1
|
Garza AL, Blangero J, Lee M, Bauer CX, Czerwinski SA, Choh AC. Endophenotype-Informed Association Analyses for Liver Fat Accumulation and Metabolic Dysfunction in the Fels Longitudinal Study. Int J Mol Sci 2025; 26:4812. [PMID: 40429953 PMCID: PMC12112654 DOI: 10.3390/ijms26104812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
The identification of causal genomic regions for liver fat accumulation in the context of metabolic dysfunction remains a challenging goal. This study aimed to identify potential endophenotypes for liver fat content and employ them in bivariate linkage searches for pleiotropic genetic regions where targeted association analysis is more likely to reveal significant variants. Multiple metabolic risk and adiposity distribution traits were assessed using the endophenotype ranking value. The top-ranked endophenotypes were then used in a bivariate linkage analysis, paired with liver fat content. Quantitative trait loci (QTLs) identified as significant or suggestive were targeted for measured genotype association analyses. The highest-ranked endophenotypes for liver fat accumulation were insulin resistance (IR), visceral adipose tissue (VAT), and high-density lipoprotein cholesterol (HDL-C). The univariate linkage analysis for liver fat content identified one significant QTL at chromosome 17p13.2 (Logarithm of odds score (LOD) = 2.90, p = 1.29 × 10-4). The bivariate linkage analysis pairing liver fat with IR and VAT improved the localization of two suggestive QTLs at 13q21.31 (LOD = 2.11, p = 9.03 × 10-4), and 6q21 (LOD = 2.35, p = 5.07 × 10-4), respectively. Targeted association analyses within the -1-LOD score regions of these QTLs revealed 17 marginally significant single nucleotide polymorphisms (SNPs) associated with liver fat content or its combination with the selected endophenotypes. The endophenotype-informed linkage analysis successfully identified regions suitable for the targeted association analysis of liver fat content, either alone or in combination with IR or VAT, leading to the discovery of marginally significant variants with potential for future functional studies.
Collapse
Affiliation(s)
- Ariana L. Garza
- School of Public Health, UT Health Science Center, Brownsville, TX 78520, USA
| | - John Blangero
- School of Medicine, South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Miryoung Lee
- School of Public Health, Division of Epidemiology, Human Genetics and Environmental Sciences, UT Health Science Center, Brownsville, TX 78520, USA; (M.L.); (A.C.C.)
| | - Cici X. Bauer
- School of Public Health, Division of Biostatistics, UT Health Science Center, Houston, TX 77030, USA
| | - Stefan A. Czerwinski
- School of Health and Rehabilitation Sciences, College of Medicine, Ohio State University, Columbus, OH 43004, USA
| | - Audrey C. Choh
- School of Public Health, Division of Epidemiology, Human Genetics and Environmental Sciences, UT Health Science Center, Brownsville, TX 78520, USA; (M.L.); (A.C.C.)
| |
Collapse
|
2
|
Carbone F, Després JP, Ioannidis JPA, Neeland IJ, Garruti G, Busetto L, Liberale L, Ministrini S, Vilahur G, Schindler TH, Macedo MP, Di Ciaula A, Krawczyk M, Geier A, Baffy G, Faienza MF, Farella I, Santoro N, Frühbeck G, Yárnoz-Esquiroz P, Gómez-Ambrosi J, Chávez-Manzanera E, Vázquez-Velázquez V, Oppert JM, Kiortsis DN, Sbraccia P, Zoccali C, Portincasa P, Montecucco F. Bridging the gap in obesity research: A consensus statement from the European Society for Clinical Investigation. Eur J Clin Invest 2025:e70059. [PMID: 40371883 DOI: 10.1111/eci.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/12/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Most forms of obesity are associated with chronic diseases that remain a global public health challenge. AIMS Despite significant advancements in understanding its pathophysiology, effective management of obesity is hindered by the persistence of knowledge gaps in epidemiology, phenotypic heterogeneity and policy implementation. MATERIALS AND METHODS This consensus statement by the European Society for Clinical Investigation identifies eight critical areas requiring urgent attention. Key gaps include insufficient long-term data on obesity trends, the inadequacy of body mass index (BMI) as a sole diagnostic measure, and insufficient recognition of phenotypic diversity in obesity-related cardiometabolic risks. Moreover, the socio-economic drivers of obesity and its transition across phenotypes remain poorly understood. RESULTS The syndemic nature of obesity, exacerbated by globalization and environmental changes, necessitates a holistic approach integrating global frameworks and community-level interventions. This statement advocates for leveraging emerging technologies, such as artificial intelligence, to refine predictive models and address phenotypic variability. It underscores the importance of collaborative efforts among scientists, policymakers, and stakeholders to create tailored interventions and enduring policies. DISCUSSION The consensus highlights the need for harmonizing anthropometric and biochemical markers, fostering inclusive public health narratives and combating stigma associated with obesity. By addressing these gaps, this initiative aims to advance research, improve prevention strategies and optimize care delivery for people living with obesity. CONCLUSION This collaborative effort marks a decisive step towards mitigating the obesity epidemic and its profound impact on global health systems. Ultimately, obesity should be considered as being largely the consequence of a socio-economic model not compatible with optimal human health.
Collapse
Affiliation(s)
- Federico Carbone
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Jean-Pierre Després
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, Québec, Canada
- VITAM - Centre de Recherche en santé Durable, Centre intégré Universitaire de santé et de Services Sociaux de la Capitale-Nationale, Québec, Québec, Canada
| | - John P A Ioannidis
- Department of Medicine, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Epidemiology and Population Health, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
- Department of Biomedical Science, Stanford Cardiovascular Institute, and Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, California, USA
| | - Ian J Neeland
- Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Cardiovascular Disease, Harrington Heart and Vascular Institute, Cleveland, Ohio, USA
| | - Gabriella Garruti
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Luca Busetto
- Department of Medicine, University of Padua, Padua, Italy
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Cardiology Department, Luzerner Kantonspital, Lucerne, Switzerland
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CiberCV, Institute Carlos III, Madrid, Spain
| | - Thomas H Schindler
- Washington University in St. Louis, Mallinckrodt Institute of Radiology, Division of Nuclear Medicine, Cardiovascular Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Maria Paula Macedo
- APDP - Diabetes Portugal, Education and Research Center, Lisbon, Portugal
- iNOVA4Health, NOVA Medical School | Faculdade de Ciências Médicas, NMS | FCM, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Agostino Di Ciaula
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Marcin Krawczyk
- Department of Gastroenterology, Hepatology and Transplant Medicine, Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Andreas Geier
- Interdisciplinary Amyloidosis Center of Northern Bavaria, University Hospital of Würzburg, Würzburg, Germany
- Department of Internal Medicine II, Hepatology, University Hospital of Würzburg, Würzburg, Germany
| | - Gyorgy Baffy
- Department of Medicine, VA Boston Healthcare System, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria Felicia Faienza
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Ilaria Farella
- Department of Medicine and Surgery, LUM University, Casamassima, Italy
| | - Nicola Santoro
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Medicine and Health Sciences, "V. Tiberio" University of Molise, Campobasso, Italy
| | - Gema Frühbeck
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Patricia Yárnoz-Esquiroz
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Department of Endocrinology and Nutrition, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- IdiSNA (Instituto de Investigación en la Salud de Navarra), Pamplona, Spain
- CIBERObn (CIBER Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Emma Chávez-Manzanera
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Jean-Michel Oppert
- Department of Nutrition, Pitié-Salpêtrière Hospital (AP-HP), Human Nutrition Research Center Ile-de-France (CRNH IdF), Sorbonne University, Paris, France
| | - Dimitrios N Kiortsis
- Atherothrombosis Research Centre, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Paolo Sbraccia
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carmine Zoccali
- Renal Research Institute, New York, New York, USA
- Institute of Molecular Biology and Genetics (Biogem), Ariano Irpino, Italy
- Associazione Ipertensione Nefrologia Trapianto Renale (IPNET), c/o Nefrologia, Grande Ospedale Metropolitano, Reggio Calabria, Italy
| | - Piero Portincasa
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari "Aldo Moro", Bari, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Genoa, Italy
| |
Collapse
|
3
|
Silva JG, Tavares L, Belew GD, Rodrigues JA, Araújo R, Gil AM, Jones JG. Impact of High-Fat Diet-induced Metabolic Dysfunction-associated Steatotic Liver Disease on Heart, Kidney, and Skeletal Muscle Metabolomes in Wild-Type Mice. J Proteome Res 2025; 24:2491-2504. [PMID: 40222045 DOI: 10.1021/acs.jproteome.5c00040] [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] [Indexed: 04/15/2025]
Abstract
Background: Metabolic dysfunction-associated steatotic liver disease (MASLD) can be recapitulated in mice fed a high-fat diet. The development of MASLD and the diet per se can both perturb metabolism in key extrahepatic tissues such as the heart, kidney, and skeletal muscle. To date, these alterations have not been well described in this animal model of diet-induced MASLD. Methodology: Male C57BL/6J mice were fed either standard (SC, n = 12) or high-fat chow (HF, n = 11) for 18 weeks. Metabolites were extracted from the heart, kidney, and skeletal muscle and analyzed by 1H nuclear magnetic resonance (NMR) spectroscopy, along with multivariate and univariate statistical analyses. Results: Kidney metabolite profiles exhibited the largest differences between HF and SC diets, followed by those of skeletal muscle and then the heart. Some alterations were common across all tissues, namely decreased trimethylamine and elevated levels of linoleic acid and polyunsaturated fatty acids in HF compared to SC (p < 0.05 for all three metabolites). Overall, the metabolite variations were consistent with shifts in carbohydrate and lipid substrate selection for oxidation, increased tissue stress in the heart and kidneys, and altered choline metabolism. These findings may serve as additional important descriptors of MASLD onset and progression.
Collapse
Affiliation(s)
- João G Silva
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
- Institute for Interdisciplinary Research (III-UC), Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra 3030-789, Portugal
- University School Vasco da Gama (EUVG), Vasco da Gama Research Center (CIVG), Coimbra 3020-210, Portugal
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ludgero Tavares
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
- University School Vasco da Gama (EUVG), Vasco da Gama Research Center (CIVG), Coimbra 3020-210, Portugal
| | - Getachew D Belew
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, United States
| | - João A Rodrigues
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Rita Araújo
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - Ana M Gil
- Department of Chemistry and CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro 3810-193, Portugal
| | - John G Jones
- Institute for Interdisciplinary Research (III-UC), Centre for Innovative Biomedicine and Biotechnology (CIBB), Metabolism, Aging and Disease, University of Coimbra, Cantanhede 3060-197, Portugal
| |
Collapse
|
4
|
Taylor R. Aetiology of type 2 diabetes: an experimental medicine odyssey. Diabetologia 2025:10.1007/s00125-025-06428-0. [PMID: 40316731 DOI: 10.1007/s00125-025-06428-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 05/04/2025]
Abstract
This review describes a prolonged research endeavour to test the twin cycle hypothesis that type 2 diabetes is caused by fat-induced dysfunction of the liver and pancreas, guided by the happenstance of clinical practice. Testing of the personal fat threshold hypothesis, that individuals exhibit different levels of tolerance to intra-organ fat accumulation, is also described. Both hypotheses predict that type 2 diabetes is potentially reversible by weight loss. The results of the Counterpoint study supported the twin cycle hypothesis, leading to a second study which determined that short-duration diabetes was more likely to remit following the 10-15 kg weight loss. It also confirmed that remission was durable over 6 months on an isoenergetic, normal diet. Subsequently, it was shown that weight loss caused an immediate decrease of pancreas fat only in people with type 2 diabetes and also that postprandial incretin spikes after bariatric surgery had no role in normalising fasting plasma glucose. DiRECT, a 2 year randomised controlled study, demonstrated clinical utility, observing functional beta cell capacity to return almost to normal over 12 months. A small group of participants regained weight and redeveloped type 2 diabetes, allowing observation that the underlying pathophysiological mechanisms during onset of diabetes were as postulated by the twin cycle hypothesis. Major clinical benefit was demonstrated after a further 3 year follow-up in routine care, halving the incidence of serious adverse effect compared with the standard treatment control group. In answer to the question of whether individuals have a personal fat threshold for tolerance of fat, stepwise weight loss in people with type 2 diabetes and BMI in the range 21-27 kg/m2 resulted in remission in 70%, with a wide range of fat thresholds. Type 2 diabetes can be regarded as a condition of homogenous aetiology in genetically heterogenous individuals.
Collapse
Affiliation(s)
- Roy Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
5
|
Huijbers A, Korzilius JW, Morsche RT, van der Graaf M, Wanten GJA. Intestinal failure-associated steatosis and fibroblast growth factor 21 plasma levels among adult chronic intestinal failure patients. Clin Nutr ESPEN 2025; 68:1-7. [PMID: 40294745 DOI: 10.1016/j.clnesp.2025.04.017] [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: 02/24/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND & AIMS Adult patients with chronic intestinal failure (CIF) may develop intestinal failure-associated steatosis. Asymptomatic steatosis can lead to steatohepatitis and its downstream complications. Monitoring steatosis in daily practice in adult CIF patients is hampered by limited, reliable, accessible, non-invasive methods to measure liver fat content (LFC). Fibroblast growth factor 21 (FGF21) is a hormone that is mainly produced by hepatocytes, and higher plasma levels are associated with the presence and the degree of liver steatosis in several clinical conditions. Furthermore, FGF21 analogues have been shown to reduce fatty liver. FGF21 has previously been suggested as a biomarker for liver steatosis. The aim of this study was to assess the diagnostic performance of FGF21 plasma levels to detect steatosis and steatosis severity in adult CIF patients. METHODS FGF21 plasma levels were quantified using enzyme-linked immunosorbent assay (ELISA) in 48 adult CIF patients who had been receiving home parenteral nutrition (HPN) or intravenous fluids for ≥3 months for ≥2 times per week. Liver fat content (LFC, %) was assessed with proton magnetic resonance spectroscopy (1H-MRS). Patient characteristics of patients with steatosis (LFC >5.5 %) and without steatosis (LFC ≤5.5 %) were compared using the Mann-Whitney U test or Fisher's exact test. The diagnostic value of FGF21 levels to diagnose the presence of steatosis (LFC >5.5 %) was performed by determining the area under the receiver operating characteristics curve (AUC), and the optimal cut-off value was determined. Furthermore, Spearman's rho correlation coefficient was calculated to evaluate the association between FGF21 levels and LFC. RESULTS FGF21 plasma levels were measured in 48 patients (median age of 56 years, 71 % female) with a median duration of HPN use of 57 months. Steatosis was diagnosed in 8/48 (17 %) patients, with a median LFC of 8.4 % (range 5.7-39.9 %). CIF patients with steatosis had higher median FGF21 plasma levels (658 pg/mL) than patients without steatosis (299 pg/mL). The area under the curve (AUC) of FGF21 to predict steatosis (LFC >5.5 %) was 0.80 [95 % CI 0.63, 0.96]. With the optimal FGF21 cut-off point at 453 pg/mL, the sensitivity as well as the specificity was 75 %. The calculated Spearman rho correlation found a significant positive correlation (ρ = 0.65, p < 0.001) between FGF21 plasma levels and LFC (%). CONCLUSION Adult CIF patients with steatosis had higher FGF21 plasma levels than CIF patients without steatosis. FGF21 is a good predictor for diagnosing steatosis and has a good correlation with LFC. FGF21 should be considered as a biomarker for steatosis in adult patients with CIF.
Collapse
Affiliation(s)
- Angelique Huijbers
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Julia W Korzilius
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rene Te Morsche
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Geert J A Wanten
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
6
|
Endsley C, Ali S, Salhadar K, Woodward A, Garland S, Santelli J, Salimabad MZ, Ren L, Yokoo T, Rosado-Mendez IM, Fetzer DT, de Gracia Lux C. Lipid Microparticle-Based Phantoms Modeling Hepatic Steatosis for the Validation of Quantitative Imaging Techniques. SMALL METHODS 2025:e2500043. [PMID: 40277165 DOI: 10.1002/smtd.202500043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/30/2025] [Indexed: 04/26/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) typically presents as "macrovesicular steatosis", where each hepatocyte contains a large fat vacuole (30-50 µm), indicating a more indolent form. In about 20% of cases, "microvesicular steatosis" occurs, with smaller vacuoles (1-15 µm) linked to steatohepatitis, cirrhosis progression, and increased risk of liver cancer. Emerging quantitative ultrasound (QUS) liver fat quantification (QUS-LFQ) tools measure various acoustic properties, but few methods compare techniques and imaging modalities, and the impact of fat vacuole size remains unclear. This study introduces a methodology to create ultrasound (US) phantoms that replicate fat vesicle size in MASLD. While imaging phantoms validate quantitative tools, no model currently links QUS-LFQ measurements to steatosis severity. Existing homogeneous phantoms assessing properties like attenuation, backscatter, and speed of sound overlook the microstructure of steatosis, despite the known effect of particle size on acoustic interactions. Here, agar-based phantoms simulate fat accumulation in steatotic hepatocytes using stable peanut oil droplets as analogs for lipid vacuoles. Microscopy and sizing confirm stability at 4 °C, 23 °C, and 50 °C. Both microscopy and US imaging confirm uniform distribution, with QUS-LFQ measurements reflecting fat content. These phantoms hold promise for validating quantitative imaging methods, particularly for US-based MASLD screening tools.
