HFD)-fed mice. PA was observed to significantly inhibit obesity and alleviate liver steatosis in mice. PA improved HFD-induced liver inflammation, oxidative stress and fibrosis. Moreover, PA improved HFD-induced colonic inflammation, gut barrier damage and systemic inflammation in mice. Furthermore, PA effectively ameliorated the decreased diversity and gut microbiota composition in HFD-fed mice. Additionally, PA decreased the abundance of harmful bacteria Proteobacteria and Desulfovibrionaceae and increased the abundance of probiotic bacteria Muribaculaceae and Lachnospiraceae. Thus, PA is effective in restoring the homeostasis of the gut-liver axis. It further provides a theoretical basis for the prevention and treatment of NAFLD in patients with obesity by the rational intake of foods containing PA.
Collapse
Affiliation(s)
- Xin Ran
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130012, China
| | - Guiqiu Hu
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130012, China
| | - Fuding He
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130012, China
| | - Kefei Li
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130012, China
| | - Feng Li
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130012, China
| | - Dianwen Xu
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130012, China
| | - Juxiong Liu
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130012, China
| | - Shoupeng Fu
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
17
|
Vachliotis ID, Anastasilakis AD, Goulas A, Goulis DG, Polyzos SA. Nonalcoholic fatty liver disease and osteoporosis: A potential association with therapeutic implications. Diabetes Obes Metab 2022; 24:1702-1720. [PMID: 35589613 DOI: 10.1111/dom.14774] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/01/2022] [Accepted: 05/17/2022] [Indexed: 11/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and osteoporosis are two highly prevalent metabolic diseases. Increasing experimental evidence supports a pathophysiological link between NAFLD and osteoporosis. A key feature could be chronic, low-grade inflammation, which characterizes NAFLD and possibly affects bone metabolism. In this context, several factors, including but not limited to receptor activator of nuclear factor kappa-B ligand, osteoprotegerin, osteopontin and osteocalcin, may serve as mediators. In the clinical setting, most but not all epidemiological evidence indicates that NAFLD is associated with lower bone mineral density or osteoporosis in adults. Although an association between NAFLD and osteoporosis has not yet been established, and thus remains speculative, pharmacological considerations already exist. Some of the current and emerging pharmacological options for NAFLD have shown possible anti-osteoporotic properties (eg, vitamin E, obeticholic acid, semaglutide), while others (eg, pioglitazone, canagliflozin) have been associated with increased risk of fractures and may be avoided in patients with NAFLD and concomitant osteoporosis, especially those at high fracture risk. Conversely, some anti-osteoporotic medications (denosumab) might benefit NAFLD, while others (raloxifene) might adversely affect it and, consequently, may be avoided in patients with osteoporosis and NAFLD. If an association between NAFLD and osteoporosis is established, a medication that could target both diseases would be a great advancement. This review summarizes the main experimental and clinical evidence on the potential association between NAFLD and osteoporosis and focuses on treatment considerations derived from this potential association.
Collapse
Affiliation(s)
- Ilias D Vachliotis
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Department of Endocrinology, 424 General Military Hospital, Thessaloniki, Greece
| | | | - Antonis Goulas
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
18
|
Combination Therapies for Nonalcoholic Fatty Liver Disease. J Pers Med 2022; 12:jpm12071166. [PMID: 35887662 PMCID: PMC9322793 DOI: 10.3390/jpm12071166] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered a highly prevalent disease associated with various co-morbidities that lead to socioeconomic burden. Despite large-scale investigation, no pharmacological treatment has been approved specifically for NAFLD to date. Lifestyle modifications and diet are regarded as highly beneficial for the management of NAFLD, albeit with poor compliance, thus rendering pharmacological treatment highly important. Based on the current failure to discover a “magic bullet” to treat all patients with NAFLD and considering the multifaceted pathophysiology of the disease, combination therapies may be considered to be a rational alternative approach. In this regard, several drug categories have been considered, including, but not limited to, lipid-lowering, anti-hypertensive, glucose-lowering, anti-obesity, anti-oxidant, anti-inflammatory and anti-fibrotic medications. The aim of this review is, in addition to summarizing some of the multiple factors contributing to the pathophysiology of NAFLD, to focus on the efficacy of pharmacological combinations on the management of NAFLD. This may provide evidence for a more personalized treatment of patients with NAFLD in the future.
