1
|
Fan W, Yang S, Wei Y, Tian M, Liu Q, Li X, Ding J, Li X, Mao M, Han X, Du Y, Qiu C, Dong Y, Wang Y. Characterization of brain morphology associated with metabolic dysfunction-associated steatotic liver disease in the UK Biobank. Diabetes Obes Metab 2025; 27:3419-3430. [PMID: 40171859 DOI: 10.1111/dom.16362] [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: 10/09/2024] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Emerging evidence has linked metabolic dysfunction-associated steatotic liver disease (MASLD) with accelerated cognitive decline and dementia. We aimed to investigate the associations of MASLD with volumes of total brain tissue and subcortical grey matter, and white matter microstructures in the UK Biobank. METHODS This cross-sectional study included 29,195 individuals (aged 45-82 years) from the UK Biobank who undertook a magnetic resonance imaging (MRI) sub-study between 2014 and 2022. The brain MRI covers three modalities (T1, T2 FLAIR, and diffusion). Volumes of grey matter, subcortical grey matter structures, and regional cortex were derived from T1-weighted images. Fractional anisotropy (FA) and mean diffusivity (MD) were derived from diffusion tensor imaging (DTI) to assess global and tract-specific microstructure. MASLD was defined as the MRI-derived proton density fat fraction (MRI-PDFF) ≥5% and the presence of at least one cardiometabolic criterion. Data were analysed using multiple linear regression models. RESULTS MASLD was significantly associated with smaller volumes of total grey matter and subcortical grey matter (p < 0.05) and reduced Alzheimer's disease (AD)-signature cortical thickness (multivariable-adjusted β = -0.04; 95% confidence interval [CI]: -0.07, -0.01). Having MASLD was associated with higher total white matter hyperintensity (WMH) volume (multivariable-adjusted β = 0.12; 95% CI: 0.10, 0.15). For white matter microstructure, MASLD was associated with increased global FA (multivariable-adjusted β = 0.05; 95% CI: 0.03, 0.08) and reduced global MD (multivariable-adjusted β = -0.04; 95% CI: -0.07, -0.01). CONCLUSIONS Brain morphology associated with MASLD is characterized by smaller subcortical grey matter volume and higher coherence but lower magnitudes of white matter microstructure.
Collapse
Affiliation(s)
- Wenxiao Fan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Shuping Yang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
| | - Yiran Wei
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Minle Tian
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Qianying Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xiaomeng Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Jiahao Ding
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xuewei Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Ming Mao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Xiaolei Han
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Yifeng Du
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Chengxuan Qiu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| | - Yi Dong
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
| | - Yongxiang Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, People's Republic of China
- Shandong Institute of Brain Science and Brain-inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People's Republic of China
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet-Stockholm University, Stockholm, Sweden
| |
Collapse
|
2
|
Triay Bagur A, Arya Z, Waddell T, Pansini M, Fernandes C, Counter D, Jackson E, Thomaides-Brears HB, Robson MD, Bulte DP, Banerjee R, Aljabar P, Brady M. Standardized pancreatic MRI-T1 measurement methods: comparison between manual measurement and a semi-automated pipeline with automatic quality control. Br J Radiol 2025; 98:965-973. [PMID: 40108439 PMCID: PMC12089764 DOI: 10.1093/bjr/tqaf062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/20/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVES Scanner-referenced T1 (srT1) is a method for measuring pancreas T1 relaxation time. The purpose of this multi-centre study is 2-fold: (1) to evaluate the repeatability of manual ROI-based analysis of srT1, (2) to validate a semi-automated measurement method with an automatic quality control (QC) module to identify likely discrepancies between automated and manual measurements. METHODS Pancreatic MRI scans from a scan-rescan cohort (46 subjects) were used to evaluate the repeatability of manual analysis. Seven hundred and eight scans from a longitudinal multi-centre study of 466 subjects were divided into training, internal validation (IV), and external validation (EV) cohorts. A semi-automated method for measuring srT1 using machine learning is proposed and compared against manual analysis on the validation cohorts with and without automated QC. RESULTS Inter-operator agreement between manual ROI-based method and semi-automated method had low bias (3.8 ms or 0.5%) and limits of agreement [-36.6, 44.1] ms. There was good agreement between the 2 methods without automated QC (IV: 3.2 [-47.1, 53.5] ms, EV: -0.5 [-35.2, 34.2] ms). After QC, agreement on the IV set improved, was unchanged in the EV set, and the agreement in both was within inter-operator bounds (IV: -0.04 [-33.4, 33.3] ms, EV: -1.9 [-37.6, 33.7] ms). The semi-automated method improved scan-rescan agreement versus manual analysis (manual: 8.2 [-49.7, 66] ms, automated: 6.7 [-46.7, 60.1] ms). CONCLUSIONS The semi-automated method for characterization of standardized pancreatic T1 using MRI has the potential to decrease analysis time while maintaining accuracy and improving scan-rescan agreement. ADVANCES IN KNOWLEDGE We provide intra-operator, inter-operator, and scan-rescan agreement values for manual measurement of srT1, a standardized biomarker for measuring pancreas fibro-inflammation. Applying a semi-automated measurement method improves scan-rescan agreement and agrees well with manual measurements, while reducing human effort. Adding automated QC can improve agreement between manual and automated measurements. SUMMARY STATEMENT We describe a method for semi-automated, standardized measurement of pancreatic T1 (srT1), which includes automated quality control. Measurements show good agreement with manual ROI-based analysis, with comparable consistency to inter-operator performance.
Collapse
Affiliation(s)
- Alexandre Triay Bagur
- Perspectum Ltd, Oxford OX4 2LL, United Kingdom
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, United Kingdom
| | - Zobair Arya
- Perspectum Ltd, Oxford OX4 2LL, United Kingdom
| | - Tom Waddell
- Perspectum Ltd, Oxford OX4 2LL, United Kingdom
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, United Kingdom
| | - Michele Pansini
- Clinica Di Radiologia EOC, Istituto Di Imaging Della Svizzera Italiana (IIMSI), Ente Ospedaliero Cantonale, Lugano 6900, Switzerland
- Department of Radiology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 0AG, United Kingdom
| | | | | | | | | | | | - Daniel P Bulte
- Department of Engineering Science, University of Oxford, Oxford OX1 3PJ, United Kingdom
| | | | | | | |
Collapse
|
3
|
Jin J, Li B, Wang X, Yang X, Li Y, Wang R, Ye C, Shu J, Fan Z, Xue F, Ge T, Ritchie MD, Pasaniuc B, Wojcik G, Zhao B. PennPRS: a centralized cloud computing platform for efficient polygenic risk score training in precision medicine. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.07.25321875. [PMID: 39990574 PMCID: PMC11844566 DOI: 10.1101/2025.02.07.25321875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Polygenic risk scores (PRS) are becoming increasingly vital for risk prediction and stratification in precision medicine. However, PRS model training presents significant challenges for broader adoption of PRS, including limited access to computational resources, difficulties in implementing advanced PRS methods, and availability and privacy concerns over individual-level genetic data. Cloud computing provides a promising solution with centralized computing and data resources. Here we introduce PennPRS (https://pennprs.org), a scalable cloud computing platform for online PRS model training in precision medicine. We developed novel pseudo-training algorithms for multiple PRS methods and ensemble approaches, enabling model training without requiring individual-level data. These methods were rigorously validated through extensive simulations and large-scale real data analyses involving over 6,000 phenotypes across various data sources. PennPRS supports online single- and multi-ancestry PRS training with seven methods, allowing users to upload their own data or query from more than 27,000 datasets in the GWAS Catalog, submit jobs, and download trained PRS models. Additionally, we applied our pseudo-training pipeline to train PRS models for over 8,000 phenotypes and made their PRS weights publicly accessible. In summary, PennPRS provides a novel cloud computing solution to improve the accessibility of PRS applications and reduce disparities in computational resources for the global PRS research community.
Collapse
Affiliation(s)
- Jin Jin
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Center for Eye-Brain Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bingxuan Li
- UCLA Samueli School of Engineering, Los Angeles, CA 90095, USA
| | - Xiyao Wang
- Department of Computer Science, Columbia University, New York, NY 10027, USA
| | - Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yujue Li
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Ruofan Wang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chenglong Ye
- Department of Statistics, University of Kentucky, Lexington, KY 40536, USA
| | - Juan Shu
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fei Xue
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bogdan Pasaniuc
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Genevieve Wojcik
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Bingxin Zhao
- Penn Center for Eye-Brain Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
4
|
Wang Y, Song SJ, Jiang Y, Lai JCT, Wong GLH, Wong VWS, Yip TCF. Role of noninvasive tests in the prognostication of metabolic dysfunction-associated steatotic liver disease. Clin Mol Hepatol 2025; 31:S51-S75. [PMID: 38934108 PMCID: PMC11925434 DOI: 10.3350/cmh.2024.0246] [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: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024] Open
Abstract
In managing metabolic dysfunction-associated steatotic liver disease, which affects over 30% of the general population, effective noninvasive biomarkers for assessing disease severity, monitoring disease progression, predicting the development of liver-related complications, and assessing treatment response are crucial. The advantage of simple fibrosis scores lies in their widespread accessibility through routinely performed blood tests and extensive validation in different clinical settings. They have shown reasonable accuracy in diagnosing advanced fibrosis and good performance in excluding the majority of patients with a low risk of liver-related complications. Among patients with elevated serum fibrosis scores, a more specific fibrosis and imaging biomarker has proved useful to accurately identify patients at risk of liver-related complications. Among specific fibrosis blood biomarkers, enhanced liver fibrosis is the most widely utilized and has been approved in the United States as a prognostic biomarker. For imaging biomarkers, the availability of vibration-controlled transient elastography has been largely improved over the past years, enabling the use of liver stiffness measurement (LSM) for accurate assessment of significant and advanced fibrosis, and cirrhosis. Combining LSM with other routinely available blood tests enhances the ability to diagnose at-risk metabolic dysfunction-associated steatohepatitis and predict liver-related complications, some reaching an accuracy comparable to that of liver biopsy. Magnetic resonance imaging-based modalities provide the most accurate quantification of liver fibrosis, though the current utilization is limited to research settings. Expanding their future use in clinical practice depends on factors such as cost and facility availability.