Collapse
Affiliation(s)
- Connor Endsley
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shariq Ali
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Karim Salhadar
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Adam Woodward
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shea Garland
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Julien Santelli
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Mehdi Zeighami Salimabad
- Departments of Medical Physics and Radiology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Liqiang Ren
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Takeshi Yokoo
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ivan M Rosado-Mendez
- Departments of Medical Physics and Radiology, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - David T Fetzer
- Department of Radiology, Collaborative for Advanced Clinical Techniques in UltraSound (CACTUS) Lab, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Caroline de Gracia Lux
- Department of Radiology, Translational Research in Ultrasound Theranostics (TRUST) Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
7
|
van den Burg EL, Schoonakker MP, van Peet PG, le Cessie S, Numans ME, Pijl H, Lamb HJ. A fasting-mimicking diet programme reduces liver fat and liver inflammation/fibrosis measured by magnetic resonance imaging in patients with type 2 diabetes. Clin Nutr 2025; 47:136-145. [PMID: 40020647 DOI: 10.1016/j.clnu.2025.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND & AIMS This study aimed to assess whether a fasting-mimicking diet (FMD) programme as an adjunct to usual care can reduce liver fat and liver inflammation/fibrosis as measured by Magnetic Resonance Imaging (MRI) in patients with type 2 diabetes. METHODS This study analyses secondary outcomes of the Fasting In diabetes Treatment (FIT) trial, which was a randomised, controlled, assessor-blinded trial in which people with type 2 diabetes using metformin only and/or diet alone for glycaemic control were randomised to receive 5-consecutive day cycles of FMD monthly as adjunct to usual care or usual care only for twelve months. Laboratory measurements, anthropometric measurements and MRI were performed at baseline, 6 and 12 months. Two MRI-derived biomarkers were measured: proton density fat-fraction (PDFF), a biomarker for liver fat, and iron content corrected T1 (cT1), a biomarker for liver inflammation/fibrosis. RESULTS Data were available of 89 participants who completed baseline visits including MRI (n = 48 in the FMD group and n = 41 in the control group). Intention-to-treat analyses, using linear mixed models, revealed significant adjusted estimated treatment effects of the FMD on PDFF (-2.8 %, 95 % CI -4.7 to -0.8, p < 0.01) and cT1 (-29.9 ms, 95 % CI -51.8 to -8.0, p < 0.01) at 12 months. In a post-hoc analysis, more participants in the FMD group compared to the control group transitioned from high to low risk for liver disease and cardiovascular disease based on PDFF ≥5.6 %. In the FMD and control group combined, every percent decrease in PDFF was associated with a decrease in HbA1c of 0.75 mmol/mol (95 % CI 0.51 to 0.99), fasting glucose of 0.14 mmol/L (95 % CI 0.08 to 0.20), triglycerides of 0.04 mmol/L (95 % CI 0.02 to 0.07), total cholesterol of 0.03 mmol/L (95 % CI 0.01 to 0.05) and weight of 0.52 kg (CI 0.33 to 0.70). Every millisecond decrease in cT1 was associated with a decrease in HbA1c of 0.05 mmol/mol (95 % CI 0.02 to 0.08), fasting glucose of 0.01 mmol/L (95 % CI 0.00 to 0.02) and weight of 0.04 kg (CI 0.01 to 0.06). CONCLUSION Following an FMD programme for 5-consecutive days per month for twelve months reduces both liver PDFF and cT1 MRI-derived biomarkers in patients with type 2 diabetes, indicating a reduction in liver fat and liver inflammation/fibrosis. Decreases in PDFF and cT1 are associated with decreases in HbA1c, fasting glucose, triglycerides and weight. Decrease in PDFF was also associated with a decrease in total cholesterol. Monthly cycles of an FMD appear to be a valuable adjunct to regular treatment of type 2 diabetes. TRIAL REGISTRATION ClinicalTrials.gov: NCT03811587.
Collapse
Affiliation(s)
- Elske L van den Burg
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands.
| | - Marjolein P Schoonakker
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Petra G van Peet
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Saskia le Cessie
- Department of Clinical Epidemiology, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Mattijs E Numans
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Hanno Pijl
- Department of Public Health and Primary Care, Leiden University Medical Centre (LUMC), Leiden, the Netherlands; Department of Internal Medicine, Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| | - Hildo J Lamb
- Department of Radiology, Cardio Vascular Imaging Group (CVIG), Leiden University Medical Centre (LUMC), Leiden, the Netherlands
| |
Collapse
|
8
|
Zhyzhneuskaya SV, Al‐Mrabeh AH, Peters C, Barnes AC, Hollingsworth KG, Welsh P, Sattar N, Lean MEJ, Taylor R. Clinical utility of liver function tests for resolution of metabolic dysfunction-associated steatotic liver disease after weight loss in the Diabetes Remission Clinical Trial. Diabet Med 2025; 42:e15462. [PMID: 39645664 PMCID: PMC11823348 DOI: 10.1111/dme.15462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 12/10/2024]
Abstract
AIMS Ectopic fat is reduced by effective weight management, but difficult to assess clinically. METHODS We evaluated paired data on 42 participants in the intervention group of the Diabetes Remission Clinical Trial (DiRECT) at baseline, 12 and 24 months after weight loss as indicators of liver fat content measured by 3-point Dixon MRI. RESULTS Baseline liver fat was elevated at 13.0 [7.8-23.3]% with fasting plasma glucose 7.9 [7.1-10.1] mmol/L. Prevalence of baseline MASLD was 86.4%. After weight loss of 11.9 ± 1.2 kg (0-37 kg) at 12 months, remission of MASLD occurred in 74% and liver fat normalised for many (1.8 [1.2-5.2]%; p < 0.0001) as did fasting glucose (5.9 [5.5-7.2] mmol/L; p < 0.0001). Alanine aminotransferase (ALT) and gamma glutamyl transferase (GGT) decreased at 12 months by 38 [19-60]% (p < 0·0001) and 38 [16-53]% (p < 0.0001) respectively. The positive predictive value for decrease in liver fat, with baseline values of >40 IU/L, was 100% for ALT and 87.5% for GGT. As expected, change in liver fat correlated with change in ALT (r = 0.64; p < 0.0001), GGT (r = 0.38; p = 0.013), AST (r = 0.36; p = 0.018), fatty liver index (r = 0.50; p < 0.0001) and hepatic steatosis index (r = 0.44; p < 0.0001). CONCLUSION Metabolic dysfunction-associated steatotic liver disease, an important marker of ill-health is improved by intentional weight loss. If enzyme levels are raised at baseline, following weight loss, changes in ALT and GGT usefully reflect change in liver fat content, with high positive predictive value. Monitoring liver enzymes can provide a simple way to assess change in liver fat following weight loss in day-to-day clinical practice.
Collapse
Affiliation(s)
- S. V. Zhyzhneuskaya
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
- University Hospital of North Durham, County Durham and Darlington NHS Foundation TrustDurhamUK
| | - A. H. Al‐Mrabeh
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of EdinburghEdinburghUK
| | - C. Peters
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
| | - A. C. Barnes
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle UniversityNewcastle upon TyneUK
| | - K. G. Hollingsworth
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
| | - P. Welsh
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - N. Sattar
- School of Cardiovascular and Metabolic Health, College of Medical Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - M. E. J. Lean
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - R. Taylor
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
9
|
Patterson WB, Young ND, Holzhausen EA, Lurmann F, Liang D, Walker DI, Jones DP, Liao J, Chen Z, Conti DV, Chatzi L, Goodrich JA, Alderete TL. Oxidative gaseous air pollutant exposure interacts with PNPLA3-I148M genotype to influence liver fat fraction and multi-omics profiles in young adults. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125692. [PMID: 39864653 PMCID: PMC11859754 DOI: 10.1016/j.envpol.2025.125692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/10/2024] [Accepted: 01/13/2025] [Indexed: 01/28/2025]
Abstract
PNPLA3-I148M genotype is the strongest predictive single-nucleotide polymorphism for liver fat. We examine whether PNPLA3-I148M modifies associations between oxidative gaseous air pollutant exposure (Oxwt) with i) liver fat and ii) multi-omics profiles of miRNAs and metabolites linked to liver fat. Participants were 69 young adults (17-22 years) from the Meta-AIR cohort. Prior-month residential Oxwt exposure (redox-weighted oxidative capacity of nitrogen dioxide and ozone) was spatially interpolated from monitoring stations via inverse-distance-squared weighting. Liver fat fraction was assessed by MRI. Serum miRNAs and metabolites were assayed via NanoString nCounter and LC-HRMS, respectively. Multi-omics factor analysis (MOFA) was used to identify latent factors with shared variance across omics layers. Multivariable linear regression models adjusted for age, sex, body mass index, and genotype with liver fat or MOFA factors as an outcome and examined PNPLA3 (rs738409; CC/CG vs. GG) as a multiplicative interaction term. Overall, a standard deviation difference in Oxwt exposure was associated with 8.9% relative increase in liver fat (p = 0.04) and this relationship differed by PNPLA3 genotype (p-value for interaction term: pintx<0.001), whereby relative increases in liver fat for GG and CC/CG participants were 71.8% and 2.4%, respectively. There was no main effect of Oxwt on MOFA Factor 1 expression (p = 0.85), but there was an interaction with PNPLA3 genotype (pintx = 0.01), whereby marginal slopes were 0.211 and -0.017 for GG and CC/CG participants, respectively. MOFA Factor 1 in turn was associated with liver fat (p = 0.006). MOFA Factor 1 miRNAs targeted genes in Fatty Acid Biosynthesis and Metabolism and Lysine Degradation pathways. MOFA Factor 9 was also associated with liver fat and was comprised of branched-chain keto acid and amino acid metabolites. The effects of Oxwt exposure on liver fat is exacerbated in young adults with two PNPLA3 risk alleles, potentially through differential effects on miRNA and/or metabolite profiles.
Collapse
Affiliation(s)
- William B Patterson
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Nathan D Young
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth A Holzhausen
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Donghai Liang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Douglas I Walker
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Dean P Jones
- Clinical Biomarkers Laboratory, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA, USA
| | - Jiawen Liao
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Zhanghua Chen
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - David V Conti
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Lida Chatzi
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Jesse A Goodrich
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Tanya L Alderete
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
10
|
Xavier A, Oyarzun JE, Zacconi F, Zanlungo S, Andia ME. Liver magnetic resonance spectroscopy as an alternative for evaluating Niemann-Pick C disease progression. RSC Adv 2025; 15:4079-4085. [PMID: 39926245 PMCID: PMC11800100 DOI: 10.1039/d4ra06781a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/30/2025] [Indexed: 02/11/2025] Open
Abstract
Niemann-Pick disease (NP) is a group of rare genetic disorders that affect normal lipid metabolism and cause an accumulation of lipids in the liver, spleen, brain, and bone marrow. NP patients develop brain alterations and a very fast progression of liver damage. The purpose of this study is to characterize the changes in liver lipid composition during the progression of this disease using ex vivo magnetic resonance spectroscopy (MRS) in mouse models with the aim of identifying potential biomarkers to support a future non-invasive technique to follow-up these patients. NP type C (NPC) and wild-type (WT) mice were fed a chow diet and euthanized at 5 weeks of age (n = 5 per group) and 9 weeks of age (n = 5 per group). We extracted lipids from their livers and analyzed them with Gas Chromatography-Mass Spectrometry (GC-MS) and MRS. With the GC-MS analysis, 7 main fatty acids (FA) and cholesterols were quantified. Using MRS, we identified 5 metabolite peaks that correspond to FA only, 3 peaks that correspond to cholesterol only, and 2 peaks that correspond to FA and cholesterol. Our results show that the increase in liver cholesterol is the key biomarker for liver damage in NPC, which is consistent with a bad liver disease prognosis due to the association of increased cholesterol levels and liver inflammation. Additionally, we identified a difference in the pool of FA stored in the NPC compared to the WT mouse livers. Those different liver spectra could provide potential biomarkers for the non-invasive follow-up of NPC patients.
Collapse
Affiliation(s)
- Aline Xavier
- Faculty of Engineering, Universidad de Santiago de Chile Santiago 9170124 Chile
- Millennium Institute for Intelligent Healthcare Engineering - IHEALTH Santiago Chile
| | - Juan E Oyarzun
- Millennium Institute for Intelligent Healthcare Engineering - IHEALTH Santiago Chile
- Radiology Department & Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile Santiago Chile
| | - Flavia Zacconi
- Faculty of Chemistry and Pharmacy, Pontificia Universidad Católica de Chile Santiago Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine, and Biological Sciences, Pontificia Universidad Católica de Chile Santiago Chile
| | - Silvana Zanlungo
- Gastroenterology Department, School of Medicine, Pontificia Universidad Católica de Chile Santiago Chile
| | - Marcelo E Andia
- Millennium Institute for Intelligent Healthcare Engineering - IHEALTH Santiago Chile
- Radiology Department & Biomedical Imaging Center, School of Medicine, Pontificia Universidad Católica de Chile Santiago Chile
| |
Collapse
|
11
|
Heinrich NS, Pedersen RP, Vestergaard MB, Lindberg U, Andersen UB, Haddock B, Fornoni A, Larsson HBW, Rossing P, Hansen TW. Kidney fat by magnetic resonance spectroscopy in type 2 diabetes with chronic kidney disease. J Diabetes Complications 2025; 39:108923. [PMID: 39647261 DOI: 10.1016/j.jdiacomp.2024.108923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/05/2024] [Accepted: 11/29/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND AND HYPOTHESIS The kidneys may be susceptible to ectopic fat and its lipotoxic effects, disposing them to chronic kidney disease (CKD) in type 2 diabetes (T2D). We investigated whether the kidney parenchyma fat content and kidney sinus fat volume would be higher in persons with T2D and CKD. METHODS Cross-sectional study including 29 controls, 27 persons with T2D and no CKD, and 48 persons with T2D and early CKD (urine albumin creatinine ratio (UACR) ≥ 30 mg/g). Kidney parenchyma fat content and kidney sinus fat volume were assessed using magnetic resonance spectroscopy and Dixon scans respectively. RESULTS In the control, T2D without CKD and T2D with CKD groups, respectively, median [1st - 3rd quartile] UACR was 5 [4 - 6], 6 [5 - 10] and 95 [43 - 278] mg/g. and mean ± standard deviation estimated glomerular filtration rate was 89 ± 11, 94 ± 11 and 77 ± 22 ml/min/1.73m2. Kidney parenchyma fat content was, respectively, 1.0 [0.5-2.4], 0.7 [0.2-1.2], 1.0 [0.3-2.0] % (p = 0.26). Kidney sinus fat volume was 2.8 [1.6-7.6], 8.0 [4.7-11.3], 10.3 [5.7-14.0] ml (p < 0.01). Around 90 % of T2D participants received a sodium-glucose cotransporter-2 inhibitor or glucagon-like peptide-1 receptor agonist. CONCLUSIONS In a setting of modern, multifactorial T2D management, kidney parenchyma fat content, evaluated with magnetic resonance spectroscopy, was similar among healthy controls and persons with T2D irrespective of CKD status. Still, kidney sinus fat volume was higher in the presence of T2D and higher still with CKD.