Collapse
|
19
|
Muzurović E, Peng CCH, Belanger MJ, Sanoudou D, Mikhailidis DP, Mantzoros CS. Nonalcoholic Fatty Liver Disease and Cardiovascular Disease: a Review of Shared Cardiometabolic Risk Factors. Hypertension 2022; 79:1319-1326. [PMID: 35465684 DOI: 10.1161/hypertensionaha.122.17982] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) is rising. NAFLD/nonalcoholic steatohepatitis (NASH) is associated not only with hepatic morbidity and mortality but also with an increased cardiovascular risk. NAFLD and cardiovascular disease (CVD) share several risk factors, such as obesity, metabolic syndrome, hypertension, dyslipidemia, type 2 diabetes, and chronic kidney disease. This review summarizes the evidence linking cardiometabolic risk factors and NAFLD in the context of risk for CVD. The cause of NAFLD/NASH is complex, involving a range of factors from genetics to lifestyle and energy balance. Genetically driven high liver fat content does not appear to be causally associated with increased CVD risk. In contrast, metabolic dysfunction not only predisposes to liver pathology but also leads to a significantly higher CVD risk. Given that NAFLD pathophysiology is influenced by multiple factors, each patient is unique as to their risk of developing CVD and liver pathology. At the same time, the rising burden of NAFLD/NASH is closely linked with the global increase in metabolic disorders, including obesity and type 2 diabetes. Therefore, both personalized therapeutic approaches that recognize individual pathophysiology, as well as public health policies that address the root causes of cardiometabolic risk factors for NAFLD may be needed to effectively address the NAFLD/NASH epidemic.
Collapse
Affiliation(s)
- Emir Muzurović
- Department of Internal Medicine, Endocrinology Section, Clinical Centre of Montenegro, Podgorica (E.M.).,Faculty of Medicine, University of Montenegro, Podgorica, Montenegro (E.M.)
| | - Carol Chiung-Hui Peng
- Section of Endocrinology, Diabetes, Nutrition & Weight Management, Boston University School of Medicine, Boston, MA (C.C.-H.P.)
| | | | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4 Department of Internal Medicine, Attikon Hospital, Medical School, National and Kapodistrian University of Athens, Greece (D.S.).,Biomedical Research Foundation of the Academy of Athens, Greece (D.S.)
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London, Medical School, University College London (UCL), United Kingdom (D.P.M.).,Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai (D.P.M.)
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (C.S.M.)
| |
Collapse
|
20
|
Petrov MS, Taylor R. Intra-pancreatic fat deposition: bringing hidden fat to the fore. Nat Rev Gastroenterol Hepatol 2022; 19:153-168. [PMID: 34880411 DOI: 10.1038/s41575-021-00551-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/04/2021] [Indexed: 02/07/2023]
Abstract
Development of advanced modalities for detection of fat within the pancreas has transformed understanding of the role of intra-pancreatic fat deposition (IPFD) in health and disease. There is now strong evidence for the presence of minimal (but not negligible) IPFD in healthy human pancreas. Diffuse excess IPFD, or fatty pancreas disease (FPD), is more frequent than type 2 diabetes mellitus (T2DM) (the most common disease of the endocrine pancreas) and acute pancreatitis (the most common disease of the exocrine pancreas) combined. FPD is not strictly a function of high BMI; it can result from the excess deposition of fat in the islets of Langerhans, acinar cells, inter-lobular stroma, acinar-to-adipocyte trans-differentiation or replacement of apoptotic acinar cells. This process leads to a wide array of diseases characterized by excess IPFD, including but not limited to acute pancreatitis, chronic pancreatitis, pancreatic cancer, T2DM, diabetes of the exocrine pancreas. There is ample evidence for FPD being potentially reversible. Weight loss-induced decrease of intra-pancreatic fat is tightly associated with remission of T2DM and its re-deposition with recurrence of the disease. Reversing FPD will open up opportunities for preventing or intercepting progression of major diseases of the exocrine pancreas in the future.