Collapse
Affiliation(s)
- Yue Wang
- Medical Data Analytic Center, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Sherlot Juan Song
- Medical Data Analytic Center, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Yichong Jiang
- Medical Data Analytic Center, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Jimmy Che-To Lai
- Medical Data Analytic Center, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Grace Lai-Hung Wong
- Medical Data Analytic Center, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Vincent Wai-Sun Wong
- Medical Data Analytic Center, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | - Terry Cheuk-Fung Yip
- Medical Data Analytic Center, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
5
|
Shah N, Sanyal AJ. A Pragmatic Management Approach for Metabolic Dysfunction-Associated Steatosis and Steatohepatitis. Am J Gastroenterol 2025; 120:75-82. [PMID: 39569874 DOI: 10.14309/ajg.0000000000003215] [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: 07/31/2024] [Accepted: 11/15/2024] [Indexed: 11/22/2024]
Abstract
Obesity and associated insulin resistance induce a chronic metaboinflammatory state that lead to injury and dysfunction of multiple organs resulting in a cluster of noncommunicable diseases such as type 2 diabetes mellitus, hypertension, cardiovascular disease, chronic kidney disease, and metabolic dysfunction-associated steatotic liver disease (MASLD). Metabolic dysfunction-associated steatohepatitis (MASH) is a histologically active form of MASLD and characterized by greater injury and inflammation and progresses to cirrhosis with greater certainty than steatosis alone. The progression to cirrhosis is characterized by increasing fibrosis. The goal of treatment of MASLD/MASH was to improve the metaboinflammatory state i.e., the root cause of the liver disease and to prevent fibrosis progression to cirrhosis whereas in those who already have cirrhosis need additional care to prevent portal hypertension-related outcomes. Fibrosis regression is thus a key objective of treatment. The recent approval of resmetirom for MASH with fibrosis and the use of glucagon-like peptide-1 receptor agonists for obesity and type 2 diabetes has increased awareness of these NCDs and resulted in the growing demand for liver assessment and care in obese individuals. Patients with MASLD also have multiple metabolic comorbidities which represent competing threats to life, and the care of the patient requires both assessment of the totality of the risk and a more holistic approach integrating the care of all of the threats to life. Here, we provide a pragmatic and easily implementable risk-based approach to the evaluation and management of MASLD.
Collapse
Affiliation(s)
- Neha Shah
- Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Arun J Sanyal
- Department of Internal Medicine, Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
6
|
Yang R, Peng H, Pan J, Wan Q, Zou C, Hu F. Native and Gd-EOB-DTPA-Enhanced T1 mapping for Assessment of Liver Fibrosis in NAFLD: Comparative Analysis of Modified Look-Locker Inversion Recovery and Water-specific T1 mapping. Acad Radiol 2025; 32:170-179. [PMID: 39043516 DOI: 10.1016/j.acra.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/25/2024]
Abstract
RATIONALE AND OBJECTIVES To investigate the diagnostic performance of water-specific T1 mapping for staging liver fibrosis in a non-alcoholic fatty liver disease (NAFLD) rabbit model, in comparison to Modified Look-Locker Inversion recovery (MOLLI) T1 mapping. MATERIALS AND METHODS 60 rabbits were randomly divided into the control group (12 rabbits) and NAFLD model groups (eight rabbits per subgroup) corresponding to different durations of high-fat high cholesterol diet feeding. All rabbits underwent MRI examination including MOLLI T1 mapping and 3D multi-echo variable flip angle (VFAME- GRE) sequences were acquired before and 20 min after the administration of Gd- EOB-DTPA. Histological assessments were performed to evaluate steatosis, inflammation, ballooning, and fibrosis. Statistical analysis included the intraclass correlation coefficient, analysis of variance, spearman correlation, multiple linear regression, and receiver operating characteristic curve. RESULTS A moderate correlation was observed between conventional native T1 and MRI-PDFF (r = -0.513, P < 0.001), as well as between conventional native T1 and liver steatosis grades (r = -0.319, P = 0.016). However, no significant correlation was found between the native wT1 and PDFF (r = 0.137, P = 0.314), or between the native wT1 and steatosis grades (r = 0.106, P = 0.435). In the multiple regression analysis, liver fibrosis, and hepatocellular ballooning were identified as independent factors influencing native wT1 in this study (R2 =0.545, P < 0.05), while steatosis was independently associated with conventional native T1 (R2 =0.321, P < 0.05). The AUC values for native T1, native wT1, HBP T1, and HBP wT1 were 0.549(0.410-0.682), 0.811(0.684-0.903), 0.775(0.644-0.876), and 0.752(0.619-0.858) for F1 or higher, 0.581(0.441-0.711), 0.828(0.704-0.916), 0.832(0.708-0.919), and 0.854(0.734-0.934) for F2 or higher, respectively. CONCLUSION The native wT1 may provide a more reliable assessment of early liver fibrosis in the context of NAFLD compared to conventional native T1.
Collapse
Affiliation(s)
- Ru Yang
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, No.278, Baoguang Road, Xindu District, Chengdu, Sichuan, China (R.Y., J.P., F.H.)
| | - Hao Peng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong, China (H.P., Q.W., C.Z.)
| | - Jing Pan
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, No.278, Baoguang Road, Xindu District, Chengdu, Sichuan, China (R.Y., J.P., F.H.)
| | - Qian Wan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong, China (H.P., Q.W., C.Z.)
| | - Chao Zou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, Guangdong, China (H.P., Q.W., C.Z.)
| | - Fubi Hu
- Department of Radiology, The First Affiliated Hospital of Chengdu Medical College, No.278, Baoguang Road, Xindu District, Chengdu, Sichuan, China (R.Y., J.P., F.H.).
| |
Collapse
|
7
|
Aimuzi R, Xie Z, Qu Y, Luo K, Jiang Y. Proteomic signatures of ambient air pollution and risk of non-alcoholic fatty liver disease: A prospective cohort study in the UK Biobank. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 957:177529. [PMID: 39547383 DOI: 10.1016/j.scitotenv.2024.177529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 10/13/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Air pollution has been linked with non-alcoholic fatty liver disease (NAFLD), but the underlying mechanisms characterized by perturbations in the circulating proteome profile are largely unknown. Therefore, we included 51,357 participants from the UK Biobank with 2941 plasma proteins measured in blood samples collected between 2006 and 2010, measurements of annual fine particular matter <2.5 μm in diameter (PM2.5) and nitrogen dioxide (NO2), and follow-up data on NAFLD (743 incident cases occurred over a median follow-up of 13.6 years). Multiple linear regression was used to identify proteins associated with PM2.5 and NO2. Cox proportional hazards models were applied to assess associations of PM2.5 and NO2 and identified proteins with incident NAFLD. Mediation analyses were conducted to explore the mediation role of proteins in the associations between air pollution and incident NAFLD. After adjusting for selected covariates, PM2.5 (hazard ratio [HR] = 2.57, 95%CI:1.27, 5.21, per ln increase) and NO2 (HR = 1.43, 95%CI: 1.10, 1.84, per ln increase) were positively associated with incident NAFLD. We identified 138 proteins associated with PM2.5 (92 positively, 46 inversely, FDR <0.05) and 143 with NO2 (100 positively, 43 inversely). Of the proteins that were significantly associated with both PM2.5 and NO2, 93 (79 positively, 14 inversely) and 79 (69 positively, 10 inversely) were significantly associated with incident NAFLD. Furthermore, 84 PM2.5-associated proteins and 66 NO2-associated proteins significantly mediated the corresponding association between air pollutants and incident NAFLD, with the proportion of mediation effects ranging from 3.2 % to 27.3 % for PM2.5 and 2.6 % to 20.8 % for NO2, respectively. Of note, the majority of significant mediating proteins were enriched in pathways of cytokine-cytokine receptor interaction, viral protein interaction with cytokine and cytokine receptor. Our findings suggested that long-term exposure to PM2.5 and NO2 was associated with an increased risk of NAFLD partially by perturbating circulating proteins involved in pathways of inflammation and immunity responses.
Collapse
Affiliation(s)
- Ruxianguli Aimuzi
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Zhilan Xie
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Yimin Qu
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Kai Luo
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Yu Jiang
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
8
|
Tavaglione F, Marafioti G, Romeo S, Jamialahmadi O. Machine Learning Reveals the Contribution of Lipoproteins to Liver Triglyceride Content and Inflammation. J Clin Endocrinol Metab 2024; 110:218-227. [PMID: 38833012 PMCID: PMC11651681 DOI: 10.1210/clinem/dgae371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
CONTEXT Metabolic dysfunction-associated steatotic liver disease (MASLD) is currently the most common chronic liver disease worldwide and is strongly associated with metabolic comorbidities, including dyslipidemia. OBJECTIVE Herein, we aim to estimate the prevalence of MASLD and metabolic dysfunction-associated steatohepatitis (MASH) in Europeans with isolated hypercholesterolemia and isolated hypertriglyceridemia in the UK Biobank and to estimate the independent contribution of lipoproteins to liver triglyceride content. METHODS We selected 218 732 Europeans from the UK Biobank without chronic viral hepatitis and other causes of liver disease, of whom 14 937 with liver magnetic resonance imaging data available. Next, to examine the relationships between traits in predicting liver triglyceride content, we compared the predictive performance of several machine learning methods and selected the best performing algorithms based on the minimum cross-validated mean squared error (MSE). RESULTS There was an approximately 3-fold and 4-fold enrichment of MASLD and MASH in individuals with isolated hypertriglyceridemia (P = 1.23 × 10-41 and P = 1.29 × 10-10, respectively), whereas individuals with isolated hypercholesterolemia had a marginal higher rate of MASLD and no difference in MASH rate compared with the control group (P = .019 and P = .97, respectively). Among machine learning methods, the feed-forward neural network had the best cross-validation MSE on the validation set. Circulating triglycerides, after body mass index, were the second strongest independent predictor of liver proton density fat fraction with the largest absolute mean Shapley additive explanation value. CONCLUSION Isolated hypertriglyceridemia is the second strongest, after obesity, independent predictor of MASLD/MASH. Individuals with hypertriglyceridemia, but not with hypercholesterolemia, should be screened for liver disease.