Collapse
Affiliation(s)
| | - Rune Ploegstra Pedersen
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Mark Bitsch Vestergaard
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Ulrich Lindberg
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Ulrik Bjørn Andersen
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Bryan Haddock
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension and Peggy and Harold Katz Drug Discovery Center, Miami, USA
| | - Henrik Bo Wiberg Larsson
- Department of Clinical Physiology and Nuclear Medicine at Rigshospitalet Glostrup, Glostrup, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
12
|
Hur YK, Lee HE, Yoo JY, Park YN, Lee IH, Bae YS. NADPH oxidase 4-SH3 domain-containing YSC84-like 1 complex participates liver inflammation and fibrosis. Free Radic Biol Med 2025; 227:246-259. [PMID: 39645205 DOI: 10.1016/j.freeradbiomed.2024.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
There is growing evidence that NADPH oxidase 4 (Nox4) in hepatocytes contributes to liver inflammation and fibrosis during the development of metabolic dysfunction-associated steatohepatitis (MASH). However, how Nox4 is regulated and leads to liver pathogenesis is unclear. Our previous studies showed that the cytosolic protein SH3 domain-containing Ysc84-like 1 (SH3YL1) regulates Nox4 activity. Here, we asked whether SH3YL1 also participates in liver inflammation and fibrosis during MASH development. We generated that whole body SH3YL1 knockout (SH3YL1-/-), Nox4 knockout (Nox4-/-) mice, and the hepatocyte-specific SH3YL1 conditional knockout (Alb-Cre/SH3YL1fl/fl) mice were fed a methionine/choline-deficient (MCD) diet to induce liver inflammation and fibrosis in pathogenesis of MASH. Palmitate-stimulated primary SH3YL1-and Nox4-deficient hepatocytes and hepatic stellate cells (HSCs) did not generate H2O2. While the liver of MCD diet-fed wild type (WT) mice demonstrated elevated 3-nitrotyrosine as a protein oxidation and 4-hydroxynonenal adducts as a lipid oxidation and increased liver inflammation, hepatocyte apoptosis, and liver fibrosis, these events were markedly reduced in SH3YL1-/-, Nox4-/-, and Alb-Cre/SH3YL1fl/fl mice. The MCD diet-fed WT mice also showed elevated hepatocyte expression of SH3YL1 protein. Similarly, liver biopsies from MASH patients demonstrated strong hepatocyte SH3YL1 protein expression, whereas hepatocytes from patients with steatosis weakly expressed SH3YL1 and histologically normal patient hepatocytes exhibited very little SH3YL1 expression. The Nox4-SH3YL1 complex in murine hepatocytes elevates their H2O2 production, which promotes the liver inflammation, hepatocyte apoptosis, and liver fibrosis that characterize MASH. This axis may also participate in MASH in humans.
Collapse
Affiliation(s)
- Yeo Kyu Hur
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea
| | - Hye Eun Lee
- Celros Biotech, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea
| | - Jung-Yeon Yoo
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea
| | - Young Nyun Park
- Department of Pathology Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-Gu, Seoul, 03722, South Korea
| | - In Hye Lee
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea.
| | - Yun Soo Bae
- Department of Life Sciences, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea; Celros Biotech, 52 Ewhayeodae-Gil, Seodaemoon-Gu, Seoul, 03760, South Korea.
| |
Collapse
|
13
|
Schenker RB, Ramirez CB, Jang C, Allayee H, Zhao X, Setchell KDR, Kohli R, Goran MI. Dihydroxyacetone phosphate is a novel predictor of hepatic fibrosis in Latino adolescents with obesity. J Pediatr Gastroenterol Nutr 2025; 80:174-181. [PMID: 39582475 DOI: 10.1002/jpn3.12420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/25/2024] [Accepted: 10/21/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVES Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common pediatric liver disease and can progress to liver fibrosis. Latino adolescents have increased MASLD and fibrosis risk. While fibrosis is diagnosed by biopsy or imaging, more accessible, noninvasive, and economical screening methods are needed. We aimed to use plasma metabolomics/lipidomics to identify potential fibrosis biomarkers in Latino adolescents with obesity. METHODS Liver stiffness (LS) was measured in 93 Latino adolescents with obesity using magnetic resonance elastography. Metabolites and lipids were extracted from plasma and identified on Compound Discoverer. Associations between metabolites/lipids and fibrosis (LS > 2.73 kPa) were determined using linear regression models after covariate adjustment. False discovery rate (FDR) adjusted Pearson's correlations were performed. Analytes yielding significant FDR-adjusted correlations were examined further by receiver operator curve analysis. RESULTS Mean (±standard deviation) alanine transaminase (ALT) was 45.7(±65.2) IU/L, hepatic fat fraction was 12.7(±9.1)%, and LS was 2.4(±0.3) kPa. We identified 795 metabolites and 413 lipids in plasma, but only one single metabolite, dihydroxyacetone phosphate (DHAP), a marker of triglyceride synthesis, was significantly associated with fibrosis after FDR adjustment (p < 0.05). In terms of predicting fibrosis, ALT had an area under the curve (AUC) of 0.79, and DHAP had an AUC of 0.79. When combined, ALT + DHAP had an AUC of 0.89. CONCLUSIONS The combination of ALT + DHAP may have the potential as an accurate, noninvasive test for liver fibrosis. Our data are limited to Latino children with obesity, and a larger cohort should be examined to further validate this novel biomarker.
Collapse
Affiliation(s)
- Rachel B Schenker
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Cuauhtemoc B Ramirez
- Department of Biologic Chemistry, University of California Irvine School of Medicine, Irvine, California, USA
| | - Cholsoon Jang
- Department of Biologic Chemistry, University of California Irvine School of Medicine, Irvine, California, USA
| | - Hooman Allayee
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, University of Southern California, Los Angeles, California, USA
| | - Xueheng Zhao
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Cincinnati Children's Hospital Medical Center, Division of Pathology and Laboratory Medicine, Cincinnati, Ohio, USA
| | - Kenneth D R Setchell
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Cincinnati Children's Hospital Medical Center, Division of Pathology and Laboratory Medicine, Cincinnati, Ohio, USA
| | - Rohit Kohli
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Michael I Goran
- Department of Pediatrics, Division of Endocrinology, Children's Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
14
|
Huang HYR, Vitali C, Zhang D, Hand NJ, Phillips MC, Creasy KT, Scorletti E, Park J, Regeneron Centre, Schneider KM, Rader DJ, Schneider CV. Deep metabolic phenotyping of humans with protein-altering variants in TM6SF2 using a genome-first approach. JHEP Rep 2025; 7:101243. [PMID: 39687601 PMCID: PMC11647476 DOI: 10.1016/j.jhepr.2024.101243] [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: 07/09/2024] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 12/18/2024] Open
Abstract
Background & Aim An unbiased genome-first approach can expand the molecular understanding of specific genes in disease-agnostic biobanks for deeper phenotyping. TM6SF2 represents a good candidate for this approach due to its known association with steatotic liver disease (SLD). Methods We screened participants with whole-exome sequences in the Penn Medicine Biobank (PMBB, n >40,000) and the UK Biobank (UKB, n >200,000) for protein-altering variants in TM6SF2 and evaluated their association with liver phenotypes and clinical outcomes. Results Missense variants in TM6SF2 (E167K, L156P, P216L) were associated with an increased risk of clinically diagnosed and imaging-proven steatosis, independent of the PNPLA3 I48M risk allele and hepatitis B/C (p <0.001). E167K homozygotes had significantly increased risk of SLD (odds ratio [OR] 5.38, p <0.001), steatohepatitis (OR 5.76, p <0.05) and hepatocellular carcinoma (OR 11.22, p <0.0001), while heterozygous carriers of L156P and P216L were also at an increased risk of steatohepatitis. In addition, carriers of E167K are at a 3-fold increased risk of at-risk MASH (OR 2.75, p <0.001). CT-derived liver fat scores were higher in E167K and L156P in an allele-dose manner (p <0.05). This corresponded with the UKB nuclear magnetic resonance-derived lipidomic analyses (n = 105,348), revealing all carriers to exhibit lower total cholesterol, triglycerides and total choline. In silico predictions suggested that these missense variants cause structural disruptions in the EXPERA domain, leading to reduced protein function. This hypothesis was supported by the association of rare loss-of-function variants in TM6SF2 with an increased risk of SLD (OR 4.9, p <0.05), primarily driven by a novel rare stop-gain variant (W35X) with the same directionality. Conclusion The functional genetic study of protein-altering variants provides insights on the association between loss of TM6SF2 function and SLD and provides the basis for future mechanistic studies. Impact and implications The genome-first approach expands insights into genetic risk factors for steatotic liver disease with TM6SF2 being a focal point due to its known association with plasma lipid traits. Our findings validated the association of two missense variants (E167K and L156P) with increased risk of hepatic steatosis on CT and MRI scans, as well as the risk of clinically diagnosed hepatocellular carcinoma independent of the common PNPLA3 I48M risk variant. Notably, we also identified a predicted deleterious missense variant (P216L) linked to steatotic risk and demonstrated that an aggregated gene burden of rare putative loss-of-function variants was associated with the risk of hepatic steatosis. Combined, this study sets the stage for future mechanistic investigations into the functional consequences of TM6SF2 variants in metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Helen Ye Rim Huang
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cecilia Vitali
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Zhang
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas J. Hand
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael C. Phillips
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kate Townsend Creasy
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eleonora Scorletti
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Institute for Translational Medicine and Therapeutics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joseph Park
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- NewYork-Presbyterian, Weill Cornell Medical Center, New York, NY 10065, USA
| | | | - Kai Markus Schneider
- Department of Medicine III, Gastroenterology, Metabolic diseases and Intensive Care, University Hospital RWTH Aachen, 52074 Aachen, Germany
- Medical Department 1, Technische Universität, Dresden, Germany
| | - Daniel J. Rader
- Department of Medicine, Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Institute for Translational Medicine and Therapeutics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Carolin Victoria Schneider
- The Institute for Translational Medicine and Therapeutics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine III, Gastroenterology, Metabolic diseases and Intensive Care, University Hospital RWTH Aachen, 52074 Aachen, Germany
| |
Collapse
|
15
|
Bahadoran Z, Mirmiran P, Kashfi K, Ghasemi A. Effects of time-restricted feeding (TRF)-model of intermittent fasting on adipose organ: a narrative review. Eat Weight Disord 2024; 29:77. [PMID: 39719521 DOI: 10.1007/s40519-024-01709-w] [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: 06/19/2024] [Accepted: 12/11/2024] [Indexed: 12/26/2024] Open
Abstract
Time-restricted feeding (TRF), an intermittent fasting approach involving a shortened eating window within 24 h, has gained popularity as a weight management approach. This review addresses how TRF may favor fat redistribution and the function of the adipose organ. TRF trials (mainly 16:8 model, with a duration of 5-48 weeks) reported a significant weight loss (1.2-10.2%, ~ 1.4-9.4 kg), with a considerable decrease in total fat mass (1.6-21%, ~ 0.5-7 kg) and visceral adipose compartment (VAC, 11-27%) in overweight and obese subjects. Experimental TRF in normal-fed and obesogenic-diet-fed mice and rats (with a fasting duration ranging between 9 and 21 h within 1-17 weeks) reported a significant reduction in body weight (~ 7-40%), total fat mass (~ 17-71%), and intrahepatic fat (~ 25-72%). TRF also improves VAC and subcutaneous adipose compartment (SAC) function by decreasing adipocyte size, macrophage infiltration, M1-macrophage polarity, and downregulating inflammatory genes. In conclusion, beyond its effect on body weight loss, total fat mass, and intrahepatic fat accumulation, TRF favors adipose organ fat redistribution in overweight and obese subjects by decreasing VAC and improving the function of VAC and SAC.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Micronutrient Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvin Mirmiran
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, 10031, USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Sahid-Erabi St, Yemen St, Chamran Exp, Tehran, Iran.
| |
Collapse
|
16
|
Misceo D, Mocciaro G, D'Amore S, Vacca M. Diverting hepatic lipid fluxes with lifestyles revision and pharmacological interventions as a strategy to tackle steatotic liver disease (SLD) and hepatocellular carcinoma (HCC). Nutr Metab (Lond) 2024; 21:112. [PMID: 39716321 DOI: 10.1186/s12986-024-00871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/13/2024] [Indexed: 12/25/2024] Open
Abstract
Steatotic liver disease (SLD) and Hepatocellular Carcinoma (HCC) are characterised by a substantial rewiring of lipid fluxes caused by systemic metabolic unbalances and/or disrupted intracellular metabolic pathways. SLD is a direct consequence of the interaction between genetic predisposition and a chronic positive energy balance affecting whole-body energy homeostasis and the function of metabolically-competent organs. In this review, we discuss how the impairment of the cross-talk between peripheral organs and the liver stalls glucose and lipid metabolism, leading to unbalances in hepatic lipid fluxes that promote hepatic fat accumulation. We also describe how prolonged metabolic stress builds up toxic lipid species in the liver, and how lipotoxicity and metabolic disturbances drive disease progression by promoting a chronic activation of wound healing, leading to fibrosis and HCC. Last, we provide a critical overview of current state of the art (pre-clinical and clinical evidence) regarding mechanisms of action and therapeutic efficacy of candidate SLD treatment options, and their potential to interfere with SLD/HCC pathophysiology by diverting lipids away from the liver therefore improving metabolic health.
Collapse
Affiliation(s)
- Davide Misceo
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Gabriele Mocciaro
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK
| | - Simona D'Amore
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Clinica Medica "G. Baccelli", "Aldo Moro" University of Bari, 70124, Bari, Italy.
| | - Michele Vacca
- Department of Interdisciplinary Medicine, Clinica Medica "C. Frugoni", "Aldo Moro" University of Bari, Piazza Giulio Cesare 11, 70124, Bari, Italy.
- Roger Williams Institute of Liver Studies, Foundation for Liver Research, London, SE5 9NT, UK.
| |
Collapse
|
17
|
Rodionov RN, Jarzebska N, Koay YC, Li M, Kuhn M, Bornstein SR, Martens-Lobenhoffer J, Eslam M, Chen FW, Rubets E, Markov AG, Weiss N, Birkenfeld A, Schwarz P, Bode-Böger SM, Perakakis N, O’Sullivan JF, George J. Symmetric dimethylguanidino valeric acid, a novel single biomarker of hepatic steatosis. iScience 2024; 27:111366. [PMID: 39660051 PMCID: PMC11629207 DOI: 10.1016/j.isci.2024.111366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/26/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
There is an unmet need for a biomarker of liver fat. We identified dimethylguanidino valeric acid (DMGV) as a circulating biomarker of liver fat. Here, we assess its two isoforms-symmetric (SDGV) and asymmetric (ADGV)-as biomarkers of steatosis. We determined plasma ADGV, SDGV, related metabolites, alanine aminotransferase (ALT), and the fatty liver index (FLI) in two cohorts and compared their diagnostic performance for liver fat detection. SDGV was the strongest predictor of moderate to severe steatosis. Changes in SDGV correlated with changes in liver fat % in a prospective cohort. In a murine model of fatty liver disease, protein levels and activity of alanine:glyoxylate aminotransferase 2 (AGXT2), which produces SDGV, were increased and coincided with elevation of SDGV concentrations. SDGV is a biomarker of liver fat and its increase in hepatic steatosis results from the upregulation of AGXT2 activity.
Collapse
Affiliation(s)
- Roman N. Rodionov
- Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, SA 5042 Australia
| | - Natalia Jarzebska
- Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Yen Chin Koay
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Mengbo Li
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Matthias Kuhn
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan R. Bornstein
- Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
- Department of Diabetes, School of Life Course Science and Medicine, King’s College London, London, UK
| | - Jens Martens-Lobenhoffer
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Mohammad Eslam
- Institute of Clinical Pharmacology, Otto-von-Guericke University, Magdeburg, Germany
| | - Fei Wen Chen
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW, Australia
| | - Elena Rubets
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | - Alexander G. Markov
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | - Norbert Weiss
- Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Andreas Birkenfeld
- German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
- Department of Internal Medicine IV, Department of Endocrinology, Diabetology and Nephrology, University Hospital of Eberhard-Karls-University Tübingen, Tübingen, Germany
- Germany and Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the Eberhard-Karls University of Tübingen, 72074 Tübingen, Germany
| | - Peter Schwarz
- Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
| | | | - Nikolaos Perakakis
- Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
- Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany
| | - John F. O’Sullivan
- Department of Internal Medicine III, University Center for Vascular Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- Cardiometabolic Medicine, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Department of Cardiology, Royal Price Alfred Hospital, Sydney, NSW, Australia
| | - Jacob George
- Institute of Clinical Pharmacology, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
18
|
Miranda J, Key Wakate Teruya A, Leão Filho H, Lahan-Martins D, Tamura Sttefano Guimarães C, de Paula Reis Guimarães V, Ide Yamauchi F, Blasbalg R, Velloni FG. Diffuse and focal liver fat: advanced imaging techniques and diagnostic insights. Abdom Radiol (NY) 2024; 49:4437-4462. [PMID: 38896247 DOI: 10.1007/s00261-024-04407-4] [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: 04/16/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024]
Abstract
The fatty liver disease represents a complex, multifaceted challenge, requiring a multidisciplinary approach for effective management and research. This article uses conventional and advanced imaging techniques to explore the etiology, imaging patterns, and quantification methods of hepatic steatosis. Particular emphasis is placed on the challenges and advancements in the imaging diagnostics of fatty liver disease. Techniques such as ultrasound, CT, MRI, and elastography are indispensable for providing deep insights into the liver's fat content. These modalities not only distinguish between diffuse and focal steatosis but also help identify accompanying conditions, such as inflammation and fibrosis, which are critical for accurate diagnosis and management.