Collapse
Affiliation(s)
- Maxim S Petrov
- School of Medicine, University of Auckland, Auckland, New Zealand.
| | - Roy Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
21
|
Devi J, Raees A, Butt AS. Redefining non-alcoholic fatty liver disease to metabolic associated fatty liver disease: Is this plausible? World J Hepatol 2022; 14:158-167. [PMID: 35126845 PMCID: PMC8790389 DOI: 10.4254/wjh.v14.i1.158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/17/2021] [Accepted: 12/22/2021] [Indexed: 02/06/2023] Open
Abstract
Recently, a single letter change has taken the world by storm. A group of experts have developed a consensus to upgrade the term non-alcoholic fatty liver disease (NAFLD) to metabolic associated fatty liver disease (MAFLD), suggesting that MAFLD would more accurately reflect not only the disease pathogenesis but would also help in patient stratification for management with NAFLD. However, the difference of opinion exists, which has made the NAFLD vs MAFLD debate the current talk of the town. This review will focus on the plausibility and implications of redefining NAFLD as MAFLD.
Collapse
Affiliation(s)
- Jalpa Devi
- Department of Gastroenterology, Liaquat University of Medical and Health Sciences, Jamshoro 74800, Pakistan
| | - Aimun Raees
- Department of Gastroenterology, Aga Khan University Hospital, Karachi 74800, Pakistan
| | - Amna Subhan Butt
- Department of Gastroenterology, Aga Khan University Hospital, Karachi 74800, Pakistan.
| |
Collapse
|
22
|
Muzurović EM, Volčanšek Š, Tomšić KZ, Janež A, Mikhailidis DP, Rizzo M, Mantzoros CS. Glucagon-Like Peptide-1 Receptor Agonists and Dual Glucose-Dependent Insulinotropic Polypeptide/Glucagon-Like Peptide-1 Receptor Agonists in the Treatment of Obesity/Metabolic Syndrome, Prediabetes/Diabetes and Non-Alcoholic Fatty Liver Disease-Current Evidence. J Cardiovasc Pharmacol Ther 2022; 27:10742484221146371. [PMID: 36546652 DOI: 10.1177/10742484221146371] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The obesity pandemic is accompanied by increased risk of developing metabolic syndrome (MetS) and related conditions: non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH), type 2 diabetes mellitus (T2DM) and cardiovascular (CV) disease (CVD). Lifestyle, as well as an imbalance of energy intake/expenditure, genetic predisposition, and epigenetics could lead to a dysmetabolic milieu, which is the cornerstone for the development of cardiometabolic complications. Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) and dual glucose-dependent insulinotropic polypeptide (GIP)/GLP-1 RAs promote positive effects on most components of the "cardiometabolic continuum" and consequently help reduce the need for polypharmacy. In this review, we highlight the main pathophysiological mechanisms and risk factors (RFs), that could be controlled by GLP-1 and dual GIP/GLP-1 RAs independently or through synergism or differences in their mode of action. We also address the evidence on the use of GLP-1 and dual GIP/GLP-1 RAs in the treatment of obesity, MetS and its related conditions (prediabetes, T2DM and NAFLD/NASH). In conclusion, GLP-1 RAs have already been established for the treatment of T2DM, obesity and cardioprotection in T2DM patients, while dual GIP/GLP-1 RAs appear to have the potential to possibly surpass them for the same indications. However, their use in the prevention of T2DM and the treatment of complex cardiometabolic metabolic diseases, such as NAFLD/NASH or other metabolic disorders, would benefit from more evidence and a thorough clinical patient-centered approach. There is a need to identify those patients in whom the metabolic component predominates, and whether the benefits outweigh any potential harm.