Collapse
Affiliation(s)
- Federica Tavaglione
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Giuseppe Marafioti
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Stefano Romeo
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, 41345 Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
- Department of Medicine, Karolinska Institutet, Huddinge, 14152 Stockholm, Sweden
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, 41345 Gothenburg, Sweden
| |
Collapse
|
9
|
Raverdy V, Tavaglione F, Chatelain E, Lassailly G, De Vincentis A, Vespasiani-Gentilucci U, Qadri SF, Caiazzo R, Verkindt H, Saponaro C, Kerr-Conte J, Baud G, Marciniak C, Chetboun M, Oukhouya-Daoud N, Blanck S, Vandel J, Olsson L, Chakaroun R, Gnemmi V, Leteurtre E, Lefebvre P, Haas JT, Yki-Järvinen H, Francque S, Staels B, Le Roux CW, Tremaroli V, Mathurin P, Marot G, Romeo S, Pattou F. Data-driven cluster analysis identifies distinct types of metabolic dysfunction-associated steatotic liver disease. Nat Med 2024; 30:3624-3633. [PMID: 39653777 PMCID: PMC11645276 DOI: 10.1038/s41591-024-03283-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 08/30/2024] [Indexed: 12/15/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) exhibits considerable variability in clinical outcomes. Identifying specific phenotypic profiles within MASLD is essential for developing targeted therapeutic strategies. Here we investigated the heterogeneity of MASLD using partitioning around medoids clustering based on six simple clinical variables in a cohort of 1,389 individuals living with obesity. The identified clusters were applied across three independent MASLD cohorts with liver biopsy (totaling 1,099 participants), and in the UK Biobank to assess the incidence of chronic liver disease, cardiovascular disease and type 2 diabetes. Results unveiled two distinct types of MASLD associated with steatohepatitis on histology and liver imaging. The first cluster, liver-specific, was genetically linked and showed rapid progression of chronic liver disease but limited risk of cardiovascular disease. The second cluster, cardiometabolic, was primarily associated with dysglycemia and high levels of triglycerides, leading to a similar incidence of chronic liver disease but a higher risk of cardiovascular disease and type 2 diabetes. Analyses of samples from 831 individuals with available liver transcriptomics and 1,322 with available plasma metabolomics highlighted that these two types of MASLD exhibited distinct liver transcriptomic profiles and plasma metabolomic signatures, respectively. In conclusion, these data provide preliminary evidence of the existence of two distinct types of clinically relevant MASLD with similar liver phenotypes at baseline, but each with specific underlying biological profiles and different clinical trajectories, suggesting the need for tailored therapeutic strategies.
Collapse
Affiliation(s)
- Violeta Raverdy
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Federica Tavaglione
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Estelle Chatelain
- US 41 - UAR 2014 - PLBS Bilille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | - Guillaume Lassailly
- Department of Hepato-Gastroenterology CHU Lille, University of Lille, Inserm INFINITE-U1286, Lille, France
| | - Antonio De Vincentis
- Operative Unit of Internal Medicine, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Internal Medicine, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Umberto Vespasiani-Gentilucci
- Operative Unit of Clinical Medicine and Hepatology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Clinical Medicine and Hepatology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Sami F Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Robert Caiazzo
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Helene Verkindt
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Chiara Saponaro
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
| | - Julie Kerr-Conte
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
| | - Gregory Baud
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Camille Marciniak
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Mikael Chetboun
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Naima Oukhouya-Daoud
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France
| | - Samuel Blanck
- ULR 2694 METRICS: Évaluation des technologies de santé et des pratiques médicales, University of Lille, CHU Lille, F-59000, Lille, France
| | - Jimmy Vandel
- US 41 - UAR 2014 - PLBS Bilille, University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, F-59000, Lille, France
| | - Lisa Olsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Rima Chakaroun
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Viviane Gnemmi
- Cancer Heterogeneity Plasticity and Resistance to Therapies, CANTHER-UMR9020-U1277 - CNRS, Inserm, CHU Lille, University of Lille, Lille, France
- Department of Pathology, CHU Lille, University of Lille, Lille, France
| | - Emmanuelle Leteurtre
- Cancer Heterogeneity Plasticity and Resistance to Therapies, CANTHER-UMR9020-U1277 - CNRS, Inserm, CHU Lille, University of Lille, Lille, France
- Department of Pathology, CHU Lille, University of Lille, Lille, France
| | - Philippe Lefebvre
- Nuclear Receptors, Metabolic and Cardiovascular Diseases - U1011, University of Lille, Inserm, CHU Lille, Institut Pasteur Lille, Lille, France
| | - Joel T Haas
- Nuclear Receptors, Metabolic and Cardiovascular Diseases - U1011, University of Lille, Inserm, CHU Lille, Institut Pasteur Lille, Lille, France
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Sven Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Edegem, Belgium
- InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Bart Staels
- Nuclear Receptors, Metabolic and Cardiovascular Diseases - U1011, University of Lille, Inserm, CHU Lille, Institut Pasteur Lille, Lille, France
| | - Carel W Le Roux
- Diabetes Complications Research Centre, University College Dublin, Dublin, Ireland
| | - Valentina Tremaroli
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Philippe Mathurin
- Department of Hepato-Gastroenterology CHU Lille, University of Lille, Inserm INFINITE-U1286, Lille, France
| | - Guillemette Marot
- ULR 2694 METRICS: Évaluation des technologies de santé et des pratiques médicales, University of Lille, CHU Lille, F-59000, Lille, France
- MODAL: Models for Data Analysis and Learning, Inria, F-59000, Lille, France
| | - Stefano Romeo
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden.
- Department of Medicine Huddinge (H7), Karolinska Institutet and University Hospital, Stockholm, Sweden.
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, Gothenburg, Sweden.
| | - François Pattou
- Translational Research for Diabetes UMR 1190, University of Lille, Inserm, Institut Pasteur Lille, CHU Lille, Lille, France.
- Department of General and Endocrine Surgery, Centre Hospitalier et Universitaire de Lille, Lille, France.
| |
Collapse
|
10
|
Bell W, Jennings A, Thompson AS, Bondonno NP, Tresserra-Rimbau A, Kühn T, Cassidy A. A flavonoid-rich diet is associated with lower risk and improved imaging biomarkers of nonalcoholic fatty liver disease: a prospective cohort study. Am J Clin Nutr 2024; 120:1325-1334. [PMID: 39341459 PMCID: PMC11619783 DOI: 10.1016/j.ajcnut.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/14/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Mechanistic studies and short-term randomized trials suggest higher intakes of dietary flavonoids may protect against nonalcoholic fatty liver disease (NAFLD). OBJECTIVES We aimed to perform the first population-based study with long-term follow-up on flavonoid consumption, incident NAFLD, and validated NAFLD biomarkers. METHODS In a prospective study, we assessed the associations between flavonoid intake based on ≥2 24-h dietary assessments and NAFLD risk among 121,064 adults aged 40-69 y by multivariable Cox regression analyses. We further assessed the associations between flavonoid intake and magnetic resonance imaging-derived liver fat (a subset of n = 11,435) and liver-corrected T1 values (cT1; a subset of n = 9570), a marker of steatosis, more sensitive to inflammatory pathology. RESULTS Over 10 y of follow-up, 1081 cases of NAFLD were identified. Participants in the highest quartile (Q4) of the flavodiet score reflecting the consumption of foods high in flavonoids, had a 19% lower risk of NAFLD compared to the lowest quartile (Q1) [hazard ratio (HR) (95% confidence interval (CI): 0.81 (0.67, 0.97), P-trend = 0.02)]. Moreover, participants in the Q4 of the flavodiet score had lower liver fat and cT1 values than those in Q1 (liver fat: relative difference Q1 compared with Q4: -5.28%, P-trend = <0.001; cT1: relative difference Q1 compared with Q4: -1.73%, P-trend = <0.001). When compared to low intakes, high intakes of apples and tea were associated with lower NAFLD risk [apples: HR (95% CI): 0.78 (0.67, 0.92), P-trend = <0.01; tea: HR (95% CI): 0.86 (0.72, 1.02), P-trend = 0.03)]. Additionally, when compared to low intakes, high apple, tea, and dark chocolate intakes were significantly associated with lower liver fat values, whereas high tea and red pepper intakes were significantly associated with lower cT1 values. CONCLUSIONS The consumption of flavonoid-rich foods was associated with a reduced risk of NAFLD among middle-aged adults.