Collapse
Affiliation(s)
- Joao Miranda
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
- Department of Radiology, University of São Paulo, R. Dr. Ovídio Pires de Campos, 75-Cerqueira César, São Paulo, SP, 05403-010, Brazil.
| | - Alexandre Key Wakate Teruya
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Hilton Leão Filho
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Daniel Lahan-Martins
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
- Departament of Radiology-FCM, State University of Campinas (UNICAMP), R. Tessália Vieira de Camargo, 126 Cidade Universitária, Campinas, SP, 13083-887, Brazil
| | - Cássia Tamura Sttefano Guimarães
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Vivianne de Paula Reis Guimarães
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Fernando Ide Yamauchi
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Roberto Blasbalg
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| | - Fernanda Garozzo Velloni
- Department of Radiology, Diagnósticos da América SA (DASA), Av Juruá 434, Alphaville Industrial, Barueri, São Paulo, SP, 06455-010, Brazil
| |
Collapse
|
19
|
Šedivý P, Dusilová T, Šetinová B, Pajuelo D, Hájek M, Rossmeislová L, Šiklová M, Šrámková V, Krauzová E, Gojda J, Koc M, Dezortová M, Kovář J. Liver fat response to two days fasting and two days isocaloric high-carbohydrate refeeding in lean and obese women. Nutr Metab Cardiovasc Dis 2024; 34:2690-2695. [PMID: 39443278 DOI: 10.1016/j.numecd.2024.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/26/2024] [Accepted: 09/28/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND AIMS Prolonged fasting, which leads to the mobilization of fat from adipose tissue, can result in the development of hepatosteatosis. However, it is not yet known whether the accumulation of fat in the liver after fasting can be affected by concurrent obesity. Therefore, this study aimed to assess how excessive adiposity influences changes in liver fat content induced by fasting and subsequent refeeding. METHODS AND RESULTS Ten lean women and eleven women with obesity (age: 36.4 ± 7.9 and 34.5 ± 7.9 years, BMI: 21.4 ± 1.7 and 34.5 ± 4.8 kg/m2) underwent a 60-h fasting period followed by 2 days of isocaloric high-carbohydrate refeeding. Magnetic resonance spectroscopy (MRS) examinations of liver were conducted at baseline, after 48 h of fasting, and at the end of refeeding period. Hepatic fat content (HFC) increased in lean women after fasting, whereas no statistically significant change in HFC was observed in women with obesity. Additionally, fasting led to significant reductions in liver volume in both groups, likely attributable to glycogen depletion, with subsequent restoration upon refeeding. Notably, changes in hepatic fat volume (HFV) rather than HFC inversely correlated with baseline liver fat content and HOMA-IR. CONCLUSION We demonstrated that prolonged fasting results in accumulation of fat in the liver in lean subjects only and that this accumulation is inversely related to baseline fat content and insulin resistance. Moreover, the study underscored the importance of evaluating hepatic fat volume rather than hepatic fat content in studies that involve considerable changes in hepatic lean volume.
Collapse
Affiliation(s)
- Petr Šedivý
- MR Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Tereza Dusilová
- MR Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Bára Šetinová
- MR Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Dita Pajuelo
- MR Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Milan Hájek
- MR Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Lenka Rossmeislová
- Department of Pathophysiology, Centre for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Michaela Šiklová
- Department of Pathophysiology, Centre for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Veronika Šrámková
- Department of Pathophysiology, Centre for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Eva Krauzová
- Department of Pathophysiology, Centre for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Internal Medicine, Third Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital, Prague, Czech Republic
| | - Jan Gojda
- Department of Pathophysiology, Centre for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Internal Medicine, Third Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital, Prague, Czech Republic
| | - Michal Koc
- Department of Pathophysiology, Centre for Research on Nutrition, Metabolism and Diabetes, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Monika Dezortová
- MR Unit, Department of Diagnostic and Interventional Radiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Jan Kovář
- Centre of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
20
|
Wang R, Wang Y, Qiu S, Ma S, Yan F, Yang GZ, Li R, Feng Y. A Comparative Study of Three Systems for Liver Magnetic Resonance Elastography. J Magn Reson Imaging 2024; 60:2472-2484. [PMID: 38449389 DOI: 10.1002/jmri.29335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/21/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Different MR elastography (MRE) systems may produce different stiffness measurements, making direct comparison difficult in multi-center investigations. PURPOSE To assess the repeatability and reproducibility of liver stiffness measured by three typical MRE systems. STUDY TYPE Prospective. POPULATION/PHANTOMS Thirty volunteers without liver disease history (20 males, aged 21-28)/5 gel phantoms. FIELD STRENGTH/SEQUENCE 3.0 T United Imaging Healthcare (UIH), 1.5 T Siemens Healthcare, 3.0 T General Electric Healthcare (GE)/Echo planar imaging-based MRE sequence. ASSESSMENT Wave images of volunteers and phantoms were acquired by three MRE systems. Tissue stiffness was evaluated by two observers, while phantom stiffness was assessed automatically by code. The reproducibility across three MRE systems was quantified based on the mean stiffness of each volunteer and phantom. STATISTICAL TESTS Intraclass correlation coefficients (ICC), coefficients of variation (CV), and Bland-Altman analyses were used to assess the interobserver reproducibility, the interscan repeatability, and the intersystem reproducibility. Paired t-tests were performed to assess the interobserver and interscan variation. Friedman tests with Dunn's multiple comparison correction were performed to assess the intersystem variation. P values less than 0.05 indicated significant difference. RESULTS The reproducibility of stiffness measured by the two observers demonstrated consistency with ICC > 0.92, CV < 4.32%, Mean bias < 2.23%, and P > 0.06. The repeatability of measurements obtained using the electromagnetic system for the liver revealed ICC > 0.96, CV < 3.86%, Mean bias < 0.19%, P > 0.90. When considering the range of reproducibility across the three systems for liver evaluations, results ranged with ICCs from 0.70 to 0.87, CVs from 6.46% to 10.99%, and Mean biases between 1.89% and 6.30%. Phantom studies showed similar results. The values of measured stiffness differed across all three systems significantly. DATA CONCLUSION Liver stiffness values measured from different MRE systems can be different, but the measurements across the three MRE systems produced consistent results with excellent reproducibility. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Runke Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy (NERC-AMRT), Shanghai Jiao Tong University, Shanghai, China
| | - Yikun Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suhao Qiu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy (NERC-AMRT), Shanghai Jiao Tong University, Shanghai, China
| | - Shengyuan Ma
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy (NERC-AMRT), Shanghai Jiao Tong University, Shanghai, China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang-Zhong Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy (NERC-AMRT), Shanghai Jiao Tong University, Shanghai, China
| | - Ruokun Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Feng
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy (NERC-AMRT), Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
21
|
Pletsch-Borba L, Wernicke C, Machann J, Meyer NM, Huong Nguyen T, Pohrt A, Hornemann S, Gerbracht C, Pfeiffer AF, Spranger J, Mai K. Increase in PUFA and protein, and decrease in carbohydrate intake improves liver fat in 12 months and the role of weight loss as a mediator: A randomized controlled trial. Clin Nutr 2024; 43:361-369. [PMID: 39577067 DOI: 10.1016/j.clnu.2024.11.010] [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: 02/14/2023] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND & AIMS Recently, a beneficial effect of high intake of unsaturated fatty acids (UFA) and protein on intrahepatic lipids (IHL) was demonstrated over 12 months within a randomized controlled trial (the NutriAct trial). We now aimed to explore the specific macronutrient components driving this IHL improvement within this trial in middle-aged and elderly subjects (50-80 y) at risk for age-related diseases. METHODS The NutriAct trial (n = 502) analyzed the effect of a high-protein and high-UFA diet on age related diseases including fatty liver disease. Individuals who completed 3-day food records with available IHL data both at baseline and at month 12 were included in this analysis. The impact of each macronutrient (E%) on IHL (measured by magnetic resonance spectroscopy) was analyzed by linear regression analyses and mediation analysis. Adherence in the intervention group was defined as intake at month 12 of ≥1 g protein/kg bodyweight or ≥25%E UFA intake; in the control group it was defined as intake of ≥15%E protein or ≥17%E UFA. RESULTS 248 participants were included in the analyses (34 % male, median age 66 y). Although BMI changed similarly in both groups within 12 months (mean change -0.41 kg/m2 in the control and -0.70 kg/m2 in the intervention group, p within groups <0.001, p between groups = 0.09), IHL improved more strongly in the compliant intervention participants than in compliant controls (estimate of relative change 0.21 % (95 % CI 0.01, 0.40), p = 0.03). Participants with stronger increase in protein and PUFA intake and a greater decrease in carbohydrate intake showed a stronger improvement in IHL (estimate for linear relative change -0.04 % (95%CI -0.06, -0.02), estimate 4th quartile vs. 1st quartile -0.40 % (95%CI -0.65, -0.16), and 0.32 % (95%CI 0.05, 0.59), respectively). These associations were partially mediated by BMI changes. Increase in PUFA intake was also directly associated with IHL improvement independently of BMI changes (estimate for linear relative change -0.03 % (95%CI -0.05, -0.01)). CONCLUSIONS Beneficial effects of increased protein and decreased carbohydrate intake on IHL are mediated by BMI changes in middle-aged and elderly subjects. The effect of high PUFA intake on IHL improvement was partly independent of weight loss. These results give insight into the understanding of a macronutrient specific effect on IHL changes in a long-term dietary intervention. CLINICAL TRIAL REGISTRATION The trial was registered at German Clinical Trials Register (drks.de) as DRKS00010049.
Collapse
Affiliation(s)
- Laura Pletsch-Borba
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Charlotte Wernicke
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Germany; Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Germany
| | - Nina Mt Meyer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany
| | - Thu Huong Nguyen
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany
| | - Anne Pohrt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biometry and Clinical Epidemiology, Germany
| | - Silke Hornemann
- Human Study Center, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany
| | - Christiana Gerbracht
- Human Study Center, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
| | - Andreas Fh Pfeiffer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Human Study Center, German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany
| | - Joachim Spranger
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany; Max Rubner Center for Cardiovascular Metabolic Renal Research, 10115, Berlin, Germany
| | - Knut Mai
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, 10117, Berlin, Germany; NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charité Center for Cardiovascular Research, 10117, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany; Max Rubner Center for Cardiovascular Metabolic Renal Research, 10115, Berlin, Germany.
| |
Collapse
|
22
|
Harreiter J, Just I, Weber M, Klepochová R, Bastian M, Winhofer Y, Wolf P, Scherer T, Leutner M, Kosi‐Trebotic L, Deischinger C, Chmelík M, Krebs MR, Trattnig S, Krššák M, Kautzky‐Willer A. Sex differences in ectopic lipid deposits and cardiac function across a wide range of glycemic control: a secondary analysis. Obesity (Silver Spring) 2024; 32:2299-2309. [PMID: 39558211 PMCID: PMC11589534 DOI: 10.1002/oby.24153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/22/2024] [Accepted: 08/20/2024] [Indexed: 11/20/2024]
Abstract
OBJECTIVE The objective of this study was to identify sex differences in ntrahepatocellular (HCL) and intramyocardial lipids (MYCL) and cardiac function in participants with different grades of glucometabolic impairment and different BMI strata. METHODS Data from 503 individuals from 17 clinical experimental studies were analyzed. HCL and MYCL were assessed with 3T and 7T scanners by magnetic resonance spectroscopy. Cardiac function was measured with a 3T scanner using electrocardiogram-gated TrueFISP sequences. Participants were classified as having normoglycemia, prediabetes, or type 2 diabetes. Three-way ANCOVA with post hoc simple effects analyses was used for statistical assessment. RESULTS Consistent increases of HCL with BMI and deterioration of glucose metabolism, especially in female individuals, were detected. MYCL increased with BMI and glucose impairment in female individuals, but not in male individuals. Sex differences were found in cardiac function loss, with significant effects found among male individuals with worsening glucose metabolism. Myocardial mass and volume of the ventricle were higher in male individuals in all groups. This sex difference narrowed with increasing BMI and with progressing dysglycemia. CONCLUSIONS Sex differences in HCL and MYCL may be associated with a higher cardiovascular disease risk observed in female individuals progressing to diabetes. Further studies are needed to elucidate possible sex differences with advancing glucometabolic impairment and obesity and their potential impact on cardiovascular outcomes.
Collapse
Affiliation(s)
- Jürgen Harreiter
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
- Department of MedicineLandesklinikum ScheibbsScheibbsAustria
| | - Ivica Just
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
- High Field MR Center, Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Michael Weber
- Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Radka Klepochová
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
- High Field MR Center, Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Magdalena Bastian
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Yvonne Winhofer
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Peter Wolf
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Michael Leutner
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Lana Kosi‐Trebotic
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Carola Deischinger
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Marek Chmelík
- High Field MR Center, Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
- Department of Technical Disciplines in Health Care at Faculty of Health CareUniversity of PrešovPrešovSlovakia
| | - Michael R. Krebs
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - Siegfried Trattnig
- High Field MR Center, Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Martin Krššák
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
- High Field MR Center, Department of Biomedical Imaging and Image‐guided TherapyMedical University of ViennaViennaAustria
| | - Alexandra Kautzky‐Willer
- Division of Endocrinology and Metabolism, Department of Internal Medicine IIIMedical University of ViennaViennaAustria
| |
Collapse
|
23
|
Mishra P, Sadananthan SA, Yaligar J, Tan KH, Chong YS, Gluckman PD, Godfrey KM, Fortier MV, Eriksson JG, Chan JKY, Chan SY, Wang D, Velan SS, Michael N. Even moderate liver fat accumulation below conventional fatty liver cutoffs is linked to multiple metabolomic alterations and gestational dysglycemia in Asian women of reproductive age. BMC Med 2024; 22:561. [PMID: 39605006 PMCID: PMC11600899 DOI: 10.1186/s12916-024-03779-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND It is not clear if conventional liver fat cutoff of 5.56% weight which has been used for identifying fatty liver in western populations is also applicable for Asians. In Asian women of reproductive age, we evaluate the optimum metabolic syndrome (MetS)-linked liver fat cutoff, the specific metabolomic alterations apparent at this cutoff, as well as prospective associations of preconception liver fat levels with gestational dysglycemia. METHODS Liver fat (measured by magnetic resonance spectroscopy), MetS, and nuclear magnetic resonance (NMR)-based plasma metabolomic profiles were assessed in 382 Asian women, who were planning to conceive. Ninety-eight women went on to become pregnant and received an oral glucose tolerance test at week 26 of gestation. RESULTS The optimum liver fat cutoff for diagnosing MetS was 2.07%weight. Preconception liver fat was categorized into Low (liver fat < 2.07%), Moderate (2.07% ≤ liver fat < 5.56%), and High (liver fat ≥ 5.56%) groups. Individual MetS traits showed worsening trends, going from Low to Moderate to High groups. Multiple plasma metabolomic alterations, previously linked to incident type 2 diabetes (T2D), were already evident in the Moderate group (adjusted for ethnicity, age, parity, educational attainment, and BMI). Both a cross-sectional multi-metabolite score for incident T2D and mid-gestational glucose area under the curve showed increasing trends, going from Low to Moderate to High groups (p < 0.001 for both). Gestational diabetes incidence was 2-fold (p = 0.23) and 7-fold (p < 0.001) higher in the Moderate and High groups relative to the Low group. CONCLUSIONS In Asian women of reproductive age, moderate liver fat accumulation below the conventional fatty liver cutoff was not metabolically benign and was linked to gestational dysglycemia. The newly derived cutoff can aid in screening individuals before adverse metabolic phenotypes have consolidated, which provides a longer window for preventive strategies.