Collapse
Affiliation(s)
- Emir M Muzurović
- Department of Internal Medicine, Endocrinology Section, Clinical Center of Montenegro, Podgorica, Montenegro.,Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Špela Volčanšek
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia.,Medical Faculty Ljubljana, Ljubljana, Slovenia
| | - Karin Zibar Tomšić
- Department of Endocrinology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Andrej Janež
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia.,Medical Faculty Ljubljana, Ljubljana, Slovenia
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, United Kingdom.,Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Manfredi Rizzo
- Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Verdú E, Homs J, Boadas-Vaello P. Physiological Changes and Pathological Pain Associated with Sedentary Lifestyle-Induced Body Systems Fat Accumulation and Their Modulation by Physical Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:13333. [PMID: 34948944 PMCID: PMC8705491 DOI: 10.3390/ijerph182413333] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
A sedentary lifestyle is associated with overweight/obesity, which involves excessive fat body accumulation, triggering structural and functional changes in tissues, organs, and body systems. Research shows that this fat accumulation is responsible for several comorbidities, including cardiovascular, gastrointestinal, and metabolic dysfunctions, as well as pathological pain behaviors. These health concerns are related to the crosstalk between adipose tissue and body systems, leading to pathophysiological changes to the latter. To deal with these health issues, it has been suggested that physical exercise may reverse part of these obesity-related pathologies by modulating the cross talk between the adipose tissue and body systems. In this context, this review was carried out to provide knowledge about (i) the structural and functional changes in tissues, organs, and body systems from accumulation of fat in obesity, emphasizing the crosstalk between fat and body tissues; (ii) the crosstalk between fat and body tissues triggering pain; and (iii) the effects of physical exercise on body tissues and organs in obese and non-obese subjects, and their impact on pathological pain. This information may help one to better understand this crosstalk and the factors involved, and it could be useful in designing more specific training interventions (according to the nature of the comorbidity).
Collapse
Affiliation(s)
- Enrique Verdú
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| | - Judit Homs
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
- Department of Physical Therapy, EUSES-University of Girona, 17190 Salt, Spain
| | - Pere Boadas-Vaello
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| |
Collapse
|
24
|
Pal P, Palui R, Ray S. Heterogeneity of non-alcoholic fatty liver disease: Implications for clinical practice and research activity. World J Hepatol 2021; 13:1584-1610. [PMID: 34904031 PMCID: PMC8637673 DOI: 10.4254/wjh.v13.i11.1584] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/29/2021] [Accepted: 10/14/2021] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a heterogeneous condition with a wide spectrum of clinical presentations and natural history and disease severity. There is also substantial inter-individual variation and variable response to a different therapy. This heterogeneity of NAFLD is in turn influenced by various factors primarily demographic/dietary factors, metabolic status, gut microbiome, genetic predisposition together with epigenetic factors. The differential impact of these factors over a variable period of time influences the clinical phenotype and natural history. Failure to address heterogeneity partly explains the sub-optimal response to current and emerging therapies for fatty liver disease. Consequently, leading experts across the globe have recently suggested a change in nomenclature of NAFLD to metabolic-associated fatty liver disease (MAFLD) which can better reflect current knowledge of heterogeneity and does not exclude concomitant factors for fatty liver disease (e.g. alcohol, viral hepatitis, etc.). Precise identification of disease phenotypes is likely to facilitate clinical trial recruitment and expedite translational research for the development of novel and effective therapies for NAFLD/MAFLD.
Collapse
Affiliation(s)
- Partha Pal
- Department of Medical Gastroenterology, Asian Institute of Gastroenterology, Hyderabad 500082, India
| | - Rajan Palui
- Department of Endocrinology, The Mission Hospital, Durgapur 713212, West Bengal, India
| | - Sayantan Ray
- Department of Endocrinology, Jagannath Gupta Institute of Medical Sciences and Hospital, Kolkata 700137, West Bengal, India
- Diabetes and Endocrinology, Apollo Clinic, Ballygunge, Kolkata 700019, West Bengal, India.
| |
Collapse
|
25
|
Makri ES, Goulas A, Polyzos SA. Sodium-glucose co-transporter 2 inhibitors in nonalcoholic fatty liver disease. Eur J Pharmacol 2021; 907:174272. [PMID: 34147478 DOI: 10.1016/j.ejphar.2021.174272] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/03/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is considered the most prevalent chronic hepatic disease, as it has been estimated that one of four individuals in the general population has been affected by NAFLD. The evolution of the referred entity, which includes nonalcoholic steatohepatitis (NASH) and hepatic fibrosis, may have crucial and even fatal consequences, leading to cirrhosis and hepatocellular carcinoma. Although NAFLD has also been linked with cardiovascular and renal diseases, and all-cause mortality increment, pharmacological therapy is as yet an unfulfilled demand. Since NAFLD is closely associated with type 2 diabetes mellitus (T2DM), a variety of anti-diabetic drugs have been investigated for their effectiveness towards NAFLD. Sodium-glucose co-transporter 2 inhibitors (SGLT-2i) improve blood glucose levels through increasing renal glucose excretion and they are recommended as one of standard therapeutic categories for T2DM patients. Based on preclinical animal studies, SGLT-2i have shown a beneficial effect on NAFLD, inducing histologically proven amelioration of hepatic steatosis, inflammation and fibrosis. Promising data have been also derived by clinical trials, which have indicated a potentially beneficial effect of SGLT-2i on NAFLD, at least in terms of liver function tests and imaging. Thus, it is not strange that there are many ongoing trials on the effect of various SGLT-2i in NAFLD. In conclusion, current evidence concerning the effect of SGLT-2i on NAFLD is encouraging; however, data from ongoing clinical trials with histological endpoints are awaited.