Collapse
Affiliation(s)
- William Bell
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Amy Jennings
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Alysha S Thompson
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Nicola P Bondonno
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Northern Ireland, United Kingdom; Danish Cancer Institute, Copenhagen, Denmark; Nutrition and Health Innovation Research Institute, School of Medical and Health Sciences, Edith Cowan University, Western Australia, Australia
| | - Anna Tresserra-Rimbau
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Northern Ireland, United Kingdom; Department of Nutrition, Food Science, and Gastronomy, XIA, School of Pharmacy and Food Sciences, Institute of Nutrition and Food Safety, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Institute of Health Carlos III, Madrid, Spain
| | - Tilman Kühn
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Northern Ireland, United Kingdom; Department of Nutritional Sciences, University of Vienna, Vienna, Austria; Center for Public Health, Medical University of Vienna, Vienna, Austria.
| | - Aedín Cassidy
- The Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Northern Ireland, United Kingdom.
| |
Collapse
|
11
|
Fan Y, Chen J, Fan Z, Chirinos J, Stein JL, Sullivan PF, Wang R, Nadig A, Zhang DY, Huang S, Jiang Z, Guan PY, Qian X, Li T, Li H, Sun Z, Ritchie MD, O’Brien J, Witschey W, Rader DJ, Li T, Zhu H, Zhao B. Mapping rare protein-coding variants on multi-organ imaging traits. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.16.24317443. [PMID: 39606337 PMCID: PMC11601754 DOI: 10.1101/2024.11.16.24317443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Human organ structure and function are important endophenotypes for clinical outcomes. Genome-wide association studies (GWAS) have identified numerous common variants associated with phenotypes derived from magnetic resonance imaging (MRI) of the brain and body. However, the role of rare protein-coding variations affecting organ size and function is largely unknown. Here we present an exome-wide association study that evaluates 596 multi-organ MRI traits across over 50,000 individuals from the UK Biobank. We identified 107 variant-level associations and 224 gene-based burden associations (67 unique gene-trait pairs) across all MRI modalities, including PTEN with total brain volume, TTN with regional peak circumferential strain in the heart left ventricle, and TNFRSF13B with spleen volume. The singleton burden model and AlphaMissense annotations contributed 8 unique gene-trait pairs including the association between an approved drug target gene of KCNA5 and brain functional activity. The identified rare coding signals elucidate some shared genetic regulation across organs, prioritize previously identified GWAS loci, and are enriched for drug targets. Overall, we demonstrate how rare variants enhance our understanding of genetic effects on human organ morphology and function and their connections to complex diseases.
Collapse
Affiliation(s)
- Yijun Fan
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julio Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jason L. Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rujin Wang
- Regeneron Genetics Center, 777 Old Saw Mill River Rd., Tarrytown, NY, 10591, USA
| | - Ajay Nadig
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David Y. Zhang
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuai Huang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zhiwen Jiang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter Yi Guan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xinjie Qian
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ting Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Haoyue Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Zehui Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marylyn D. Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Joan O’Brien
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Diseases, Philadelphia, PA 19104, USA
| | - Walter Witschey
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J. Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxin Zhao
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Population Aging Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Center for Eye-Brain Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Hudson D, Afzaal T, Bualbanat H, AlRamdan R, Howarth N, Parthasarathy P, AlDarwish A, Stephenson E, Almahanna Y, Hussain M, Diaz LA, Arab JP. Modernizing metabolic dysfunction-associated steatotic liver disease diagnostics: the progressive shift from liver biopsy to noninvasive techniques. Therap Adv Gastroenterol 2024; 17:17562848241276334. [PMID: 39553445 PMCID: PMC11565685 DOI: 10.1177/17562848241276334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 07/27/2024] [Indexed: 11/19/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a growing public health concern worldwide. Liver biopsy is the gold standard for diagnosing and staging MASLD, but it is invasive and carries associated risks. In recent years, there has been significant progress in developing noninvasive techniques for evaluation. This review article discusses briefly current available noninvasive assessments and the various liver biopsy techniques available for MASLD, including invasive techniques such as transjugular and transcutaneous needle biopsy, intraoperative/laparoscopic biopsy, and the evolving role of endoscopic ultrasound-guided biopsy. In addition to discussing the various biopsy techniques, we review the current state of knowledge on the histopathologic evaluation of MASLD, including the various scoring systems used to grade and stage the disease. We also explore current and alternative modalities for histopathologic evaluation, such as whole slide imaging and the utility of immunohistochemistry. Overall, this review article provides a comprehensive overview of the progress in liver biopsy techniques for MASLD and compares invasive and noninvasive modalities. However, beyond clinical trials, the practical application of liver biopsy may be limited, as ongoing advancements in noninvasive fibrosis assessments are expected to more effectively identify candidates for MASLD treatment in real-world settings.
Collapse
Affiliation(s)
- David Hudson
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Tamoor Afzaal
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Hasan Bualbanat
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Raaed AlRamdan
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Nisha Howarth
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Pavithra Parthasarathy
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Alia AlDarwish
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Emily Stephenson
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Yousef Almahanna
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Maytham Hussain
- Division of Gastroenterology, Department of Medicine, Schulich School of Medicine, Western University and London Health Sciences Centre, London, ON, Canada
| | - Luis Antonio Diaz
- Departamento de Gastroenterologia, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
- MASLD Research Center, Division of MASLD Research Center, Division of Gastroenterology and Hepatology, University of California San Diego, San Diego, CA, USA
| | - Juan Pablo Arab
- Stravitz-Sanyal Institute of Liver Disease and Metabolic Health, Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, 1201 E. Broad St. P.O. Box 980341, Richmond, VA 23284, USA
| |
Collapse
|
13
|
Thompson RB, Sherrington R, Beaulieu C, Kirkham A, Paterson DI, Seres P, Grenier J. Reference Values for Water-Specific T1 of the Liver at 3 T: T2*-Compensation and the Confounding Effects of Fat. J Magn Reson Imaging 2024; 60:2063-2075. [PMID: 38305588 DOI: 10.1002/jmri.29262] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND T1 mapping of the liver is confounded by the presence of fat. Multiparametric T1 mapping combines fat-water separation with T1-weighting to enable imaging of water-specific T1 (T1Water), proton density fat fraction (PDFF), and T2* values. However, normative T1Water values in the liver and its dependence on age/sex is unknown. PURPOSE Determine normative values for T1Water in the liver with comparison to MOLLI and evaluate a T2*-compensation approach to reduce T1 variability. STUDY TYPE Prospective observational; phantoms. POPULATIONS One hundred twenty-four controls (56 male, 18-75 years), 50 patients at-risk for liver disease (18 male, 30-76 years). FIELD STRENGTH/SEQUENCE 2.89 T; Saturation-recovery chemical-shift encoded T1 Mapping (SR-CSE); MOLLI. ASSESSMENT SR-CSE provided T1Water measurements, PDFF and T2* values in the liver across three slices in 6 seconds. These were compared with MOLLI T1 values. A new T2*-compensation approach to reduce T1 variability was evaluated test/re-test reproducibility. STATISTICAL TESTS Linear regression, ANCOVA, t-test, Bland and Altman, intraclass correlation coefficient (ICC). P < 0.05 was considered statistically significant. RESULTS Liver T1 values were significantly higher in healthy females (F) than males (M) for both SR-CSE (F-973 ± 78 msec, M-930 ± 72 msec) and MOLLI (F-802 ± 55 msec, M-759 ± 69 msec). T1 values were negatively correlated with age, with similar sex- and age-dependencies observed in T2*. The T2*-compensation model reduced the variability of T1 values by half and removed sex- and age-differences (SR-CSE: F-946 ± 36 msec, M-941 ± 43 msec; MOLLI: F-775 ± 35 msec, M-770 ± 35 msec). At-risk participants had elevated PDFF and T1 values, which became more distinct from the healthy cohort after T2*-compensation. MOLLI systematically underestimated liver T1 values by ~170 msec with an additional positive T1-bias from fat content (~11 msec/1% in PDFF). Reproducibility ICC values were ≥0.96 for all parameters. DATA CONCLUSION Liver T1Water values were lower in males and decreased with age, as observed for SR-CSE and MOLLI acquisitions. MOLLI underestimated liver T1 with an additional large positive fat-modulated T1 bias. T2*-compensation removed sex- and age-dependence in liver T1, reduced the range of healthy values and increased T1 group differences between healthy and at-risk groups. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 1.
Collapse
Affiliation(s)
- Richard B Thompson
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Rachel Sherrington
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - Christian Beaulieu
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Amy Kirkham
- Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - David I Paterson
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Peter Seres
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - Justin Grenier
- Department of Radiology and Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
14
|
Brewster F, Middleton Z, McWilliam A, Brocklehurst A, Radhakrishna G, Chuter R. Feasibility of using contrast-free quantitative magnetic resonance imaging for liver sparing stereotactic ablative body radiotherapy. Clin Transl Radiat Oncol 2024; 49:100859. [PMID: 39376618 PMCID: PMC11456905 DOI: 10.1016/j.ctro.2024.100859] [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: 07/15/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 10/09/2024] Open
Abstract
Background and purpose Tumours in the liver often develop on a background of liver cirrhosis and impaired liver function. As a result, radiotherapy treatments are limited by radiation-induced liver disease, parameterised by the liver mean dose (LMD). Liver function is highly heterogeneous, especially in liver cancer, but the use of LMD does not take this into account. One possible way to improve liver treatments is to use quantitative imaging techniques to assess liver health and prioritise the sparing of healthy liver tissue. Materials and methods Anatomical T2 and quantitative iron-corrected T1 (cT1) images were made available for 10 patients with liver metastases. Functional liver volumes were automatically segmented on the quantitative images using a threshold. Liver stereotactic ablative body radiotherapy (SABR) plans were made using a departmental protocol. Liver-sparing plans were then made by reducing the dose to the functional sub-volume. Results The sparing plans achieved a statistically significant ( p = 0.002 ) reduction in the functional liver mean dose, with a mean reduction of 1.4 Gy. The LMD was also significantly different ( p = 0.002 ) but had a smaller magnitude with a mean reduction of 0.7 Gy. There were some differences in the planning target volume D99% ( p = 0.04 ) but the sparing plans remained within the optimal tolerance and the D95% was not significantly different ( p = 0.2 ). Conclusions This study has, for the first time, demonstrated the use of cT1 maps in radiotherapy showing significant reductions in dose to the healthy liver. Further work is needed to validate this in liver cancer patients, who would likely benefit most.