Collapse
Affiliation(s)
- Priti Mishra
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
| | - Suresh Anand Sadananthan
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
| | - Jadegoud Yaligar
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
| | - Kok Hian Tan
- Academic Clinical Program in Obstetrics and Gynaecology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Maternal-Fetal Medicine, KK Women's and Children's Hospital (KKH), Singapore, Singapore
| | - Yap Seng Chong
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Peter D Gluckman
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Keith M Godfrey
- NIHR Southampton Biomedical Research Centre, Southampton University Hospital NHS Foundation Trust and University of Southampton, Southampton, UK
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Marielle V Fortier
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
- Department of Diagnostic and Interventional Imaging, KK Women's and Children's Hospital (KKH), Singapore, Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Folkhalsan Research Centre, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| | - Jerry Kok Yen Chan
- Academic Clinical Program in Obstetrics and Gynaecology, Duke-National University of Singapore Medical School, Singapore, Singapore
- Department of Reproductive Medicine, KK Women's and Children's Hospital (KKH), Singapore, Singapore
| | - Shiao-Yng Chan
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dennis Wang
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore, 138671, Singapore
- National Heart and Lung Institute, Imperial College London, London, UK
| | - S Sendhil Velan
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore
| | - Navin Michael
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, 117609, Singapore.
| |
Collapse
|
24
|
Schuppelius B, Schüler R, Pivovarova-Ramich O, Hornemann S, Busjahn A, Machann J, Kruse M, Park SQ, Kabisch S, Csanalosi M, Ost AC, Pfeiffer AFH. Alterations in Glucagon Levels and the Glucagon-to-Insulin Ratio in Response to High Dietary Fat or Protein Intake in Healthy Lean Adult Twins: A Post Hoc Analysis. Nutrients 2024; 16:3905. [PMID: 39599691 PMCID: PMC11597242 DOI: 10.3390/nu16223905] [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: 10/14/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Emerging data support evidence of the essential role of glucagon for lipid metabolism. However, data on the role of dietary fat intake for glucagon secretion is limited. This analysis investigated whether altering nutritional fat intake affects glucagon levels in healthy subjects. Methods: A total of 92 twins (age: 31 ± 14 years, BMI: 23 ± 3 kg/m2) consumed two 6-week diets: first a low-fat, high-carbohydrate diet (LFD) followed by an isocaloric high-fat, low-carbohydrate diet (HFD). In total, 24 twins (age: 39 ± 15 years, BMI: 24 ± 2 kg/m2) continued with a high-protein diet (HPD). Clinical investigations were performed after 6 weeks of the LFD, after 1 and 6 weeks of the HFD and after 6 weeks of the HPD. Results: The LFD caused a significant decrease in fasting glucagon (-27%, p < 0.001) compared to baseline. After 6 weeks of the HFD, glucagon increased (117%, p < 0.001 vs. LFD), while free fatty acids decreased. Six weeks of the HPD further increased glucagon levels (72%, p = 0.502 vs. HFD), although fasting amino acid levels remained constant. Fasting insulin and HOMA-IR moderately increased after one week of the HFD, while six weeks of the HPD significantly decreased both. The fasting glucagon-to-insulin ratio decreased during the LFD (p < 0.001) but increased after the HFD (p < 0.001) and even further increased after the HPD (p = 0.018). Liver fat, triglycerides and blood glucose did not increase during the HFD. The heritability of glucagon levels was 45% with the LFD. Conclusions: An HFD increases glucagon levels and the glucagon-to-insulin ratio under isocaloric conditions compared to an LFD in healthy lean subjects. This rise in glucagon may represent a metabolic response to prevent hepatic steatosis, as glucagon increases have been previously shown to induce hepatic fat oxidation.
Collapse
Affiliation(s)
- Bettina Schuppelius
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Rita Schüler
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Olga Pivovarova-Ramich
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- Department of Molecular Metabolism and Precision Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
| | - Silke Hornemann
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Andreas Busjahn
- HealthTwiSt GmbH, Robert-Rössle-Strasse 10, 13125 Berlin, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Centre Munich, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
- Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, University of Tübingen, Geissweg 3, 72076 Tübingen, Germany
| | - Michael Kruse
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Soyoung Q. Park
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
- Department of Decision Neuroscience and Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- Neuroscience Research Center, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany
| | - Stefan Kabisch
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
| | - Marta Csanalosi
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Anne-Cathrin Ost
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology and Metabolism, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764 München-Neuherberg, Germany
| |
Collapse
|
25
|
Chen L, Tan KML, Xu J, Mishra P, Mir SA, Gong M, Narasimhan K, Ng B, Lai JS, Tint MT, Cai S, Sadananthan SA, Michael N, Yaligar J, Velan SS, Leow MKS, Tan KH, Chan J, Meaney MJ, Chan SY, Chong YS, Eriksson JG. Exploring multi-omics and clinical characteristics linked to accelerated biological aging in Asian women of reproductive age: insights from the S-PRESTO study. Genome Med 2024; 16:128. [PMID: 39516835 PMCID: PMC11549770 DOI: 10.1186/s13073-024-01403-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Phenotypic age (PhenoAge), a widely used marker of biological aging, has been shown to be a robust predictor of all-cause mortality and morbidity in different populations. Existing studies on biological aging have primarily focused on individual domains, resulting in a lack of a comprehensive understanding of the multi-systemic dysregulation that occurs in aging. METHODS PhenoAge was evaluated based on a linear combination of chronological age (CA) and 9 clinical biomarkers in 952 multi-ethnic Asian women of reproductive age. Phenotypic age acceleration (PhenoAgeAccel), an aging biomarker, represents PhenoAge after adjusting for CA. This study conducts an in-depth association analysis of PhenoAgeAccel with clinical, nutritional, lipidomic, gut microbiome, and genetic factors. RESULTS Higher adiposity, glycaemia, plasma saturated fatty acids, kynurenine pathway metabolites, GlycA, riboflavin, nicotinamide, and insulin-like growth factor binding proteins were positively associated with PhenoAgeAccel. Conversely, a healthier diet and higher levels of pyridoxal phosphate, all-trans retinol, betaine, tryptophan, glutamine, histidine, apolipoprotein B, and insulin-like growth factors were inversely associated with PhenoAgeAccel. Lipidomic analysis found 132 lipid species linked to PhenoAgeAccel, with PC(O-36:0) showing the strongest positive association and CE(24:5) demonstrating the strongest inverse association. A genome-wide association study identified rs9864994 as the top genetic variant (P = 5.69E-07) from the ZDHHC19 gene. Gut microbiome analysis revealed that Erysipelotrichaceae UCG-003 and Bacteroides vulgatus were inversely associated with PhenoAgeAccel. Integrative network analysis of aging-related factors underscored the intricate links among clinical, nutritional and lipidomic variables, such as positive associations between kynurenine pathway metabolites, amino acids, adiposity, and insulin resistance. Furthermore, potential mediation effects of blood biomarkers related to inflammation, immune response, and nutritional and energy metabolism were observed in the associations of diet, adiposity, genetic variants, and gut microbial species with PhenoAgeAccel. CONCLUSIONS Our findings provide a comprehensive analysis of aging-related factors across multiple platforms, delineating their complex interconnections. This study is the first to report novel signatures in lipidomics, gut microbiome and blood biomarkers specifically associated with PhenoAgeAccel. These insights are invaluable in understanding the molecular and metabolic mechanisms underlying biological aging and shed light on potential interventions to mitigate accelerated biological aging by targeting modifiable factors.
Collapse
Affiliation(s)
- Li Chen
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Karen Mei-Ling Tan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore.
| | - Jia Xu
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Priti Mishra
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sartaj Ahmad Mir
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Min Gong
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kothandaraman Narasimhan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Bryan Ng
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jun Shi Lai
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mya Thway Tint
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Shirong Cai
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suresh Anand Sadananthan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Navin Michael
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jadegoud Yaligar
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sambasivam Sendhil Velan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Medicine and Human Potential Translational Research Programme. Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Melvin Khee Shing Leow
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kok Hian Tan
- Duke-NUS Medical School, Singapore, Singapore
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Jerry Chan
- Duke-NUS Medical School, Singapore, Singapore
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Michael J Meaney
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Sackler Program for Epigenetics & Psychobiology, McGill University, Montréal, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montréal, Canada
| | - Shiao-Yng Chan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yap Seng Chong
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Folkhalsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| |
Collapse
|
26
|
de Abreu JDMF, Azulay RS, Rodrigues V, de Abreu SLL, da Glória Tavares M, Pinheiro FCM, de Oliveira Neto CP, Andrade C, Facundo A, Sá AG, Azevedo PR, de Almeida AGP, Costa DCDA, Castro RS, Magalhães M, Nascimento GC, Faria MDS, Ferreira ADSP. Predictors of Hepatic Fibrosis in Type 2 Diabetes Patients with Metabolic-Dysfunction-Associated Steatotic Liver Disease. Biomedicines 2024; 12:2542. [PMID: 39595107 PMCID: PMC11592232 DOI: 10.3390/biomedicines12112542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Approximately 25% of the world's population and more than 60% of patients with type 2 diabetes (T2D) have metabolic-dysfunction-associated steatotic liver disease (MASLD). The association between these pathologies is an important cause of morbidity and mortality in Brazil and worldwide due to the high frequency of advanced fibrosis and cirrhosis. The objective of this study was to determine the epidemiologic and clinical-laboratory profile of patients with T2D and MASLD treated at an endocrinology reference service in a state in northeastern Brazil, and to investigate the association of liver fibrosis with anthropometric and laboratory measurements. METHODS A cross-sectional study was performed in a specialized outpatient clinic with 240 patients evaluated from July 2022 to February 2024, using a questionnaire, physical examination, laboratory tests, and liver elastography with FibroScan®. RESULTS Estimates showed that women (adjusted OR = 2.69, 95% CI = 1.35-5.35, p = 0.005), obesity (adjusted OR = 2.23, 95% CI = 1.22-4.07, p = 0.009), high GGT (adjusted OR = 3.78, 95% CI = 2.01-7.14, p < 0. 001), high AST (adjusted OR = 6.07, 95% CI = 2.27-16.2, p < 0.001), and high ALT (adjusted OR = 3.83, 95% CI = 1.80-8.11, p < 0.001) were associated with the risk of liver fibrosis even after adjusted analysis. CONCLUSIONS The study findings suggested that female sex and BMI were associated with an increased risk of liver fibrosis, highlighting the importance of comprehensive evaluation of these patients. In addition, FIB-4 and MAF-5 provided a good estimate of liver fibrosis in our population and may serve as a useful tool in a public health setting with limited resources.
Collapse
Affiliation(s)
- Joana D’Arc Matos França de Abreu
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Rossana Sousa Azulay
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Vandilson Rodrigues
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Sterffeson Lamare Lucena de Abreu
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Maria da Glória Tavares
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Flávia Coelho Mohana Pinheiro
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Clariano Pires de Oliveira Neto
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Caio Andrade
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis 65020-070, Brazil
| | - Alexandre Facundo
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Adriana Guimarães Sá
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Patrícia Ribeiro Azevedo
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Ana Gregória Pereira de Almeida
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Debora Camelo de Abreu Costa
- Service of Hepatology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (D.C.d.A.C.); (R.S.C.); (A.d.S.P.F.)
| | - Rogério Soares Castro
- Service of Hepatology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (D.C.d.A.C.); (R.S.C.); (A.d.S.P.F.)
| | - Marcelo Magalhães
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Gilvan Cortês Nascimento
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
| | - Manuel dos Santos Faria
- Service of Endocrinology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (M.d.G.T.); (C.P.d.O.N.); (A.F.); (G.C.N.); (M.d.S.F.)
- Research Group in Endocrinology and Clinical and Molecular Metabolism (ENDOCLIM), Sao Luis 65020-070, Brazil; (V.R.); (S.L.L.d.A.); (F.C.M.P.); (C.A.); (A.G.S.); (P.R.A.); (A.G.P.d.A.); (M.M.)
- Post-Graduate Program in Adult Health (PPGSAD), Federal University of Maranhão (UFMA), São Luis 65020-070, Brazil
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil
| | - Adalgisa de Souza Paiva Ferreira
- Service of Hepatology, University Hospital of the Federal University of Maranhão (HUUFMA/EBSERH), São Luis 65020-070, Brazil; (D.C.d.A.C.); (R.S.C.); (A.d.S.P.F.)
- Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís 65080-805, Brazil
| |
Collapse
|
27
|
Haghshomar M, Antonacci D, Smith AD, Thaker S, Miller FH, Borhani AA. Diagnostic Accuracy of CT for the Detection of Hepatic Steatosis: A Systematic Review and Meta-Analysis. Radiology 2024; 313:e241171. [PMID: 39499183 DOI: 10.1148/radiol.241171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Background CT plays an important role in the opportunistic identification of hepatic steatosis. CT performance for steatosis detection has been inconsistent across various studies, and no clear guidelines on optimum thresholds have been established. Purpose To conduct a systematic review and meta-analysis to assess CT diagnostic accuracy in hepatic steatosis detection and to determine reliable cutoffs for the commonly mentioned measures in the literature. Materials and Methods A systematic search of the PubMed, Embase, and Scopus databases (English-language studies published from September 1977 to January 2024) was performed. Studies evaluating the diagnostic accuracy of noncontrast CT (NCCT), contrast-enhanced (CECT), and dual-energy CT (DECT) for hepatic steatosis detection were included. Reference standards included biopsy, MRI proton density fat fraction (PDFF), or NCCT. In several CECT and DECT studies, NCCT was used as the reference standard, necessitating subgroup analysis. Statistical analysis included a random-effects meta-analysis, assessment of heterogeneity with use of the I2 statistic, and meta-regression to explore potential sources of heterogeneity. When available, mean liver attenuation, liver-spleen attenuation difference, liver to spleen attenuation ratio, and the DECT-derived fat fraction for hepatic steatosis diagnosis were assessed. Results Forty-two studies (14 186 participants) were included. NCCT had a sensitivity and specificity of 72% and 88%, respectively, for steatosis (>5% fat at biopsy) detection and 82% and 94% for at least moderate steatosis (over 20%-33% fat at biopsy) detection. CECT had a sensitivity and specificity of 66% and 90% for steatosis detection and 68% and 93% for at least moderate steatosis detection. DECT had a sensitivity and specificity of 85% and 88% for steatosis detection. In the subgroup analysis, the sensitivity and specificity for detecting steatosis were 80% and 99% for CECT and 84% and 93% for DECT. There was heterogeneity among studies focusing on CECT and DECT. Liver attenuation less than 40-45 HU, liver-spleen attenuation difference less than -5 to 0 HU, and liver to spleen attenuation ratio less than 0.9-1 achieved high specificity for detection of at least moderate steatosis. Conclusion NCCT showed high performance for detection of at least moderate steatosis. © RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Maryam Haghshomar
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Dominic Antonacci
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Andrew D Smith
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Sarang Thaker
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Frank H Miller
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| | - Amir A Borhani
- From the Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N St. Clair St, Arkes Family Pavilion, Ste 800, Chicago, IL 60611 (M.H., D.A., S.T., F.H.M., A.A.B.); and Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, Tenn (A.D.S.)
| |
Collapse
|
28
|
Schenker RB, Machle CJ, Allayee H, Lurmann F, Patterson WB, Kohli R, Goran MI, Alderete TL. Ambient air pollution exposure is associated with liver fat and stiffness in Latino youth with a more pronounced effect in those with PNPLA3 genotype and more advanced liver disease. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117234. [PMID: 39454357 PMCID: PMC11578286 DOI: 10.1016/j.ecoenv.2024.117234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/10/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Exposure to ambient air pollutants has emerged as a risk for metabolic-dysfunction associated steatotic liver disease (MASLD). OBJECTIVES We sought to examine associations between short-term (prior month) and long-term (prior year) ambient air pollution exposure with hepatic fat fraction (HFF) and liver stiffness in Latino youth with obesity. A secondary aim was to investigate effect modification by patatin-like phospholipase domain-containing protein 3 (PNPLA3) genotype and liver disease severity. METHODS Data was analyzed from 113 Latino youth (age 11-19) with obesity in Southern California. Individual exposure to particulate matter with aerodynamic diameter ≤ 2.5μm (PM2.5), ≤ 10μm (PM10), nitrogen dioxide (NO2), 8-hour maximum ozone (8hrMax-O3), 24-hr O3, and redox-weighted oxidative capacity (Oxwt) were estimated using residential address histories and United States Environmental Protection Agency air quality observations. HFF and liver stiffness were measured using magnetic resonance imaging. Linear models were used to determine associations between short-term and long-term exposure to air pollutants with HFF and liver stiffness. Modification by PNPLA3 and liver disease severity was then examined. RESULTS Short-term exposure to 8hrMax-O3 was positively associated with HFF. Relationships between air pollution exposure and HFF were not impacted by PNPLA3 genotype or liver disease severity. Long-term exposure to 8hrMax-O3 and Oxwt were positively associated with liver stiffness. Associations between air pollution exposure and liver stiffness depended on PNPLA3 genotype, such that individuals with GG genotypes exhibited stronger, more positive relationships between short-term exposure to PM10, 8hrMax-O3, 24-hr O3, and Oxwt and liver stiffness than individuals with CC/CG genotypes. In addition, relationships between short-term exposure to NO2 and liver stiffness were stronger in those with severe liver disease. DISCUSSION Air pollution exposure may be a risk factor for liver disease among Latino youth with obesity, particularly in those with other preexisting risks for liver damage.