Collapse
Affiliation(s)
- Evangelia S Makri
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Campus of Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| | - Antonis Goulas
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Campus of Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Campus of Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece.
| |
Collapse
|
26
|
Boutari C, Polyzos SA, Mantzoros CS. Addressing the epidemic of fatty liver disease: A call to action, a call to collaboration, a call to moving the field forward. Metabolism 2021; 122:154781. [PMID: 33901501 DOI: 10.1016/j.metabol.2021.154781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Affiliation(s)
- Chrysoula Boutari
- Second Propedeutic Department of Internal Medicine, School of Medicine, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
27
|
Cholico GN, Fling RR, Zacharewski NA, Fader KA, Nault R, Zacharewski TR. Thioesterase induction by 2,3,7,8-tetrachlorodibenzo-p-dioxin results in a futile cycle that inhibits hepatic β-oxidation. Sci Rep 2021; 11:15689. [PMID: 34344994 PMCID: PMC8333094 DOI: 10.1038/s41598-021-95214-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), a persistent environmental contaminant, induces steatosis by increasing hepatic uptake of dietary and mobilized peripheral fats, inhibiting lipoprotein export, and repressing β-oxidation. In this study, the mechanism of β-oxidation inhibition was investigated by testing the hypothesis that TCDD dose-dependently repressed straight-chain fatty acid oxidation gene expression in mice following oral gavage every 4 days for 28 days. Untargeted metabolomic analysis revealed a dose-dependent decrease in hepatic acyl-CoA levels, while octenoyl-CoA and dicarboxylic acid levels increased. TCDD also dose-dependently repressed the hepatic gene expression associated with triacylglycerol and cholesterol ester hydrolysis, fatty acid binding proteins, fatty acid activation, and 3-ketoacyl-CoA thiolysis while inducing acyl-CoA hydrolysis. Moreover, octenoyl-CoA blocked the hydration of crotonyl-CoA suggesting short chain enoyl-CoA hydratase (ECHS1) activity was inhibited. Collectively, the integration of metabolomics and RNA-seq data suggested TCDD induced a futile cycle of fatty acid activation and acyl-CoA hydrolysis resulting in incomplete β-oxidation, and the accumulation octenoyl-CoA levels that inhibited the activity of short chain enoyl-CoA hydratase (ECHS1).
Collapse
Affiliation(s)
- Giovan N Cholico
- Department of Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Russell R Fling
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
- Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Nicholas A Zacharewski
- Department of Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
| | - Kelly A Fader
- Department of Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Rance Nault
- Department of Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Timothy R Zacharewski
- Department of Biochemistry and Molecular Biology, Michigan State University, Biochemistry Building, 603 Wilson Road, East Lansing, MI, 48824, USA.