Collapse
Affiliation(s)
- Frank Brewster
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | | | - Alan McWilliam
- Department of Radiotherapy Related Research, Division of Clinical Cancer Science, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| | - Andrew Brocklehurst
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Ganesh Radhakrishna
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Robert Chuter
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
- Department of Radiotherapy Related Research, Division of Clinical Cancer Science, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, UK
| |
Collapse
|
15
|
Shumbayawonda E, Beyer C, de Celis Alonso B, Hidalgo-Tobon S, López-Martínez B, Klunder-Klunder M, Miranda-Lora AL, Thomas EL, Bell JD, Breen DJ, Janowski K, Pronicki M, Grajkowska W, Wozniak M, Jurkiewicz E, Banerjee R, Socha P, So PW. Reference Range of Quantitative MRI Metrics Corrected T1 and Liver Fat Content in Children and Young Adults: Pooled Participant Analysis. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1230. [PMID: 39457195 PMCID: PMC11506660 DOI: 10.3390/children11101230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Multiparametric MRI markers of liver health corrected T1 (cT1) and proton density fat fraction (PDFF) have shown utility in the management of various chronic liver diseases. We assessed the normal population reference range of both cT1 and PDFF in healthy child and adult volunteers without any known liver disease. METHODS A retrospective multi-centre pooled analysis of 102 child and young adult (9.1 years (6-18)) volunteers from three centres: Children's Memorial Health Institute (N = 21), University Hospital Southampton (N = 28) and Hospital Infantil de Mexico (N = 53). Sex and ethnic differences were investigated for both cT1 and PDFF. Age effects were investigated with comparison to a pooled adult cohort from the UK Biobank (N = 500) and CoverScan (N = 71), covering an age range of 21 to 81 years. RESULTS cT1 values were normally distributed with a median of 748 ms (IQR: 725-768 ms; 2.5-97.5 percentiles: 683-820 ms). PDFF values followed a normal distribution with a median of 1.7% (IQR: 1.3-1.9%; 2.5-97.5 percentiles: 1-4.4%). There were no significant age and sex differences in cT1 and PDFF between children and young adults. No differences in cT1 and PDFF were found between ethnicities. Age comparisons showed statistically significant, but clinically negligible, cT1 (748 ms vs. 732 ms) and PDFF (2.4% vs. 1.9%) differences between paediatric and adult groups, respectively. CONCLUSIONS Median healthy cT1 and PDFF reference ranges in children and young adults fall within the reported limits for normal of 800 ms and 5%, respectively.
Collapse
Affiliation(s)
| | | | - Benito de Celis Alonso
- Faculty of Physical and Mathematical Sciences, Benemérita Universidad Autónoma de Puebla, Puebla 72000, Mexico
| | - Silvia Hidalgo-Tobon
- Imaging Department, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
- Physics Department, Universidad Autónoma Metropolitana, Campus Iztapalapa, Mexico City 09340, Mexico
| | - Briceida López-Martínez
- Sub Direction of Research, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
| | - Miguel Klunder-Klunder
- Research Committee, Latin American Society for Pediatric Gastroenterology, Hepatology and Nutrition (SLAGHNP/LASPGHAN), Mexico City 06720, Mexico
- Epidemiological Research Unit in Endocrinology and Nutrition, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
| | - América Liliana Miranda-Lora
- Epidemiological Research Unit in Endocrinology and Nutrition, Children’s Hospital of Mexico Federico Gómez, Mexico City 06720, Mexico
| | - E. Louise Thomas
- Research Centre for Optimal Health, University of Westminster, London W1B 2HW, UK
| | - Jimmy D. Bell
- Research Centre for Optimal Health, University of Westminster, London W1B 2HW, UK
| | - David J. Breen
- Department of Radiology, University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton SO16 6YD, UK
| | - Kamil Janowski
- Department of Gastroenterology, Hepatology, Nutritional Disorders and Pediatrics, The Children’s Memorial Health Institute, 20 04-736 Warsaw, Poland
| | - Maciej Pronicki
- Department of Pathology, The Children’s Memorial Health Institute, 20 04-736 Warsaw, Poland
| | - Wieslawa Grajkowska
- Department of Pathology, The Children’s Memorial Health Institute, 20 04-736 Warsaw, Poland
| | - Malgorzata Wozniak
- Department of Gastroenterology, Hepatology, Nutritional Disorders and Pediatrics, The Children’s Memorial Health Institute, 20 04-736 Warsaw, Poland
| | - Elzbieta Jurkiewicz
- Department of Diagnostic Imaging, The Children’s Memorial Health Institute, 20 04-736 Warsaw, Poland
| | | | - Piotr Socha
- Department of Gastroenterology, Hepatology, Nutritional Disorders and Pediatrics, The Children’s Memorial Health Institute, 20 04-736 Warsaw, Poland
| | - Po-Wah So
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| |
Collapse
|
16
|
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
|
17
|
Liu M, Li Y, Sun L, Sun M, Hu X, Li Q, Yu M, Wang C, Ren X, Ma J. Integrating Multi-Organ Imaging-Derived Phenotypes and Genomic Information for Predicting the Occurrence of Common Diseases. Bioengineering (Basel) 2024; 11:872. [PMID: 39329614 PMCID: PMC11428582 DOI: 10.3390/bioengineering11090872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 09/28/2024] Open
Abstract
As medical imaging technologies advance, these tools are playing a more and more important role in assisting clinical disease diagnosis. The fusion of biomedical imaging and multi-modal information is profound, as it significantly enhances diagnostic precision and comprehensiveness. Integrating multi-organ imaging with genomic information can significantly enhance the accuracy of disease prediction because many diseases involve both environmental and genetic determinants. In the present study, we focused on the fusion of imaging-derived phenotypes (IDPs) and polygenic risk score (PRS) of diseases from different organs including the brain, heart, lung, liver, spleen, pancreas, and kidney for the prediction of the occurrence of nine common diseases, namely atrial fibrillation, heart failure (HF), hypertension, myocardial infarction, asthma, type 2 diabetes, chronic kidney disease, coronary artery disease (CAD), and chronic obstructive pulmonary disease, in the UK Biobank (UKBB) dataset. For each disease, three prediction models were developed utilizing imaging features, genomic data, and a fusion of both, respectively, and their performances were compared. The results indicated that for seven diseases, the model integrating both imaging and genomic data achieved superior predictive performance compared to models that used only imaging features or only genomic data. For instance, the Area Under Curve (AUC) of HF risk prediction was increased from 0.68 ± 0.15 to 0.79 ± 0.12, and the AUC of CAD diagnosis was increased from 0.76 ± 0.05 to 0.81 ± 0.06.
Collapse
Affiliation(s)
- Meng Liu
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Yan Li
- Department of Radiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China;
| | - Longyu Sun
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Mengting Sun
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Xumei Hu
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Qing Li
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Mengyao Yu
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Chengyan Wang
- Human Phenome Institute, Fudan University, Shanghai 201203, China; (M.L.); (L.S.); (M.S.); (Q.L.); (M.Y.); (C.W.)
| | - Xinping Ren
- Ultrasound Department, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Jinlian Ma
- Radiology Department, Jiangyin Affiliated Hospital of Nanjing University of Chinese Medicine, 130 Renmin Middle Road, Jiangyin 214400, China
| |
Collapse
|
18
|
Serai SD, Robson MD, Tirkes T, Trout AT. T 1 Mapping of the Abdomen, From the AJR "How We Do It" Special Series. AJR Am J Roentgenol 2024. [PMID: 39194308 DOI: 10.2214/ajr.24.31643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
By exploiting different tissues' characteristic T1 relaxation times, T1-weighted images help distinguish normal and abnormal tissues, aiding assessment of diffuse and local pathologies. However, such images do not provide quantitative T1 values. Advances in abdominal MRI techniques have enabled measurement of abdominal organs' T1 relaxation times, which can be used to create color-coded quantitative maps. T1 mapping is sensitive to tissue microenvironments including inflammation and fibrosis and has received substantial interest for noninvasive imaging of abdominal organ pathology. In particular, quantitative mapping provides a powerful tool for evaluation of diffuse disease by making apparent changes in T1 occurring across organs that may otherwise be difficult to identify. Quantitative measurement also facilitates sensitive monitoring of longitudinal T1 changes. Increased T1 in liver helps to predict parenchymal fibro-inflammation, in pancreas is associated with reduced exocrine function from chronic or autoimmune pancreatitis, and in kidney is associated with impaired renal function and aids diagnosis of chronic kidney disease. In this review, we describe the acquisition, postprocessing, and analysis of T1 maps in the abdomen and explore applications in liver, spleen, pancreas, and kidney. We highlight practical aspects of implementation and standardization, technical pitfalls and confounding factors, and areas of likely greatest clinical impact.
Collapse
Affiliation(s)
- Suraj D Serai
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Temel Tirkes
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrew T Trout
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
19
|
Yang X, Sullivan PF, Li B, Fan Z, Ding D, Shu J, Guo Y, Paschou P, Bao J, Shen L, Ritchie MD, Nave G, Platt ML, Li T, Zhu H, Zhao B. Multi-organ imaging-derived polygenic indexes for brain and body health. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.04.18.23288769. [PMID: 38883759 PMCID: PMC11177904 DOI: 10.1101/2023.04.18.23288769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The UK Biobank (UKB) imaging project is a crucial resource for biomedical research, but is limited to 100,000 participants due to cost and accessibility barriers. Here we used genetic data to predict heritable imaging-derived phenotypes (IDPs) for a larger cohort. We developed and evaluated 4,375 IDP genetic scores (IGS) derived from UKB brain and body images. When applied to UKB participants who were not imaged, IGS revealed links to numerous phenotypes and stratified participants at increased risk for both brain and somatic diseases. For example, IGS identified individuals at higher risk for Alzheimer's disease and multiple sclerosis, offering additional insights beyond traditional polygenic risk scores of these diseases. When applied to independent external cohorts, IGS also stratified those at high disease risk in the All of Us Research Program and the Alzheimer's Disease Neuroimaging Initiative study. Our results demonstrate that, while the UKB imaging cohort is largely healthy and may not be the most enriched for disease risk management, it holds immense potential for stratifying the risk of various brain and body diseases in broader external genetic cohorts.