Collapse
Affiliation(s)
- Rachel B Schenker
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA.
| | - Christopher J Machle
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA; Department of Psychology, University of Oregon, Eugene, OR, USA.
| | - Hooman Allayee
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, University of Southern California, Los Angeles, CA, USA.
| | | | - William B Patterson
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA.
| | - Rohit Kohli
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA.
| | - Michael I Goran
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA.
| | - Tanya L Alderete
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
29
|
Basaria S, Taplin ME, McDonnell M, Simonson DC, Lin AP, Dufour AB, Habtemariam D, Nguyen PL, Ravi P, Kibel AS, Sweeney CJ, D’ Amico AV, Roberts DA, Xu W, Wei XX, Sunkara R, Choudhury AD, Mantia C, Beltran H, Pomerantz M, Berchuck JE, Martin NE, Leeman JE, Mouw KW, Kilbridge KE, Bearup R, Kackley H, Kafel H, Huang G, Reid KF, Storer T, Braga-Basaria M, Travison TG. Insulin resistance during androgen deprivation therapy in men with prostate cancer. Cancer 2024; 130:3671-3685. [PMID: 38881266 PMCID: PMC11464184 DOI: 10.1002/cncr.35443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) in prostate cancer (PCa) has been associated with development of insulin resistance. However, the predominant site of insulin resistance remains unclear. METHODS The ADT & Metabolism Study was a single-center, 24-week, prospective observational study that enrolled ADT-naive men without diabetes who were starting ADT for at least 24 weeks (ADT group, n = 42). The control group comprised men without diabetes with prior history of PCa who were in remission after prostatectomy (non-ADT group, n = 23). Prevalent diabetes mellitus was excluded in both groups using all three laboratory criteria defined in the American Diabetes Association guidelines. All participants were eugonadal at enrollment. The primary outcome was to elucidate the predominant site of insulin resistance (liver or skeletal muscle). Secondary outcomes included assessments of body composition, and hepatic and intramyocellular fat. Outcomes were assessed at baseline, 12, and 24 weeks. RESULTS At 24 weeks, there was no change in hepatic (1.2; 95% confidence interval [CI], -2.10 to 4.43; p = .47) or skeletal muscle (-3.2; 95% CI, -7.07 to 0.66; p = .10) insulin resistance in the ADT group. No increase in hepatic or intramyocellular fat deposition or worsening of glucose was seen. These changes were mirrored by those observed in the non-ADT group. Men undergoing ADT gained 3.7 kg of fat mass. CONCLUSIONS In men with PCa and no diabetes, 24 weeks of ADT did not change insulin resistance despite adverse body composition changes. These findings should be reassuring for treating physicians and for patients who are being considered for short-term ADT.
Collapse
Affiliation(s)
- Shehzad Basaria
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mary-Ellen Taplin
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Marie McDonnell
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Donald C. Simonson
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander P. Lin
- Center for Clinical Spectroscopy, Department of Radiology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Alyssa B. Dufour
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel Habtemariam
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Paul L. Nguyen
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Praful Ravi
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Adam S. Kibel
- Division of Urology, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher J. Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Anthony V. D’ Amico
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel A. Roberts
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Wenxin Xu
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiao X. Wei
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Rajitha Sunkara
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Atish D. Choudhury
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Charlene Mantia
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Himisha Beltran
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark Pomerantz
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob E. Berchuck
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Neil E. Martin
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan E. Leeman
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Kent W. Mouw
- Department of Radiation Oncology, Brigham and Women’s Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Kerry E. Kilbridge
- Lank Center for Genitourinary Oncology and Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Richelle Bearup
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hannah Kackley
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hussein Kafel
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Grace Huang
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Kieran F. Reid
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Storer
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Milena Braga-Basaria
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas G. Travison
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Stangl TA, Wiepjes CM, Smit RAJ, van Hylckama Vlieg A, Lamb HJ, van der Velde JHPM, Winters-van Eekelen E, Boone SC, Brouwers MCGJ, Rosendaal FR, den Heijer M, Heijboer AC, de Mutsert R. Association Between Low Sex Hormone-Binding Globulin and Increased Risk of Type 2 Diabetes Is Mediated by Increased Visceral and Liver Fat: Results From Observational and Mendelian Randomization Analyses. Diabetes 2024; 73:1793-1804. [PMID: 39106187 DOI: 10.2337/db23-0982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 07/29/2024] [Indexed: 08/09/2024]
Abstract
The aim of this study was to investigate the associations among sex hormone-binding globulin (SHBG), visceral adipose tissue (VAT), liver fat content, and risk of type 2 diabetes (T2D). In the Netherlands Epidemiology of Obesity study, 5,690 women (53%) and men (47%) without preexisting diabetes were included and followed for incident T2D. SHBG concentrations were measured in all participants, VAT was measured using MRI, and liver fat content was measured using proton magnetic resonance spectroscopy in a random subset of 1,822 participants. We examined associations between SHBG and liver fat using linear regression and bidirectional Mendelian randomization analyses and between SHBG and T2D using Cox regression adjusted for confounding and additionally for VAT and liver fat to examine mediation. Mean age was 56 (SD 6) years, mean BMI was 30 (SD 4) kg/m2, median SHBG was 47 (interquartile range [IQR] 34-65) nmol/L in women and 34 (26-43) nmol/L in men, and median liver fat was 3.4% (IQR 1.6-8.2%) in women and 6.0% (2.9-13.5%) in men. Compared with the highest SHBG quartile, liver fat was 2.9-fold (95% CI 2.4, 3.4) increased in women and 1.6-fold (95% CI 1.3, 1.8) increased in men, and the hazard ratio of T2D was 4.9 (95% CI 2.4, 9.9) in women and 1.8 (1.1, 2.9) in men. Genetically predicted SHBG was associated with liver fat content (women: SD -0.45 [95% CI -0.55, -0.35]; men: natural logarithm, -0.25 [95% CI -0.34, -0.16]). VAT and liver fat together mediated 43% (women) and 60% (men) of the SHBG-T2D association. To conclude, in a middle-aged population with overweight, the association between low SHBG and increased risk of T2D was, for a large part, mediated by increased VAT and liver fat. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Theresa A Stangl
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Chantal M Wiepjes
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Roelof A J Smit
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Hildo J Lamb
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Sebastiaan C Boone
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Martijn C G J Brouwers
- Division of Endocrinology and Metabolic Diseases, Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Martin den Heijer
- Amsterdam UMC, location Vrije Universiteit Amsterdam, Department of Endocrinology and Metabolism, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Annemieke C Heijboer
- Amsterdam UMC, location Vrije Universiteit Amsterdam and location University of Amsterdam, Endocrine Laboratory, Department of Laboratory Medicine, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam Reproduction Development, Amsterdam, the Netherlands
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
31
|
Arold D, Bornstein SR, Perakakis N, Ehrlich S, Bernardoni F. Regional gray matter changes in steatotic liver disease provide a neurobiological link to depression: A cross-sectional UK Biobank cohort study. Metabolism 2024; 159:155983. [PMID: 39089490 DOI: 10.1016/j.metabol.2024.155983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Steatotic liver disease (SLD) is characterized by excessive accumulation of lipids in the liver. It is associated with elevated risk of hepatic and cardiometabolic diseases, as well as mental disorders such as depression. Previous studies revealed global gray matter reduction in SLD. To investigate a possible shared neurobiology with depression, we examined liver fat-related regional gray matter alterations in SLD and its most significant clinical subgroup metabolic dysfunction-associated steatotic liver disease (MASLD). METHODS We analyzed regional cortical thickness and area obtained from brain MRI in 29,051 participants in UK Biobank. Liver fat amount was computed as proton density fat fraction (PDFF) from liver MRI scans. We examined the relationship between brain structure and PDFF, adjusting for sociodemographic, physical, lifestyle, and environmental factors, as well as alcohol intake and a spectrum of cardiometabolic covariates. Finally, we compared patterns of brain alterations in SLD/MASLD and major depressive disorder (MDD) using previously published results. RESULTS PDFF-related gray matter alterations were region-specific, involving both increases and decreases in cortical thickness, and increased cortical area. In several regions, PDFF effects on gray matter could also be attributed to cardiometabolic covariates. However, PDFF was consistently associated with lower cortical thickness in middle and superior temporal regions and higher cortical thickness in pericalcarine and right frontal pole regions. PDFF-related alterations for the SLD and the MASLD group correlated with those observed in MDD (Pearson r = 0.45-0.54, p < 0.01). CONCLUSION These findings suggest the presence of shared biological mechanisms linking MDD to SLD and MASLD. They might explain the well-known elevated risk of depression in these groups and support early lifestyle interventions and treatment of metabolic risk factors for the successful management of the interconnected diseases depression and SLD/MASLD.
Collapse
Affiliation(s)
- Dominic Arold
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Nikolaos Perakakis
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Paul Langerhans Institute Dresden (PLID), Helmholtz Center Munich, University Hospital and Faculty of Medicine, TU Dresden, Dresden, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Stefan Ehrlich
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Fabio Bernardoni
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany.
| |
Collapse
|
32
|
Lee Y, Yoon S, Paek M, Han D, Choi MH, Park SH. Advanced MRI techniques in abdominal imaging. Abdom Radiol (NY) 2024; 49:3615-3636. [PMID: 38802629 DOI: 10.1007/s00261-024-04369-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024]
Abstract
Magnetic resonance imaging (MRI) is a crucial modality for abdominal imaging evaluation of focal lesions and tissue properties. However, several obstacles, such as prolonged scan times, limitations in patients' breath-hold capacity, and contrast agent-associated artifacts, remain in abdominal MR images. Recent techniques, including parallel imaging, three-dimensional acquisition, compressed sensing, and deep learning, have been developed to reduce the scan time while ensuring acceptable image quality or to achieve higher resolution without extending the scan duration. Quantitative measurements using MRI techniques enable the noninvasive evaluation of specific materials. A comprehensive understanding of these advanced techniques is essential for accurate interpretation of MRI sequences. Herein, we therefore review advanced abdominal MRI techniques.
Collapse
Affiliation(s)
- Yoonhee Lee
- Department of Radiology, Gil Medical Center, Gachon University College of Medicine, 21, Namdong-daero 774beon-gil, Namdong-gu, Incheon, 21565, Republic of Korea
| | - Sungjin Yoon
- Department of Radiology, Gil Medical Center, Gachon University College of Medicine, 21, Namdong-daero 774beon-gil, Namdong-gu, Incheon, 21565, Republic of Korea
| | | | - Dongyeob Han
- Siemens Healthineers Ltd, Seoul, Republic of Korea
| | - Moon Hyung Choi
- Department of Radiology, Catholic University of Korea Eunpyeong St Mary's Hospital, Seoul, Republic of Korea
| | - So Hyun Park
- Department of Radiology, Gil Medical Center, Gachon University College of Medicine, 21, Namdong-daero 774beon-gil, Namdong-gu, Incheon, 21565, Republic of Korea.
| |
Collapse
|
33
|
Spinelli SMC, Mantovani MDF, Madalozzo Schieferdecker ME, Paes RG. Nutritional diagnoses and interventions in people with metabolic dysfunction-associated fatty liver disease:Cross-sectional study. CLINICAL NUTRITION OPEN SCIENCE 2024; 57:266-274. [DOI: 10.1016/j.nutos.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
|
34
|
Kim MN, Han JW, An J, Kim BK, Jin YJ, Kim SS, Lee M, Lee HA, Cho Y, Kim HY, Shin YR, Yu JH, Kim MY, Choi Y, Chon YE, Cho EJ, Lee EJ, Kim SG, Kim W, Jun DW, Kim SU, on behalf of The Korean Association for the Study of the Liver (KASL). KASL clinical practice guidelines for noninvasive tests to assess liver fibrosis in chronic liver disease. Clin Mol Hepatol 2024; 30:S5-S105. [PMID: 39159947 PMCID: PMC11493350 DOI: 10.3350/cmh.2024.0506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024] Open
Affiliation(s)
- Mi Na Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Ji Won Han
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jihyun An
- Department of Gastroenterology and Hepatology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - Young-Joo Jin
- Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Seung-seob Kim
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Minjong Lee
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Han Ah Lee
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Yuri Cho
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, Korea
| | - Hee Yeon Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yu Rim Shin
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Hwan Yu
- Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Moon Young Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - YoungRok Choi
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Young Eun Chon
- Department of Internal Medicine, Institute of Gastroenterology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Eun Ju Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Joo Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Gyune Kim
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Won Kim
- Department of Internal Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Dae Won Jun
- Department of Internal Medicine, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
| | - on behalf of The Korean Association for the Study of the Liver (KASL)
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Gastroenterology and Hepatology, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
- Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
- Center for Liver and Pancreatobiliary Cancer, National Cancer Center, Goyang, Korea
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Bucheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Thoracic and Cardiovascular Surgery, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Institute of Gastroenterology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
- Department of Internal Medicine, Seoul Metropolitan Government Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Hanyang University Hospital, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Yuan W, Ran Y, Wang J, Pei F, Cui L, Chen S, Wu S, Zhou L. Mediating effect of diabetes on the relationship between nonalcoholic fatty liver disease and atherosclerotic cardiovascular disease: a prospective cohort study. Eur J Gastroenterol Hepatol 2024; 36:1133-1140. [PMID: 39101442 DOI: 10.1097/meg.0000000000002794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
OBJECTIVE This study explored the mediating effect of diabetes on the relationship between nonalcoholic fatty liver disease (NAFLD) and atherosclerotic cardiovascular disease (ASCVD). METHODS In this prospective community cohort study, 82 975 participants were enrolled, with the primary outcome being the incidence of new-onset ASCVD. Using the Cox proportional hazards model, the hazard ratio (HR) and 95% confidence interval (CI) for ASCVD occurrence were computed between NAFLD and non-NAFLD groups. The correlation between NAFLD and diabetes was assessed using a binary logistic regression model, and that between NAFLD, diabetes and ASCVD using a mediation model. RESULTS During follow-up, 9471 ASCVD cases were observed. Compared with individuals without NAFLD, those with NAFLD showed an increased ASCVD risk (HR: 1.424; 95% CI: 1.363-1.488; P < 0.001). Stratifying NAFLD based on metabolic subphenotypes revealed a higher ASCVD risk in the NAFLD combined with diabetes subgroup than in the non-NAFLD subgroup (HR: 1.960; 95% CI: 1.817-2.115; P < 0.001). NAFLD was positively associated with baseline diabetes (odds ratio: 2.983; 95% CI: 2.813-3.163; P < 0.001). Furthermore, NAFLD severity was positively correlated with diabetes risk. Mediation analysis indicated that diabetes partially mediated the effect of NAFLD on ASCVD incidence, accounting for 20.33% of the total effect. CONCLUSION NAFLD is an independent predictor of increased ASCVD risk, which may be slightly mediated by diabetes in patients with NAFLD. Evaluating NAFLD and diabetes may be crucial in the early screening and prevention of ASCVD.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University
- Tianjin Institute of Digestive Diseases
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, Departments of
- Rheumatology and Immunology
| | - Ying Ran
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University
- Tianjin Institute of Digestive Diseases
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, Departments of
| | | | | | | | - Shuohua Chen
- Cardiology, Kailuan General Hospital, Tangshan, China
| | - Shouling Wu
- Cardiology, Kailuan General Hospital, Tangshan, China
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University
- Tianjin Institute of Digestive Diseases
- Tianjin Key Laboratory of Digestive Diseases, Tianjin, Departments of
| |
Collapse
|
36
|
Taylor R. Understanding the cause of type 2 diabetes. Lancet Diabetes Endocrinol 2024; 12:664-673. [PMID: 39038473 DOI: 10.1016/s2213-8587(24)00157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 07/24/2024]
Abstract
Type 2 diabetes has long been thought to have heterogenous causes, even though epidemiological studies uniformly show a tight relationship with overnutrition. The twin cycle hypothesis postulated that interaction of self-reinforcing cycles of fat accumulation inside the liver and pancreas, driven by modest but chronic positive calorie balance, could explain the development of type 2 diabetes. This hypothesis predicted that substantial weight loss would bring about a return to the non-diabetic state, permitting observation of the pathophysiology determining the transition. These changes were postulated to reflect the basic mechanisms of causation in reverse. A series of studies over the past 15 years has elucidated these underlying mechanisms. Together with other research, the interaction of environmental and genetic factors has been clarified. This knowledge has led to successful implementation of a national programme for remission of type 2 diabetes. This Review discusses the paucity of evidence for heterogeneity in causes of type 2 diabetes and summarises the in vivo pathophysiological changes, which cause this disease of overnutrition. Type 2 diabetes has a homogenous cause expressed in genetically heterogenous individuals.