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
28
|
Xue C, Li Y, Lv H, Zhang L, Bi C, Dong N, Shan A, Wang J. Oleanolic Acid Targets the Gut-Liver Axis to Alleviate Metabolic Disorders and Hepatic Steatosis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:7884-7897. [PMID: 34251802 DOI: 10.1021/acs.jafc.1c02257] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
This study investigated the effects of oleanolic acid (OA) on hepatic lipid metabolism and gut-liver axis homeostasis in an obesity-related non-alcoholic fatty liver disease (NAFLD) nutritional animal model and explored possible molecular mechanisms behind its effects. The results revealed that OA ameliorated the development of metabolic disorders, insulin resistance, and hepatic steatosis in obese rats. Meanwhile, OA restored high-fat-diet (HFD)-induced intestinal barrier dysfunction and endotoxin-mediated induction of toll-like-receptor-4-related pathways, subsequently inhibiting endotoxemia and systemic inflammation and balancing the homeostasis of the gut-liver axis. OA also reshaped the composition of the gut microbiota of HFD-fed rats by reducing the Firmicutes/Bacteroidetes ratio and increasing the abundance of butyrate-producing bacteria. Our results support the applicability of OA as a treatment for obesity-related NAFLD through its anti-inflammatory, antioxidant, and prebiotic integration responses mediated by the gut-liver axis.
Collapse
Affiliation(s)
- Chenyu Xue
- Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Ying Li
- Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Hao Lv
- Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Lei Zhang
- Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Chongpeng Bi
- Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Na Dong
- Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Anshan Shan
- Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, Heilongjiang 150030, People's Republic of China
| | - Jiali Wang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| |
Collapse
|
29
|
Lim S, Kim JW, Targher G. Links between metabolic syndrome and metabolic dysfunction-associated fatty liver disease. Trends Endocrinol Metab 2021; 32:500-514. [PMID: 33975804 DOI: 10.1016/j.tem.2021.04.008] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 02/08/2023]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a chronic condition characterized by hepatic fat accumulation combined with underlying metabolic dysregulation. Having evolved from the previous term of nonalcoholic fatty liver disease (NAFLD), the term MAFLD more closely implicates the presence of overweight/obesity, type 2 diabetes, or metabolic dysregulation as essential pathogenic factors, leading to better identification of individuals with this metabolic liver disease. Low-grade inflammation, increased oxidative stress, mitochondrial dysfunction, and intestinal dysbiosis are also involved in its pathogenesis. MAFLD is not only associated with liver-related complications, but also with adverse cardiometabolic outcomes. Further studies are needed to assess whether the newly proposed definition of MAFLD is more accurate than the NAFLD in predicting the adverse liver-related and extrahepatic outcomes.
Collapse
Affiliation(s)
- Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seoul, Korea.
| | - Jin-Wook Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seoul, Korea
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Verona, Verona, Italy.
| |
Collapse
|
30
|
Natural products and analogs as preventive agents for metabolic syndrome via peroxisome proliferator-activated receptors: An overview. Eur J Med Chem 2021; 221:113535. [PMID: 33992930 DOI: 10.1016/j.ejmech.2021.113535] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/19/2021] [Accepted: 05/01/2021] [Indexed: 12/20/2022]
Abstract
Natural products and synthetic analogs have drawn much attention as potential therapeutical drugs to treat metabolic syndrome. We reviewed the underlying mechanisms of 32 natural products and analogs with potential pharmacological effects in vitro, and especially in rodent models and/or patients, that usually act on the PPAR pathway, along with other molecular targets. Recent outstanding total syntheses or semisyntheses of these lead compounds are stated. In general, they can activate the transcriptional activity of PPARα, PPARγ, PPARα/γ, PPARβ/δ, PPARα/δ, PPARγ/δ and panPPAR as weak, partial agonists or selective PPARγ modulators (SPPARγM), which may be useful for managing obesity, type 2 diabetes (T2D), dyslipidemia and non-fatty liver disease (NAFLD). Terpenoids is the largest group of compounds that act as potential modulators on PPARs and are comprised from small lipophilic cannabinoids to lipophilic pentacyclic triterpenes and polar saponins. Shikimates-phenylpropanoids include polar heterocyclic flavonoids and phenolic compounds containing at least one C3-C6 unit and usually a double bond on the propyl chain. Quercetin (19), resveratrol (24) and curcumin (27), stand out from this group for exhibiting beneficial effects on patients. Alkaloids, the minor group of potential modulators on PPARs, include berberine (30), which has been widely explored in preclinical and clinical studies for its potential beneficial effects on T2D and dyslipidemia. However, large-scale clinical trials may be warranted for the promising compounds.