Collapse
Affiliation(s)
- Xiaochen Yang
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxuan Li
- UCLA Samueli School of Engineering, Los Angeles, CA 90095, USA
| | - Zirui Fan
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dezheng Ding
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Juan Shu
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Yuxin Guo
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Gideon Nave
- Marketing Department, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael L. Platt
- Marketing Department, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hongtu Zhu
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Applied Mathematics and Computational Science Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Population Aging Research Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Nauffal V, Klarqvist MDR, Hill MC, Pace DF, Di Achille P, Choi SH, Rämö JT, Pirruccello JP, Singh P, Kany S, Hou C, Ng K, Philippakis AA, Batra P, Lubitz SA, Ellinor PT. Noninvasive assessment of organ-specific and shared pathways in multi-organ fibrosis using T1 mapping. Nat Med 2024; 30:1749-1760. [PMID: 38806679 DOI: 10.1038/s41591-024-03010-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/22/2024] [Indexed: 05/30/2024]
Abstract
Fibrotic diseases affect multiple organs and are associated with morbidity and mortality. To examine organ-specific and shared biologic mechanisms that underlie fibrosis in different organs, we developed machine learning models to quantify T1 time, a marker of interstitial fibrosis, in the liver, pancreas, heart and kidney among 43,881 UK Biobank participants who underwent magnetic resonance imaging. In phenome-wide association analyses, we demonstrate the association of increased organ-specific T1 time, reflecting increased interstitial fibrosis, with prevalent diseases across multiple organ systems. In genome-wide association analyses, we identified 27, 18, 11 and 10 independent genetic loci associated with liver, pancreas, myocardial and renal cortex T1 time, respectively. There was a modest genetic correlation between the examined organs. Several loci overlapped across the examined organs implicating genes involved in a myriad of biologic pathways including metal ion transport (SLC39A8, HFE and TMPRSS6), glucose metabolism (PCK2), blood group antigens (ABO and FUT2), immune function (BANK1 and PPP3CA), inflammation (NFKB1) and mitosis (CENPE). Finally, we found that an increasing number of organs with T1 time falling in the top quintile was associated with increased mortality in the population. Individuals with a high burden of fibrosis in ≥3 organs had a 3-fold increase in mortality compared to those with a low burden of fibrosis across all examined organs in multivariable-adjusted analysis (hazard ratio = 3.31, 95% confidence interval 1.77-6.19; P = 1.78 × 10-4). By leveraging machine learning to quantify T1 time across multiple organs at scale, we uncovered new organ-specific and shared biologic pathways underlying fibrosis that may provide therapeutic targets.
Collapse
Affiliation(s)
- Victor Nauffal
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Matthew C Hill
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Danielle F Pace
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paolo Di Achille
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joel T Rämö
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Pulkit Singh
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cody Hou
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenney Ng
- Center for Computational Health, IBM Research, Cambridge, MA, USA
| | - Anthony A Philippakis
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Eric and Wendy Schmidt Center, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Puneet Batra
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven A Lubitz
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
21
|
Mak AL, Wassenaar N, van Dijk AM, Troelstra M, Houttu V, van Son K, Driessen S, Zwirs D, van den Berg-Faay S, Shumbayawonda E, Runge J, Doukas M, Verheij J, Beuers U, Nieuwdorp M, Cahen DL, Nederveen A, Gurney-Champion O, Holleboom A. Intrapancreatic fat deposition is unrelated to liver steatosis in metabolic dysfunction-associated steatotic liver disease. JHEP Rep 2024; 6:100998. [PMID: 38379586 PMCID: PMC10877191 DOI: 10.1016/j.jhepr.2023.100998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 11/21/2023] [Accepted: 12/21/2023] [Indexed: 02/22/2024] Open
Abstract
Background & Aims Individuals with obesity may develop intrapancreatic fat deposition (IPFD) and fatty pancreas disease (FPD). Whether this causes inflammation and fibrosis and leads to pancreatic dysfunction is less established than for liver damage in metabolic dysfunction-associated steatotic liver disease (MASLD). Moreover, the interrelations of FPD and MASLD are poorly understood. Therefore, we aimed to assess IPFD and fibro-inflammation in relation to pancreatic function and liver disease severity in individuals with MASLD. Methods Seventy-six participants from the Amsterdam MASLD-MASH cohort (ANCHOR) study underwent liver biopsy and multiparametric MRI of the liver and pancreas, consisting of proton-density fat fraction sequences, T1 mapping and intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI). Results The prevalence of FPD was 37.3%. There was a clear correlation between pancreatic T1 relaxation time, which indicates fibro-inflammation, and parameters of glycemic dysregulation, namely HbA1c (R = 0.59; p <0.001), fasting glucose (R = 0.51; p <0.001) and the presence of type 2 diabetes (mean 802.0 ms vs. 733.6 ms; p <0.05). In contrast, there was no relation between IPFD and hepatic fat content (R = 0.03; p = 0.80). Pancreatic IVIM diffusion (IVIM-D) was lower in advanced liver fibrosis (p <0.05) and pancreatic perfusion (IVIM-f), reflecting vessel density, inversely correlated to histological MASLD activity (p <0.05). Conclusions Consistent relations exist between pancreatic fibro-inflammation on MRI and endocrine function in individuals with MASLD. However, despite shared dysmetabolic drivers, our study suggests IPFD is a separate pathophysiological process from MASLD. Impact and implications Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide and 68% of people with type 2 diabetes have MASLD. However, fat infiltration and inflammation in the pancreas are understudied in individuals with MASLD. In this cross-sectional MRI study, we found no relationship between fat accumulation in the pancreas and liver in a cohort of patients with MASLD. However, our results show that inflammatory and fibrotic processes in the pancreas may be interrelated to features of type 2 diabetes and to the severity of liver disease in patients with MASLD. Overall, the results suggest that pancreatic endocrine dysfunction in individuals with MASLD may be more related to glucotoxicity than to lipotoxicity. Clinical trial number NTR7191 (Dutch Trial Register).
Collapse
Affiliation(s)
- Anne Linde Mak
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nienke Wassenaar
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Anne-Marieke van Dijk
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marian Troelstra
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Veera Houttu
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Koen van Son
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Radboudumc, Nijmegen, The Netherlands
| | - Stan Driessen
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Diona Zwirs
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Sandra van den Berg-Faay
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | - Jurgen Runge
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joanne Verheij
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ulrich Beuers
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Djuna L. Cahen
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Aart Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Oliver Gurney-Champion
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Adriaan Holleboom
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Meloni A, Carnevale A, Gaio P, Positano V, Passantino C, Pepe A, Barison A, Todiere G, Grigoratos C, Novani G, Pistoia L, Giganti M, Cademartiri F, Cossu A. Liver T1 and T2 mapping in a large cohort of healthy subjects: normal ranges and correlation with age and sex. MAGMA (NEW YORK, N.Y.) 2024; 37:93-100. [PMID: 38019376 DOI: 10.1007/s10334-023-01135-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/05/2023] [Accepted: 10/20/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVE We established normal ranges for native T1 and T2 values in the human liver using a 1.5 T whole-body imager (General Electric) and we evaluated their variation across hepatic segments and their association with age and sex. MATERIALS AND METHODS One-hundred healthy volunteers aged 20-70 years (50% females) underwent MRI. Modified Look-Locker inversion recovery and multi-echo fast-spin-echo sequences were used to measure hepatic native global and segmental T1 and T2 values, respectively. RESULTS T1 and T2 values exhibited good intra- and inter-observer reproducibility (coefficient of variation < 5%). T1 value over segment 4 was significantly lower than the T1 values over segments 2 and 3 (p < 0.0001). No significant regional T2 variability was detected. Segmental and global T1 values were not associated with age or sex. Global T2 values were independent from age but were significantly lower in males than in females. The lower and upper limits of normal for global T1 values were, respectively, 442 ms and 705 ms. The normal range for global T2 values was 35 ms-54 ms in males and 39 ms-54 ms in females. DISCUSSION Liver T1 and T2 mapping is feasible and reproducible and the provided normal ranges may help to establish diagnosis and progression of various liver diseases.
Collapse
Affiliation(s)
- Antonella Meloni
- Radiology Department, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1-56124, Pisa, Italy
- Bioengineering Unit, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | - Aldo Carnevale
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Paolo Gaio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Vincenzo Positano
- Radiology Department, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1-56124, Pisa, Italy
- Bioengineering Unit, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | | | - Alessia Pepe
- Institute of Radiology, University of Padua, Padua, Italy
| | - Andrea Barison
- Division of Cardiology and Cardiovascular Medicine, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | - Giancarlo Todiere
- Division of Cardiology and Cardiovascular Medicine, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | - Chrysanthos Grigoratos
- Division of Cardiology and Cardiovascular Medicine, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | - Giovanni Novani
- Radiology Department, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1-56124, Pisa, Italy
| | - Laura Pistoia
- Radiology Department, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1-56124, Pisa, Italy
- U.O.S.V.D. Ricerca Clinica, Fondazione G. Monasterio CNR-Regione Toscana, Pisa, Italy
| | | | - Filippo Cademartiri
- Radiology Department, Fondazione G. Monasterio CNR-Regione Toscana, Via Moruzzi, 1-56124, Pisa, Italy.