Collapse
Affiliation(s)
- Roy Taylor
- Newcastle Magnetic Resonance Centre, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
37
|
Willis SA, Malaikah S, Bawden SJ, Sherry AP, Sargeant JA, Coull NA, Bradley CR, Rowlands A, Naim I, Ennequin G, Yates T, Waheed G, Gowland P, Stensel DJ, Webb DR, Davies MJ, Aithal GP, King JA. Greater hepatic lipid saturation is associated with impaired glycaemic regulation in men with metabolic dysfunction-associated steatotic liver disease but is not altered by 6 weeks of exercise training. Diabetes Obes Metab 2024; 26:4030-4042. [PMID: 38978184 DOI: 10.1111/dom.15755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/05/2024] [Accepted: 06/14/2024] [Indexed: 07/10/2024]
Abstract
AIMS To examine the impact of impaired glycaemic regulation (IGR) and exercise training on hepatic lipid composition in men with metabolic dysfunction-associated steatotic liver disease (MASLD). MATERIALS AND METHODS In Part A (cross-sectional design), 40 men with MASLD (liver proton density fat fraction [PDFF] ≥5.56%) were recruited to one of two groups: (1) normal glycaemic regulation (NGR) group (glycated haemoglobin [HbA1c] < 42 mmol∙mol-1 [<6.0%]; n = 14) or (2) IGR group (HbA1c ≥ 42 mmol∙mol-1 [≥6.0%]; n = 26). In Part B (randomized controlled trial design), participants in the IGR group were randomized to one of two 6-week interventions: (1) exercise training (EX; 70%-75% maximum heart rate; four sessions/week; n = 13) or (2) non-exercise control (CON; n = 13). Saturated (SI; primary outcome), unsaturated (UI) and polyunsaturated (PUI) hepatic lipid indices were determined using proton magnetic resonance spectroscopy. Additional secondary outcomes included liver PDFF, HbA1c, fasting plasma glucose (FPG), homeostatic model assessment of insulin resistance (HOMA-IR), peak oxygen uptake (VO2 peak), and plasma cytokeratin-18 (CK18) M65, among others. RESULTS In Part A, hepatic SI was higher and hepatic UI was lower in the IGR versus the NGR group (p = 0.038), and this hepatic lipid profile was associated with higher HbA1c levels, FPG levels, HOMA-IR and plasma CK18 M65 levels (rs ≥0.320). In Part B, hepatic lipid composition and liver PDFF were unchanged after EX versus CON (p ≥ 0.257), while FPG was reduced and VO2 peak was increased (p ≤ 0.030). ΔVO2 peak was inversely associated with Δhepatic SI (r = -0.433) and positively associated with Δhepatic UI and Δhepatic PUI (r ≥ 0.433). CONCLUSIONS Impaired glycaemic regulation in MASLD is characterized by greater hepatic lipid saturation; however, this composition is not altered by 6 weeks of moderate-intensity exercise training.
Collapse
Affiliation(s)
- Scott A Willis
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
| | - Sundus Malaikah
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Clinical Nutrition Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Stephen J Bawden
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Aron P Sherry
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham, UK
| | - Jack A Sargeant
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Leicester Diabetes Centre, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Nicole A Coull
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Christopher R Bradley
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Alex Rowlands
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Iyad Naim
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - Gaël Ennequin
- Laboratory of Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Université of Clermont Auvergne, Clermont-Ferrand, France
| | - Thomas Yates
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Ghazala Waheed
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Penny Gowland
- Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
| | - David J Stensel
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Faculty of Sport Sciences, Waseda University, Tokorozawa, Japan
| | - David R Webb
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Melanie J Davies
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Guruprasad P Aithal
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - James A King
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
| |
Collapse
|
38
|
Suzuki M, Hayashi T, Nashiki K, Kawata H, Nagata S, Abe T. Influence of Gd-EOB-DTPA on proton-density fat fraction in the liver using chemical shift-encoded magnetic resonance imaging at 3-T. Radiol Phys Technol 2024; 17:637-644. [PMID: 38730134 DOI: 10.1007/s12194-024-00811-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its advanced stage, non-alcoholic steatohepatitis (NASH), have become increasingly prevalent owing to the rise in metabolic syndromes. Accurate assessment of hepatic fat deposition and inflammation is crucial for diagnosing and managing NAFLD/NASH. We investigated the influence of Gd-EOB-DTPA, (EOB) on proton-density fat fraction (PDFF) measurements using chemical shift-encoded magnetic resonance imaging (CSE-MRI) at 3-T. In total, 431 patients who underwent EOB contrast-enhanced MRI were included. PDFF measurements were obtained from pre- and post-contrast CSE-MRI. Linear regression and Bland-Altman analyses were performed to assess the correlation and agreement between pre- and post-EOB PDFF measurements. Relative enhancement (RE) of the liver was calculated as an EOB uptake index. There was a significant decrease in PDFF following EOB administration compared with the pre-contrast values (P < 0.0001), which was observed across all PDFF ranges (< 10% and ≥ 10%). Linear regression analysis revealed high correlation between pre- and post-EOB PDFF measurements. Bland-Altman analysis indicated a small bias between pre- and post-EOB PDFF values. Subgroup analysis based on RE showed a significant difference in ΔPDFF between patients with high RE (> 120%) and those with lower RE levels. EOB administration resulted in a slight decrease in PDFF measurements obtained using CSE-MRI at 3-T. We were able to generalize and clarify that the PDFF of the liver on 3D CSE-MRI at 3-T was slightly decreased after EOB administration as we used a larger group of patients compared to previous studies.
Collapse
Affiliation(s)
- Makoto Suzuki
- Department of Radiology, Kurume University Hospital, Asahimachi 67, Kurume, Japan.
| | - Tatsuya Hayashi
- Department of Radiological Technology, Faculty of Medical Technology, Teikyo University, 2-11-1, Kaga, Itabashi-Ku, Tokyo, Japan
| | - Kazutaka Nashiki
- Department of Radiology, Kurume University Hospital, Asahimachi 67, Kurume, Japan
| | - Hidemichi Kawata
- Department of Radiology, Kurume University Hospital, Asahimachi 67, Kurume, Japan
| | - Shuji Nagata
- Department of Radiology, Kurume University School of Medicine, Asahimachi 67, Kurume, Japan
| | - Toshi Abe
- Department of Radiology, Kurume University School of Medicine, Asahimachi 67, Kurume, Japan
| |
Collapse
|
39
|
Lietzén MS, Mari A, Ojala R, Hentilä J, Koskensalo K, Lautamäki R, Löyttyniemi E, Parkkola R, Saunavaara V, Kirjavainen AK, Rajander J, Malm T, Lahti L, Rinne JO, Pietiläinen KH, Iozzo P, Hannukainen JC. Effects of Obesity and Exercise on Hepatic and Pancreatic Lipid Content and Glucose Metabolism: PET Studies in Twins Discordant for BMI. Biomolecules 2024; 14:1070. [PMID: 39334836 PMCID: PMC11430379 DOI: 10.3390/biom14091070] [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: 08/06/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Obesity and sedentarism are associated with increased liver and pancreatic fat content (LFC and PFC, respectively) as well as impaired organ metabolism. Exercise training is known to decrease organ ectopic fat but its effects on organ metabolism are unclear. Genetic background affects susceptibility to obesity and the response to training. We studied the effects of regular exercise training on LFC, PFC, and metabolism in monozygotic twin pairs discordant for BMI. We recruited 12 BMI-discordant monozygotic twin pairs (age 40.4, SD 4.5 years; BMI 32.9, SD 7.6, 8 female pairs). Ten pairs completed six months of training intervention. We measured hepatic insulin-stimulated glucose uptake using [18F]FDG-PET and fat content using magnetic resonance spectroscopy before and after the intervention. At baseline LFC, PFC, gamma-glutamyl transferase (GT), and hepatic glucose uptake were significantly higher in the heavier twins compared to the leaner co-twins (p = 0.018, p = 0.02 and p = 0.01, respectively). Response to training in liver glucose uptake and GT differed between the twins (Time*group p = 0.04 and p = 0.004, respectively). Liver glucose uptake tended to decrease, and GT decreased only in the heavier twins (p = 0.032). In BMI-discordant twins, heavier twins showed higher LFC and PFC, which may underlie the observed increase in liver glucose uptake and GT. These alterations were mitigated by exercise. The small number of participants makes the results preliminary, and future research with a larger pool of participants is warranted.
Collapse
Affiliation(s)
| | - Andrea Mari
- Institute of Neuroscience, National Research Council (CNR), 35128 Padua, Italy
| | - Ronja Ojala
- Turku PET Centre, University of Turku, 20521 Turku, Finland
| | - Jaakko Hentilä
- Turku PET Centre, University of Turku, 20521 Turku, Finland
| | - Kalle Koskensalo
- Department of Medical Physics, Turku University Hospital, 20520 Turku, Finland
| | | | | | - Riitta Parkkola
- Department of Radiology, Turku University Hospital and University of Turku, 20520 Turku, Finland
| | - Virva Saunavaara
- Turku PET Centre, University of Turku, 20521 Turku, Finland
- Department of Medical Physics, Turku University Hospital, 20520 Turku, Finland
| | - Anna K Kirjavainen
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, 20521 Turku, Finland
| | - Johan Rajander
- Turku PET Centre, Accelerator Laboratory, Åbo Akademi University, 20500 Turku, Finland
| | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, 20521 Turku, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku, 20521 Turku, Finland
- Turku PET Centre, Turku University Hospital, 20520 Turku, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Abdominal Center, Obesity Center, Endocrinology, University of Helsinki and Helsinki University Central Hospital, 00014 Helsinki, Finland
| | - Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), 56124 Pisa, Italy
| | | |
Collapse
|
40
|
Habib S. Team players in the pathogenesis of metabolic dysfunctions-associated steatotic liver disease: The basis of development of pharmacotherapy. World J Gastrointest Pathophysiol 2024; 15:93606. [PMID: 39220834 PMCID: PMC11362842 DOI: 10.4291/wjgp.v15.i4.93606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 08/22/2024] Open
Abstract
Nutrient metabolism is regulated by several factors. Social determinants of health with or without genetics are the primary regulator of metabolism, and an unhealthy lifestyle affects all modulators and mediators, leading to the adaptation and finally to the exhaustion of cellular functions. Hepatic steatosis is defined by presence of fat in more than 5% of hepatocytes. In hepatocytes, fat is stored as triglycerides in lipid droplet. Hepatic steatosis results from a combination of multiple intracellular processes. In a healthy individual nutrient metabolism is regulated at several steps. It ranges from the selection of nutrients in a grocery store to the last step of consumption of ATP as an energy or as a building block of a cell as structural component. Several hormones, peptides, and genes have been described that participate in nutrient metabolism. Several enzymes participate in each nutrient metabolism as described above from ingestion to generation of ATP. As of now several publications have revealed very intricate regulation of nutrient metabolism, where most of the regulatory factors are tied to each other bidirectionally, making it difficult to comprehend chronological sequence of events. Insulin hormone is the primary regulator of all nutrients' metabolism both in prandial and fasting states. Insulin exerts its effects directly and indirectly on enzymes involved in the three main cellular function processes; metabolic, inflammation and repair, and cell growth and regeneration. Final regulators that control the enzymatic functions through stimulation or suppression of a cell are nuclear receptors in especially farnesoid X receptor and peroxisome proliferator-activated receptor/RXR ligands, adiponectin, leptin, and adiponutrin. Insulin hormone has direct effect on these final modulators. Whereas blood glucose level, serum lipids, incretin hormones, bile acids in conjunction with microbiota are intermediary modulators which are controlled by lifestyle. The purpose of this review is to overview the key players in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) that help us understand the disease natural course, risk stratification, role of lifestyle and pharmacotherapy in each individual patient with MASLD to achieve personalized care and target the practice of precision medicine. PubMed and Google Scholar databases were used to identify publication related to metabolism of carbohydrate and fat in states of health and disease states; MASLD, cardiovascular disease and cancer. More than 1000 publications including original research and review papers were reviewed.
Collapse
Affiliation(s)
- Shahid Habib
- Department of Hepatology, Liver Institute PLLC, Tucson, AZ 85712, United States
| |
Collapse
|
41
|
Sommer-Ballarini M, Nguyen TH, Pletsch-Borba L, Wernicke C, Tacke F, Schwerdtle T, Pellowski D, Machann J, Spranger J, Wirth EK, Mai K. Impact of peripheral thyroid hormone balance on liver fat: insights from the NutriAct trial. Eur J Endocrinol 2024; 191:183-191. [PMID: 39049801 DOI: 10.1093/ejendo/lvae093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE Hypothyroidism has been proposed as a potential contributor to steatotic liver disease (SLD), but existing data shows conflicting results in euthyroid subjects. Therefore, we investigated the association between thyroid function and intrahepatic lipids (IHLs) during a 36-month randomized controlled trial evaluating a diet known to reduce liver fat. DESIGN 502 eligible subjects (aged 50-80 years, ≥1 risk factor for unhealthy aging) were randomly assigned to either follow a diet rich in unsaturated fatty acids, plant protein, and fiber (intervention group, IG), or dietary recommendations of the German Nutrition Society (control group, CG). METHODS Serum levels of thyroid hormones (THs) as well as IHLs, defined via magnetic resonance spectroscopy, were measured within an euthyroid subgroup without significant alcohol consumption at baseline (n = 332) and after 12 months (n = 243). A ratio of T3/T4 was used to assess whole-body deiodinase activity. Estimates of glucose and lipid metabolism were analyzed. RESULTS Only fT3 and T3/T4 ratios showed a significant positive correlation with IHL at baseline. We observed a significant decline in fT3, T3, fT3/fT4 ratio, and T3/T4 ratio in CG and IG after 12 months without significant differences between groups. TSH, fT4, and T4 remained stable. A larger improvement of IHL during dietary intervention was seen in those subjects with a lower decline in T3 concentrations. CONCLUSIONS Altered TH balance indicates a possible compensatory upregulation of whole-body TH activity in subjects with increased liver fat. This might be also relevant during the improvement of hepatic steatosis.
Collapse
Affiliation(s)
- Miriam Sommer-Ballarini
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, 10115 Berlin, Germany
| | - Thu-Huong Nguyen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, 10115 Berlin, Germany
| | - Laura Pletsch-Borba
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, 10115 Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Charlotte Wernicke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, 10115 Berlin, Germany
| | - Frank Tacke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), 10115 Berlin, Germany
| | - Tanja Schwerdtle
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, 14558 Nuthetal, Germany
- German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany
| | - Denny Pellowski
- TraceAge-DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, 14558 Nuthetal, Germany
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany
- Institute of Nutritional Science, Department Food Chemistry, University of Potsdam,14469 Potsdam, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076 Tübingen, Germany
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Joachim Spranger
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, 10115 Berlin, Germany
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10115 Berlin, Germany
| | - Eva Katrin Wirth
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, 10115 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10115 Berlin, Germany
| | - Knut Mai
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Endocrinology and Metabolism, 10115 Berlin, Germany
- NutriAct-Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany
- German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 10115 Berlin, Germany
- Department of Human Nutrition, German Institute of Human Nutrition, Potsdam-Rehbruecke, 14558 Nuthetal, Germany
| |
Collapse
|
42
|
Duan Y, Yang Y, Zhao S, Bai Y, Yao W, Gao X, Yin J. Crosstalk in extrahepatic and hepatic system in NAFLD/NASH. Liver Int 2024; 44:1856-1871. [PMID: 38717072 DOI: 10.1111/liv.15967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/28/2024] [Accepted: 04/26/2024] [Indexed: 07/17/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as the most prevalent chronic liver disease globally. Non-alcoholic steatohepatitis (NASH) represents an extremely progressive form of NAFLD, which, without timely intervention, may progress to cirrhosis or hepatocellular carcinoma. Presently, a definitive comprehension of the pathogenesis of NAFLD/NASH eludes us, and pharmacological interventions targeting NASH specifically remain constrained. The aetiology of NAFLD encompasses a myriad of external factors including environmental influences, dietary habits and gender disparities. More significantly, inter-organ and cellular interactions within the human body play a role in the development or regression of the disease. In this review, we categorize the influences affecting NAFLD both intra- and extrahepatically, elaborating meticulously on the mechanisms governing the onset and progression of NAFLD/NASH. This exploration delves into progress in aetiology and promising therapeutic targets. As a metabolic disorder, the development of NAFLD involves complexities related to nutrient metabolism, liver-gut axis interactions and insulin resistance, among other regulatory functions of extraneous organs. It further encompasses intra-hepatic interactions among hepatic cells, Kupffer cells (KCs) and hepatic stellate cells (HSCs). A comprehensive understanding of the pathogenesis of NAFLD/NASH from a macroscopic standpoint is instrumental in the formulation of future therapies for NASH.
Collapse
Affiliation(s)
- Yiliang Duan
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yan Yang
- The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Shuqiang Zhao
- Jiangsu Institute for Food and Drug Control, NMPA Key Laboratory for Impurity Profile of Chemical Drugs, Nanjing, Jiangsu, China
| | - Yuesong Bai
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenbing Yao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jun Yin
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
43
|
Schenker RB, Machle CJ, Schmidt KA, Allayee H, Kohli R, Goran MI. Associations of dietary sugars with liver stiffness in Latino adolescents with obesity differ on PNPLA3 and liver disease severity. Liver Int 2024; 44:1768-1774. [PMID: 38634702 PMCID: PMC11251846 DOI: 10.1111/liv.15946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common paediatric liver disease. Latinos have high MASLD risk due to 50% prevalence of GG genotype of PNPLA3. Our primary aim was to evaluate associations between dietary carbohydrates/sugars and liver stiffness in Latino adolescents with obesity. Our secondary aim was to examine effect modification by (a) PNPLA3 genotype or (b) liver disease severity. Data were obtained from 114 Latino adolescents with obesity involved in two prior studies. No associations were seen between dietary carbohydrates/sugars and liver stiffness in the group as a whole. In subjects with GG genotype of PNPLA3, total sugar, fructose, sucrose, and glucose were associated with liver stiffness. Positive relationships between carbohydrate, total sugar, and sucrose and liver stiffness were stronger in those with MASLD and fibrosis compared to those with healthy livers and MASLD without fibrosis.