Collapse
|
31
|
Effects of Ethanol Feeding in Early-Stage NAFLD Mice Induced by Western Diet. LIVERS 2021. [DOI: 10.3390/livers1010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: The prevalence of metabolic liver diseases is increasing and approved pharmacological treatments are still missing. Many animal models of nonalcoholic fatty liver disease (NAFLD) show a full spectrum of fibrosis, inflammation and steatosis, which does not reflect the human situation since only up to one third of the patients develop fibrosis and nonalcoholic steatohepatitis (NASH). Methods: Seven week old C57Bl/J mice were treated with ethanol, Western diet (WD) or both. The animals’ liver phenotypes were determined through histology, immunohistochemistry, Western blotting, hepatic triglyceride content and gene expression levels. In a human cohort of 80 patients stratified by current alcohol misuse and body mass index, liver histology and gene expression analysis were performed. Results: WD diet and ethanol-treated animals showed severe steatosis, with high hepatic triglyceride content and upregulation of fatty acid synthesis. Mild fibrosis was revealed using Sirius-red stains and gene expression levels of collagen. Inflammation was detected using histology, immunohistochemistry and upregulation of proinflammatory genes. The human cohort of obese drinkers showed similar upregulation in genes related to steatosis, fibrosis and inflammation. Conclusions: We provide a novel murine model for early-stage fatty liver disease suitable for drug testing and investigation of pathophysiology.
Collapse
|
32
|
Makri E, Goulas A, Polyzos SA. Epidemiology, Pathogenesis, Diagnosis and Emerging Treatment of Nonalcoholic Fatty Liver Disease. Arch Med Res 2020; 52:25-37. [PMID: 33334622 DOI: 10.1016/j.arcmed.2020.11.010] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/26/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, with an estimated rising prevalence, in concert with the epidemics of obesity and type 2 diabetes. The pathogenesis of NAFLD is not fully elucidated. Besides weight gain and insulin resistance, many other factors seem to contribute, including adipokines, gut microbiota and genetic predisposition. The disease starts as hepatic steatosis, which may proceed to nonalcoholic steatohepatitis (NASH); if fibrosis is added, the risk of cirrhosis and/or hepatocellular carcinoma is augmented. Liver biopsy is considered the gold standard for the diagnosis and staging of NAFLD; the early use of reliable and easily applied diagnostic tools, such as noninvasive biomarkers, is needed to identify patients at different-preferably early-stages of disease however. Whilst lifestyle modification is the first step to manage NAFLD, there is poor compliance, leading to the need of drug therapy. Accordingly, a variety of medications is under investigation. Given the multifaceted pathophysiology of NAFLD, probably, a combination of approaches in an individualized basis may be a more appropriate management. This review summarizes evidence on the epidemiology, pathogenesis, diagnosis and treatment of NAFLD.
Collapse
MESH Headings
- Biomarkers/analysis
- Biomarkers/blood
- Carcinoma, Hepatocellular/complications
- Carcinoma, Hepatocellular/diagnosis
- Carcinoma, Hepatocellular/epidemiology
- Carcinoma, Hepatocellular/therapy
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/diagnosis
- Diabetes Mellitus, Type 2/epidemiology
- Diabetes Mellitus, Type 2/therapy
- Humans
- Life Style
- Liver Cirrhosis/diagnosis
- Liver Cirrhosis/epidemiology
- Liver Cirrhosis/etiology
- Liver Cirrhosis/therapy
- Liver Neoplasms/complications
- Liver Neoplasms/diagnosis
- Liver Neoplasms/epidemiology
- Liver Neoplasms/therapy
- Non-alcoholic Fatty Liver Disease/diagnosis
- Non-alcoholic Fatty Liver Disease/epidemiology
- Non-alcoholic Fatty Liver Disease/etiology
- Non-alcoholic Fatty Liver Disease/therapy
- Obesity/complications
- Obesity/diagnosis
- Obesity/epidemiology
- Obesity/therapy
- Prevalence
- Risk Factors
- Therapies, Investigational/methods
- Therapies, Investigational/trends
Collapse
Affiliation(s)
- Evangelia Makri
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Antonis Goulas
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
33
|
Mantovani A, Dalbeni A. NAFLD, MAFLD and DAFLD. Dig Liver Dis 2020; 52:1519-1520. [PMID: 33012657 DOI: 10.1016/j.dld.2020.09.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/08/2020] [Accepted: 09/16/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Alessandro Mantovani
- Section of Endocrinology, Diabetes and Metabolism, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy.