| | - Alberto Cossu
- University Radiology Unit, University of Ferrara, Ferrara, Italy
| |
Collapse
|
23
|
Wan Q, Peng H, Liu F, Liu X, Cheng C, Tie C, Deng J, Lyu J, Jia Y, Wang Y, Zheng H, Liang D, Liu X, Zou C. Ability of dynamic gadoxetic acid-enhanced magnetic resonance imaging combined with water-specific T1 mapping to reflect inflammation in a rat model of early-stage nonalcoholic steatohepatitis. Quant Imaging Med Surg 2024; 14:1591-1601. [PMID: 38415124 PMCID: PMC10895110 DOI: 10.21037/qims-23-482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/23/2023] [Indexed: 02/29/2024]
Abstract
Background Gadolinium ethoxybenzyl-diethylenetriaminepentaacetic acid (Gd-EOB-DTPA) has shown potential in reflecting the hepatic function alterations in nonalcoholic steatohepatitis (NASH). The purpose of this study was to evaluate whether Gd-EOB-DTPA combined with water-specific T1 (wT1) mapping can be used to detect liver inflammation in the early-stage of NASH in rats. Methods In this study, 54 rats with methionine- and choline-deficient (MCD) diet-induced NASH and 10 normal control rats were examined. A multiecho variable flip angle gradient echo (VFA-GRE) sequence was performed and repeated 40 times after the injection of Gd-EOB-DTPA. The wT1 of the liver and the reduction rate of wT1 (rrT1) were calculated. All rats were histologically evaluated and grouped according to the NASH Clinical Research Network scoring system. Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to detect the expression of Gd-EOB-DTPA transport genes. Analysis of variance and least significant difference tests were used for multiple comparisons of quantitative results between all groups. Multiple regression analysis was applied to identify variables associated with precontrast wT1 (wT1pre), and receiver operating characteristic (ROC) analysis was performed to assess the diagnostic performance. Results The rats were grouped according to inflammatory stage (G0 =4, G1 =15, G2 =12, G3 =23) and fibrosis stage (F0 =26, F1 =19, F2 =9). After the infusion of Gd-EOB-DTPA, the rrT1 showed significant differences between the control and NASH groups (P<0.05) but no difference between the different inflammation and fibrosis groups at any time points. The areas under curve (AUCs) of rrT1 at 10, 20, and 30 minutes were only 0.53, 0.58, and 0.61, respectively, for differentiating between low inflammation grade (G0 + G1) and high inflammation grade (G2 + G3). The MRI findings were verified by qRT-PCR examination, in which the Gd-EOB-DTPA transporter expressions showed no significant differences between any inflammation groups. Conclusions The wT1 mapping quantitative method combined with Gd-EOB-DTPA was not capable of discerning the inflammation grade in a rat model of early-stage NASH.
Collapse
Affiliation(s)
- Qian Wan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hao Peng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Feng Liu
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, China
| | - Xiaoyi Liu
- Departments of Radiology, Peking University People’s Hospital, Beijing, China
| | - Chuanli Cheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Changjun Tie
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jie Deng
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jianxun Lyu
- Department of Radiology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yizhen Jia
- Departments of Research Services, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yi Wang
- Departments of Radiology, Peking University People’s Hospital, Beijing, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dong Liang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chao Zou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
24
|
Boncan DAT, Yu Y, Zhang M, Lian J, Vardhanabhuti V. Machine learning prediction of hepatic steatosis using body composition parameters: A UK Biobank Study. NPJ AGING 2024; 10:4. [PMID: 38195699 PMCID: PMC10776620 DOI: 10.1038/s41514-023-00127-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 10/16/2023] [Indexed: 01/11/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as the most prevalent chronic liver disease worldwide, yet detection has remained largely based on surrogate serum biomarkers, elastography or biopsy. In this study, we used a total of 2959 participants from the UK biobank cohort and established the association of dual-energy X-ray absorptiometry (DXA)-derived body composition parameters and leveraged machine learning models to predict NAFLD. Hepatic steatosis reference was based on MRI-PDFF which has been extensively validated previously. We found several significant associations with traditional measurements such as abdominal obesity, as defined by waist-to-hip ratio (OR = 2.50 (male), 3.35 (female)), android-gynoid ratio (OR = 3.35 (male), 6.39 (female)) and waist circumference (OR = 1.79 (male), 3.80 (female)) with hepatic steatosis. Similarly, A Body Shape Index (Quantile 4 OR = 1.89 (male), 5.81 (female)), and for fat mass index, both overweight (OR = 6.93 (male), 2.83 (female)) and obese (OR = 14.12 (male), 5.32 (female)) categories were likewise significantly associated with hepatic steatosis. DXA parameters were shown to be highly associated such as visceral adipose tissue mass (OR = 8.37 (male), 19.03 (female)), trunk fat mass (OR = 8.64 (male), 25.69 (female)) and android fat mass (OR = 7.93 (male), 21.77 (female)) with NAFLD. We trained machine learning classifiers with logistic regression and two histogram-based gradient boosting ensembles for the prediction of hepatic steatosis using traditional body composition indices and DXA parameters which achieved reasonable performance (AUC = 0.83-0.87). Based on SHapley Additive exPlanations (SHAP) analysis, DXA parameters that had the largest contribution to the classifiers were the features predicted with significant association with NAFLD. Overall, this study underscores the potential utility of DXA as a practical and potentially opportunistic method for the screening of hepatic steatosis.
Collapse
Affiliation(s)
- Delbert Almerick T Boncan
- Snowhill Science Ltd, Units 801-803, Level 8, Core C, Hong Kong SAR, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Yan Yu
- Snowhill Science Ltd, Units 801-803, Level 8, Core C, Hong Kong SAR, China
| | - Miaoru Zhang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jie Lian
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Varut Vardhanabhuti
- Snowhill Science Ltd, Units 801-803, Level 8, Core C, Hong Kong SAR, China.
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
25
|
Machado MV. MASLD treatment-a shift in the paradigm is imminent. Front Med (Lausanne) 2023; 10:1316284. [PMID: 38146424 PMCID: PMC10749497 DOI: 10.3389/fmed.2023.1316284] [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: 10/10/2023] [Accepted: 11/24/2023] [Indexed: 12/27/2023] Open
Abstract
MASLD prevalence is growing towards the leading cause of end-stage liver disease. Up to today, the most effective treatment is weight loss. Weight loss interventions are moving from lifestyle changes to bariatric surgery or endoscopy, and, more recently, to a new wave of anti-obesity drugs that can compete with bariatric surgery. Liver-targeted therapy is a necessity for those patients who already present liver fibrosis. The field is moving fast, and in the near future, we will testify to a disruptive change in MASLD treatment, similar to the paradigm-shift that occurred for hepatitis C almost one decade ago with direct antiviral agents.
Collapse
Affiliation(s)
- Mariana Verdelho Machado
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Hospital de Vila Franca de Xira, Vila Franca de Xira, Portugal
| |
Collapse
|
26
|
Li J, Lu X, Zhu Z, Kalutkiewicz KJ, Mounajjed T, Therneau TM, Venkatesh SK, Sui Y, Glaser KJ, Hoodeshenas S, Manduca A, Shah VH, Ehman RL, Allen AM, Yin M. Head-to-head comparison of magnetic resonance elastography-based liver stiffness, fat fraction, and T1 relaxation time in identifying at-risk NASH. Hepatology 2023; 78:1200-1208. [PMID: 37080558 PMCID: PMC10521779 DOI: 10.1097/hep.0000000000000417] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/14/2023] [Accepted: 03/24/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND AND AIMS The presence of at-risk NASH is associated with an increased risk of cirrhosis and complications. Therefore, noninvasive identification of at-risk NASH with an accurate biomarker is a critical need for pharmacologic therapy. We aim to explore the performance of several magnetic resonance (MR)-based imaging parameters in diagnosing at-risk NASH. APPROACH AND RESULTS This prospective clinical trial (NCT02565446) includes 104 paired MR examinations and liver biopsies performed in patients with suspected or diagnosed NAFLD. Magnetic resonance elastography-assessed liver stiffness (LS), 6-point Dixon-derived proton density fat fraction (PDFF), and single-point saturation-recovery acquisition-calculated T1 relaxation time were explored. Among all predictors, LS showed the significantly highest accuracy in diagnosing at-risk NASH [AUC LS : 0.89 (0.82, 0.95), AUC PDFF : 0.70 (0.58, 0.81), AUC T1 : 0.72 (0.61, 0.82), z -score test z >1.96 for LS vs any of others]. The optimal cutoff value of LS to identify at-risk NASH patients was 3.3 kPa (sensitivity: 79%, specificity: 82%, negative predictive value: 91%), whereas the optimal cutoff value of T1 was 850 ms (sensitivity: 75%, specificity: 63%, and negative predictive value: 87%). PDFF had the highest performance in diagnosing NASH with any fibrosis stage [AUC PDFF : 0.82 (0.72, 0.91), AUC LS : 0.73 (0.63, 0.84), AUC T1 : 0.72 (0.61, 0.83), |z| <1.96 for all]. CONCLUSION Magnetic resonance elastography-assessed LS alone outperformed PDFF, and T1 in identifying patients with at-risk NASH for therapeutic trials.
Collapse
Affiliation(s)
- Jiahui Li
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Xin Lu
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Zhu
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Taofic Mounajjed
- Division of Anatomic Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Terry M. Therneau
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Yi Sui
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin J. Glaser
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Armando Manduca
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vijay H. Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Alina M. Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Meng Yin
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
27
|
Liu CY, Noda C, van der Geest RJ, Triaire B, Kassai Y, Bluemke DA, Lima JAC. Sex-specific associations in multiparametric 3 T MRI measurements in adult livers. Abdom Radiol (NY) 2023; 48:3072-3078. [PMID: 37378865 DOI: 10.1007/s00261-023-03981-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND MRI relaxometry mapping and proton density fat fraction (PDFF) have been proposed for the evaluation of hepatic fibrosis. However, sex-specific relationships of age and body fat with these MRI parameters have not been studied in detail among adults without clinically manifest hepatic disease. We aimed to determine the sex-specific correlation of multiparametric MRI parameters with age and body fat and to evaluate their interplay associations. METHODS 147 study participants (84 women, mean age 48±14 years, range 19-85 years) were prospectively enrolled. 3 T MRI including T1, T2 and T1ρ mapping and PDFF and R2* map were acquired. Visceral and subcutaneous fat were measured on the fat images from Dixon water-fat separation sequence. RESULTS All MRI parameters demonstrated sex difference except for T1ρ. PDFF was more related to visceral than subcutaneous fat. Per 100 ml gain of visceral or subcutaneous fat is associated with 1 or 0.4% accretion of liver fat, respectively. PDFF and R2* were higher in men (both P = 0.01) while T1 and T2 were higher in women (both P < 0.01). R2* was positively but T1 and T2 were negatively associated with age in women (all P < 0.01), while T1ρ was positively related to age in men (P < 0.05). In all studies, R2* was positively and T1ρ was negatively associated with PDFF (both P <0.0001). CONCLUSION Visceral fat plays an essential role in the elevated liver fat. When using MRI parametric measures for liver disease evaluation, the interplay between these parameters should be considered.