Collapse
Affiliation(s)
- Rachel B Schenker
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Christopher J Machle
- Department of Pediatrics, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
- Department of Psychology, University of Oregon, Eugene, Oregon, USA
| | - Kelsey A Schmidt
- Department of Pediatrics, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Hooman Allayee
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, University of Southern California, Los Angeles, California, USA
| | - Rohit Kohli
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Michael I Goran
- Department of Pediatrics, Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, USA
| |
Collapse
|
44
|
Horn F, Ittermann T, Kromrey ML, Seppelt D, Völzke H, Kühn JP, Schön F. Exploring factors associated with non-alcoholic fatty liver disease using longitudinal MRI. BMC Gastroenterol 2024; 24:229. [PMID: 39044153 PMCID: PMC11267668 DOI: 10.1186/s12876-024-03300-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND To identify factors associated with non-alcoholic fatty liver disease over a 5-year period. METHODS Three hundred seven participants, including 165 women, with a mean age of 55.6 ± 12.0 years underwent continuous quantitative MRI of the liver using the proton-density fat fraction (PDFF). The liver's fat fractions were determined at baseline and 5 years later, and the frequency of participants who developed fatty liver disease and potential influencing factors were explored. Based on significant factors, a model was generated to predict the development of fatty liver disease. RESULTS After excluding participants with pre-existing fatty liver, the baseline PDFF of 3.1 ± 0.9% (n = 190) significantly increased to 7.67 ± 3.39% within 5 years (p < 0.001). At baseline, age (OR = 1.04, p = 0.006, CI = 1.01-1.07), BMI (OR = 1.11, p = 0.041, CI = 1.01-1.23), and waist circumference (OR = 1.05, p = 0.020, CI = 1.01-1.09) were identified as risk factors. Physical activity was negatively associated (OR = 0.43, p = 0.049, CI = 0.18-0.99). In the prediction model, age, physical activity, diabetes mellitus, diastolic blood pressure, and HDL-cholesterol remained as independent variables. Combining these risk factors to predict the development of fatty liver disease revealed an AUC of 0.7434. CONCLUSIONS Within a five-year follow-up, one-quarter of participants developed fatty liver disease influenced by the triggering factors of age, diabetes mellitus, low HDL-cholesterol, and diastolic blood pressure. Increased physical activity has a protective effect on the development of fatty liver.
Collapse
Affiliation(s)
- Friedrich Horn
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Till Ittermann
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Marie-Luise Kromrey
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
- Institute and Policlinic for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Danilo Seppelt
- Institute and Policlinic for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Jens-Peter Kühn
- Institute and Policlinic for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | - Felix Schön
- Institute and Policlinic for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
45
|
Nakamura A, Yoshimura T, Ichikawa T, Okuyama K. Prognostic significance of low hepatic fat content in advanced chronic liver disease: MRI-PDFF insights. Ann Hepatol 2024; 29:101507. [PMID: 38723748 DOI: 10.1016/j.aohep.2024.101507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/19/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION AND OBJECTIVES The mechanisms of hepatic fat loss in late-stage metabolic dysfunction-associated fatty liver disease (MASLD) are enigmatic and the prognostic significance of low hepatic fat content (LHF) in chronic liver disease (CLD) is unknown. Proton density fat fraction (PDFF), measured by magnetic resonance imaging (MRI), is considered the most accurate noninvasive method for quantifying hepatic fat content. This study aimed to address these issues by evaluating PDFF. PATIENTS AND METHODS This is a single-center, retrospective study involving 762 patients with CLD, measuring liver stiffness (LS) using MR elastography and PDFF using MRI. LHF was defined as a PDFF ≤ 2.7 % and hepatic reserve function was assessed using the albumin-bilirubin (ALBI) score. Multivariate analysis explored associations between variables. RESULTS LHF was 27 % in the entire cohort, and PDFF was significantly decreased with LS ≥ 5.5 kPa (p < 0.05). On the multivariate analysis, low body mass index and ALBI score were independently associated with LHF (p < 0.05). In advanced CLD (n = 288), ALBI score and PDFF showed a significant negative correlation regardless of etiology (MASLD/non-MASLD: r= -0.613/-0.233), and the prevalence of LHF increased with progression of ALBI grade (p < 0.01 each). In addition, lower PDFF was associated with increased liver-related and all-cause mortality (p < 0.01), and Cox proportional hazards models extracted LHF as an independent prognostic factor, along with ALBI score and hepatocellular carcinoma (p < 0.05 each). CONCLUSIONS In ACLD, hepatic reserve dysfunction contributed to hepatic fat loss independent of nutritional status, suggesting that LHF may be a poor prognostic factor in all etiologies.
Collapse
Affiliation(s)
- Atsushi Nakamura
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan.
| | - Tsubasa Yoshimura
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| | - Takeshi Ichikawa
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| | - Keiji Okuyama
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| |
Collapse
|
46
|
Richter MM, Kemp IM, Heebøll S, Winther-Sørensen M, Kjeldsen SAS, Jensen NJ, Nybing JD, Linden FH, Høgh-Schmidt E, Boesen MP, Madsbad S, Schiødt FV, Nørgaard K, Schmidt S, Gluud LL, Haugaard SB, Holst JJ, Nielsen S, Rungby J, Wewer Albrechtsen NJ. Glucagon augments the secretion of FGF21 and GDF15 in MASLD by indirect mechanisms. Metabolism 2024; 156:155915. [PMID: 38631460 DOI: 10.1016/j.metabol.2024.155915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION Glucagon receptor agonism is currently explored for the treatment of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD). The metabolic effects of glucagon receptor agonism may in part be mediated by increases in circulating levels of Fibroblast Growth Factor 21 (FGF21) and Growth Differentiation Factor 15 (GDF15). The effect of glucagon agonism on FGF21 and GDF15 levels remains uncertain, especially in the context of elevated insulin levels commonly observed in metabolic diseases. METHODS We investigated the effect of a single bolus of glucagon and a continuous infusion of glucagon on plasma concentrations of FGF21 and GDF15 in conditions of endogenous low or high insulin levels. The studies included individuals with overweight with and without MASLD, healthy controls (CON) and individuals with type 1 diabetes (T1D). The direct effect of glucagon on FGF21 and GDF15 was evaluated using our in-house developed isolated perfused mouse liver model. RESULTS FGF21 and GDF15 correlated with plasma levels of insulin, but not glucagon, and their secretion was highly increased in MASLD compared with CON and T1D. Furthermore, FGF21 levels in individuals with overweight with or without MASLD did not increase after glucagon stimulation when insulin levels were kept constant. FGF21 and GDF15 levels were unaffected by direct stimulation with glucagon in the isolated perfused mouse liver. CONCLUSION The glucagon-induced secretion of FGF21 and GDF15 is augmented in MASLD and may depend on insulin. Thus, glucagon receptor agonism may augment its metabolic benefits in patients with MASLD through enhanced secretion of FGF21 and GDF15.
Collapse
Affiliation(s)
- Michael M Richter
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Ida M Kemp
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sara Heebøll
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus 8200, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Marie Winther-Sørensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sasha A S Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Nicole J Jensen
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Janus D Nybing
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Frederik H Linden
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Erik Høgh-Schmidt
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Mikael P Boesen
- Department of Radiology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital - Hvidovre, Hvidovre 2650, Denmark
| | - Frank Vinholt Schiødt
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kirsten Nørgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Signe Schmidt
- Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Lise Lotte Gluud
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Gastro Unit, Copenhagen University Hospital - Hvidovre, Hvidovre 2650, Denmark
| | - Steen B Haugaard
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Søren Nielsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Medicine, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jørgen Rungby
- Department of Endocrinology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Nicolai J Wewer Albrechtsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen 2400, Denmark; Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
47
|
Garza AL, Lee M, Blangero J, Bauer CX, Czerwinski SA, Choh AC. Genetic correlations between liver fat content, metabolic health, and adiposity distribution in the Fels Longitudinal Study. Nutr Metab Cardiovasc Dis 2024; 34:1610-1618. [PMID: 38555241 DOI: 10.1016/j.numecd.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/13/2024] [Accepted: 03/01/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND AND AIMS Hepatic steatosis is known to be heritable, but its genetic basis is mostly uncharacterized. Steatosis is associated with metabolic and adiposity features; recent studies hypothesize that shared genetic effects between these traits could account for some of the unexplained heritability. This study aimed to quantify these genetic associations in a family-based sample of non-Hispanic white adults. METHODS AND RESULTS 704 participants (18-95 years, 55.8% female) from the Fels Longitudinal Study with an MRI assessment of liver fat were included. Quantitative genetic analyses estimated the age- and sex-adjusted heritability of individual traits and the genetic correlations within trait pairs. Mean liver fat was 5.95% (SE = 0.23) and steatosis (liver fat >5.56%) was present in 29.8% of participants. Heritability (h2± SE) of steatosis was 0.72 ± 0.17 (p = 6.80e-6). All other traits including liver enzymes, fasting glucose, HOMA-IR, visceral and subcutaneous adipose tissue (VAT, SAT), body mass index, body fat percent, waist circumference, lipids and blood pressure were also heritable. Significant genetic correlations were found between liver fat and all traits except aspartate aminotransferase (AST), and among most trait pairs. Highest genetic correlations were between liver fat and HOMA-IR (0.85 ± 0.08, p = 1.73e-8), fasting glucose and ALT (0.89 ± 0.26, p = 6.68e-5), and HOMA-IR with: waist circumference (0.81 ± 0.12, p = 3.76e-6), body fat percent (0.78 ± 0.12 p = 2.42e-5) and VAT (0.73 ± 0.07, p = 6.37e-8). CONCLUSIONS Common genes may exist between liver fat accumulation, metabolic features and adiposity phenotypes.
Collapse
Affiliation(s)
- Ariana L Garza
- UTHealth Houston School of Public Health, Brownsville, TX, United States.
| | - Miryoung Lee
- Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth Houston School of Public Health, Brownsville, TX, United States
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, School of Medicine, Brownsville, TX, United States
| | - Cici X Bauer
- Division of Biostatistics, UTHealth Houston School of Public Health, Houston, TX, United States
| | - Stefan A Czerwinski
- School of Health and Rehabilitation Sciences, College of Medicine, Ohio State University, Columbus, OH, United States
| | - Audrey C Choh
- Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth Houston School of Public Health, Brownsville, TX, United States
| |
Collapse
|
48
|
Yin H, Fan Y, Yu J, Xiong B, Zhou B, Sun Y, Wang L, Zhu Y, Xu H. Quantitative US fat fraction for noninvasive assessment of hepatic steatosis in suspected metabolic-associated fatty liver disease. Insights Imaging 2024; 15:159. [PMID: 38902550 PMCID: PMC11190099 DOI: 10.1186/s13244-024-01728-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 05/19/2024] [Indexed: 06/22/2024] Open
Abstract
OBJECTIVES To evaluate the agreement between quantitative ultrasound system fat fraction (USFF) and proton magnetic resonance spectroscopy (1H-MRS) and the diagnostic value of USFF in assessing metabolic-associated fatty liver disease (MAFLD). METHODS The participants with or suspected of MAFLD were prospectively recruited and underwent 1H-MRS, USFF, and controlled attenuation parameter (CAP) measurements. The correlation between USFF and 1H-MRS was assessed using Pearson correlation coefficients. The USFF diagnostic performance for different grades of steatosis was evaluated using receiver operating characteristic curve analysis (ROC) and was compared with CAP, visual hepatic steatosis grade (VHSG). RESULTS A total of 113 participants (mean age 44.79 years ± 13.56 (SD); 71 males) were enrolled, of whom 98 (86.73%) had hepatic steatosis (1H-MRS ≥ 5.56%). USFF showed a good correlation (Pearson r = 0.76) with 1H-MRS and showed a linear relationship, which was superior to the correlation between CAP and 1H-MRS (Pearson r = 0.61). The USFF provided high diagnostic performance for different grades of hepatic steatosis, with ROC from 0.84 to 0.98, and the diagnostic performance was better than that of the CAP and the VHSG. The cut-off values of the USFF were different for various grades of steatosis, and the cut-off values for S1, S2, and S3 were 12.01%, 19.98%, and 22.22%, respectively. CONCLUSIONS There was a good correlation between USFF and 1H-MRS. Meanwhile, USFF had good diagnostic performance for hepatic steatosis and was superior to CAP and VHSG. USFF represents a superior method for noninvasive quantitative assessment of MAFLD. CRITICAL RELEVANCE STATEMENT Quantitative ultrasound system fat fraction (USFF) accurately assesses liver fat content and has a good correlation with magnetic resonance spectroscopy (1H-MRS) for the assessment of metabolic-associated fatty liver disease (MAFLD), as well as for providing an accurate quantitative assessment of hepatic steatosis. KEY POINTS Current diagnostic and monitoring modalities for metabolic-associated fatty liver disease have limitations. USFF correlated well with 1H-MRS and was superior to the CAP. USFF has good diagnostic performance for steatosis, superior to CAP and VHSG.
Collapse
Affiliation(s)
- Haohao Yin
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Yunling Fan
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Jifeng Yu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Bing Xiong
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China
| | - Boyang Zhou
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Yikang Sun
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Lifan Wang
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China
| | - Yuli Zhu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China.
| | - Huixiong Xu
- Department of Ultrasound, Zhongshan Hospital, Institute of Ultrasound in Medicine and Engineering, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
49
|
Yao S, Wei Y, Ye Z, Chen J, Duan T, Zhang Z, Song B. Hepatic Steatosis Has No Effect in Diagnosis Accuracy of LI-RADS v2018 Categorization of Hepatocellular Carcinoma in MR Imaging. J Magn Reson Imaging 2024; 59:2060-2070. [PMID: 34121266 DOI: 10.1002/jmri.27783] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND In clinical practice, hepatocellular carcinoma (HCC) is widely diagnosed by using MRI, however, whether the imaging features are affected by hepatic steatosis (HS) is still unknown. PURPOSE To investigate and compare the differences in HCC related imaging features between with- and without-HS groups, and to further determine whether HS affects the diagnosis accuracy of Liver Imaging Reporting and Data System (LI-RADS) v2018 of HCC in MRI. STUDY TYPE Prospective. SUBJECTS One hundred and seventy-one patients (mean age, 52 ± 11 years; range, 26-83 years) including 137 men and 34 women. FIELD STRENGTH/SEQUENCE 3.0 T, gradient echo (GRE). ASSESSMENT Subjects were classified as HS and non-HS groups according to MRI-proton density fat-fraction (PDFF). HS was defined as MRI-PDFF >5.6%. Three radiologists accessed HCC features and assigned LI-RADS categories in MRI independently based on LI-RADS v2018. Frequencies of HCC major features and LR categorization assignment between the two groups as well as interobserver agreement between the two radiologists were assessed. STATISTICAL TESTS Unpaired t-test, Chi-square test, Fisher's exact test, kappa statistic, intraclass correlation coefficient (ICC). A two-sided P value <0.05 was considered as statistically significant. RESULTS Major features including arterial hyperenhancement (APHE), enhancing "capsule" and nonperipheral "washout" observed between HS and non-HS groups were not significantly different (78.95% vs.78.62%, P = 0.866; 57.89% vs.52.98%, P = 0.483; and 75% vs.81.46%, P = 0.257, respectively), and the assessment of observation size showed a borderline difference (P = 0.059). No significant difference in LR-5 assignment between the two groups (69.74% vs. 72.85% for reader 1, P = 0.641; 71.05% vs. 72.19% for reader 2, P = 0.877). Interobserver agreement between the two radiologists showed almost perfect in LR-5 assignment (κ = 0.869) and size observation (ICC = 0.997). DATA CONCLUSION The diagnosis of HCC based on LI-RADS v2018 in MRI is of comparable performance regardless of HS, in which there is no significant difference in either the major imaging features or LR categorization. LEVEL OF EVIDENCE 2 Technical Efficacy Stage: 2.
Collapse
Affiliation(s)
- Shan Yao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Wei
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Zheng Ye
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Chen
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Duan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Brouwers MCGJ, Cassiman D. Rare monogenic causes of steatotic liver disease masquerading as MASLD. J Hepatol 2024; 80:e252-e253. [PMID: 38458321 DOI: 10.1016/j.jhep.2024.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/10/2024]
Affiliation(s)
- Martijn C G J Brouwers
- Division of Endocrinology and Metabolic Diseases, Maastricht University Medical Centre, Maastricht, the Netherlands; CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.
| | - David Cassiman
- Department Chrometa, KU Leuven, Leuven, Belgium; Department of Gastroenterology-Hepatology and Metabolic Centre, University Hospitals Leuven, Belgium
| |
Collapse
|