| | - Andrea Dalbeni
- Section of General Medicine, Hypertension and Liver Unit, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| |
Collapse
|
34
|
Polyzos SA, Kang ES, Tsochatzis EA, Kechagias S, Ekstedt M, Xanthakos S, Lonardo A, Mantovani A, Tilg H, Côté I, Grefhorst A, Greene MW, Araujo-Vilar D, Alisi A, Casanueva F, Mantzoros CS. Commentary: Nonalcoholic or metabolic dysfunction-associated fatty liver disease? The epidemic of the 21st century in search of the most appropriate name. Metabolism 2020; 113:154413. [PMID: 33164861 DOI: 10.1016/j.metabol.2020.154413] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eun Seok Kang
- Severance Hospital Diabetes Center, Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | - Stergios Kechagias
- Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Mattias Ekstedt
- Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Stavra Xanthakos
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Amedeo Lonardo
- Internal Medicine, Ospedale Civile di Baggiovara, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Alessandro Mantovani
- Section of Endocrinology, Diabetes and Metabolism, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Herbert Tilg
- Department of Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Austria
| | - Isabelle Côté
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Aldo Grefhorst
- Department of Experimental Vascular Medicine, Amsterdam University Medical Center, location AMC, Amsterdam, Netherlands
| | | | - David Araujo-Vilar
- Thyroid and Metabolic Diseases Unit (UETeM), Biomedical Research Institute (CIMUS/IDIS), Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, University of Santiago de Compostela, Spain
| | - Anna Alisi
- Research Unit of Molecular Genetics of Complex Phenotypes, Bambino Gesu' Children Hospital, IRCCS, Rome, Italy
| | - Felipe Casanueva
- Division of Endocrinology, Department of Medicine, Santiago de Compostela University (USC), Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago, Spain
| | - Christos S Mantzoros
- Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA; Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
35
|
Polyzos SA, Mousiolis A, Mintziori G, Goulis DG. Nonalcoholic fatty liver disease in males with low testosterone concentrations. Diabetes Metab Syndr 2020; 14:1571-1577. [PMID: 32947757 DOI: 10.1016/j.dsx.2020.07.049] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/18/2020] [Accepted: 07/25/2020] [Indexed: 12/11/2022]
Abstract
AIMS There are limited clinical data on the association between serum testosterone concentrations and nonalcoholic fatty liver disease (NAFLD) in men. The main aim of this study was to evaluate the association between testosterone concentrations and NAFLD in adult men, in terms of noninvasive indices of NAFLD and hepatic fibrosis. METHODS In this cross-sectional study, 98 men were recruited on an outpatient basis and were divided into low-testosterone (<12 nmol/l or <346 ng/dl, n = 37) or high-testosterone groups (≥12 nmol/l or ≥346 ng/dl, n = 61). Serum testosterone concentrations were measured by immuno-chemiluminescence. Hepatic steatosis index (HSI) and Triglyceride-to-HDL-C ratio (THR), as non-invasive indices of NAFLD, as well as AST-to-Platelet Ratio Index (APRI), fibrosis-4 index (FIB-4) and NAFLD fibrosis score (NFS), as non-invasive indices of hepatic fibrosis, were calculated based on standard formulas. RESULTS Both the non-invasive indices of NAFLD (HSI and THR) were higher in low-testosterone compared with high-testosterone group (HSI: 47.5 ± 2.9 vs. 38.4 ± 1.0, p = 0.005; THR: 1.70 ± 0.16 vs. 0.98 ± 0.07, p < 0.001). On the contrary, none of the non-invasive indices of hepatic fibrosis was different between groups. HSI (p = 0.038), but not THR, remained inversely independently associated with serum testosterone, after adjustment for potential confounders, including sex hormone-binding globulin. CONCLUSIONS Men with low testosterone concentrations have higher non-invasive indices of NAFLD (HSI and THR), but not of hepatic fibrosis (APRI, FIB-4, NFS), compared with counterparts of high testosterone concentrations. HSI was inversely and independently associated with testosterone concentrations.
Collapse
Affiliation(s)
- Stergios A Polyzos
- First Laboratory of Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | - Athanasios Mousiolis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gesthimani Mintziori
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|