Collapse
Affiliation(s)
| | - Chikara Noda
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rob J van der Geest
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - David A Bluemke
- Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - João A C Lima
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Tipirneni-Sajja A, Brasher S, Shrestha U, Johnson H, Morin C, Satapathy SK. Quantitative MRI of diffuse liver diseases: techniques and tissue-mimicking phantoms. MAGMA (NEW YORK, N.Y.) 2023; 36:529-551. [PMID: 36515810 DOI: 10.1007/s10334-022-01053-z] [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: 08/03/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022]
Abstract
Quantitative magnetic resonance imaging (MRI) techniques are emerging as non-invasive alternatives to biopsy for assessment of diffuse liver diseases of iron overload, steatosis and fibrosis. For testing and validating the accuracy of these techniques, phantoms are often used as stand-ins to human tissue to mimic diffuse liver pathologies. However, currently, there is no standardization in the preparation of MRI-based liver phantoms for mimicking iron overload, steatosis, fibrosis or a combination of these pathologies as various sizes and types of materials are used to mimic the same liver disease. Liver phantoms that mimic specific MR features of diffuse liver diseases observed in vivo are important for testing and calibrating new MRI techniques and for evaluating signal models to accurately quantify these features. In this study, we review the liver morphology associated with these diffuse diseases, discuss the quantitative MR techniques for assessing these liver pathologies, and comprehensively examine published liver phantom studies and discuss their benefits and limitations.
Collapse
Affiliation(s)
- Aaryani Tipirneni-Sajja
- Department of Biomedical Engineering, The University of Memphis, Memphis, TN, USA.
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Sarah Brasher
- Department of Biomedical Engineering, The University of Memphis, Memphis, TN, USA
| | - Utsav Shrestha
- Department of Biomedical Engineering, The University of Memphis, Memphis, TN, USA
| | - Hayden Johnson
- Department of Biomedical Engineering, The University of Memphis, Memphis, TN, USA
| | - Cara Morin
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sanjaya K Satapathy
- Northwell Health Center for Liver Diseases and Transplantation, Northshore University Hospital/Northwell Health, Manhasset, NY, USA
| |
Collapse
|
29
|
Tan HC, Shumbayawonda E, Beyer C, Cheng LTE, Low A, Lim CH, Eng A, Chan WH, Lee PC, Tay MF, Kin S, Chang JPE, Bee YM, Goh GBB. Multiparametric Magnetic Resonance Imaging and Magnetic Resonance Elastography to Evaluate the Early Effects of Bariatric Surgery on Nonalcoholic Fatty Liver Disease. Int J Biomed Imaging 2023; 2023:4228321. [PMID: 37521027 PMCID: PMC10372298 DOI: 10.1155/2023/4228321] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 08/01/2023] Open
Abstract
Background Bariatric surgery is the most effective treatment for morbid obesity and reduces the severity of nonalcoholic fatty liver disease (NAFLD) in the long term. Less is known about the effects of bariatric surgery on liver fat, inflammation, and fibrosis during the early stages following bariatric surgery. Aims This exploratory study utilises advanced imaging methods to investigate NAFLD and fibrosis changes during the early metabolic transitional period following bariatric surgery. Methods Nine participants with morbid obesity underwent sleeve gastrectomy. Multiparametric MRI (mpMRI) and magnetic resonance elastography (MRE) were performed at baseline, during the immediate (1 month), and late (6 months) postsurgery period. Liver fat was measured using proton density fat fraction (PDFF), disease activity using iron-correct T1 (cT1), and liver stiffness using MRE. Repeated measured ANOVA was used to assess longitudinal changes and Dunnett's method for multiple comparisons. Results All participants (Age 45.1 ± 9.0 years, BMI 39.7 ± 5.3 kg/m2) had elevated hepatic steatosis at baseline (PDFF >5%). In the immediate postsurgery period, PDFF decreased significantly from 14.1 ± 7.4% to 8.9 ± 4.4% (p = 0.016) and cT1 from 826.9 ± 80.6 ms to 768.4 ± 50.9 ms (p = 0.047). These improvements continued to the later postsurgery period. Bariatric surgery did not reduce liver stiffness measurements. Conclusion Our findings support using MRI as a noninvasive tool to monitor NAFLD in patient with morbid obesity during the early stages following bariatric surgery.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Alvin Eng
- Singapore General Hospital, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Tavaglione F, Jamialahmadi O, De Vincentis A, Qadri S, Mowlaei ME, Mancina RM, Ciociola E, Carotti S, Perrone G, Bruni V, Gallo IF, Tuccinardi D, Bianco C, Prati D, Manfrini S, Pozzilli P, Picardi A, Caricato M, Yki-Järvinen H, Valenti L, Vespasiani-Gentilucci U, Romeo S. Development and Validation of a Score for Fibrotic Nonalcoholic Steatohepatitis. Clin Gastroenterol Hepatol 2023; 21:1523-1532.e1. [PMID: 35421583 DOI: 10.1016/j.cgh.2022.03.044] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Noninvasive assessment of histological features of nonalcoholic fatty liver disease (NAFLD) has been an intensive research area over the last decade. Herein, we aimed to develop a simple noninvasive score using routine laboratory tests to identify, among individuals at high risk for NAFLD, those with fibrotic nonalcoholic steatohepatitis (NASH) defined as NASH, NAFLD activity score ≥4, and fibrosis stage ≥2. METHODS The derivation cohort included 264 morbidly obese individuals undergoing intraoperative liver biopsy in Rome, Italy. The best predictive model was developed and internally validated using a bootstrapping stepwise logistic regression analysis (2000 bootstrap samples). Performance was estimated by the area under the receiver operating characteristic curve (AUROC). External validation was assessed in 3 independent European cohorts (Finland, n = 370; Italy, n = 947; England, n = 5368) of individuals at high risk for NAFLD. RESULTS The final predictive model, designated as Fibrotic NASH Index (FNI), combined aspartate aminotransferase, high-density lipoprotein cholesterol, and hemoglobin A1c. The performance of FNI for fibrotic NASH was satisfactory in both derivation and external validation cohorts (AUROC = 0.78 and AUROC = 0.80-0.95, respectively). In the derivation cohort, rule-out and rule-in cutoffs were 0.10 for sensitivity ≥0.89 (negative predictive value, 0.93) and 0.33 for specificity ≥0.90 (positive predictive value, 0.57), respectively. In the external validation cohorts, sensitivity ranged from 0.87 to 1 (negative predictive value, 0.99-1) and specificity from 0.73 to 0.94 (positive predictive value, 0.12-0.49) for rule-out and rule-in cutoff, respectively. CONCLUSION FNI is an accurate, simple, and affordable noninvasive score which can be used to screen for fibrotic NASH in individuals with dysmetabolism in primary health care.
Collapse
Affiliation(s)
- Federica Tavaglione
- Clinical Medicine and Hepatology Unit, Department of Internal Medicine and Geriatrics, Campus Bio-Medico University, Rome, Italy; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden.
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Antonio De Vincentis
- Internal Medicine Unit, Department of Internal Medicine and Geriatrics, Campus Bio-Medico University, Rome, Italy; Clinical Lecturer of Internal Medicine, Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Sami Qadri
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Mohammad Erfan Mowlaei
- Department of Computer & Information Sciences, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Rosellina Margherita Mancina
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Ester Ciociola
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Simone Carotti
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, Campus Bio-Medico University, Rome, Italy; Predictive Molecular Diagnostic Unit, Department of Pathology, Campus Bio-Medico University Hospital, Rome, Italy
| | - Giuseppe Perrone
- Predictive Molecular Diagnostic Unit, Department of Pathology, Campus Bio-Medico University Hospital, Rome, Italy; Research Unit of Pathology, Campus Bio-Medico University, Rome, Italy
| | - Vincenzo Bruni
- Bariatric Surgery Unit, Campus Bio-Medico University, Rome, Italy
| | | | - Dario Tuccinardi
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Rome, Italy
| | - Cristiana Bianco
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Daniele Prati
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Silvia Manfrini
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Rome, Italy
| | - Paolo Pozzilli
- Department of Endocrinology and Diabetes, University Campus Bio-Medico, Rome, Italy
| | - Antonio Picardi
- Clinical Medicine and Hepatology Unit, Department of Internal Medicine and Geriatrics, Campus Bio-Medico University, Rome, Italy
| | - Marco Caricato
- Unit of Colon and Rectal Surgery, Department of General Surgery, Campus Bio-Medico University, Rome, Italy
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Luca Valenti
- Translational Medicine, Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy; Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milano, Italy
| | - Umberto Vespasiani-Gentilucci
- Clinical Medicine and Hepatology Unit, Department of Internal Medicine and Geriatrics, Campus Bio-Medico University, Rome, Italy.
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden; Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden; Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.
| |
Collapse
|
31
|
Roberts NT, Tamada D, Muslu Y, Hernando D, Reeder SB. Confounder-corrected T 1 mapping in the liver through simultaneous estimation of T 1 , PDFF, R 2 * , and B 1 + in a single breath-hold acquisition. Magn Reson Med 2023; 89:2186-2203. [PMID: 36656152 PMCID: PMC10139739 DOI: 10.1002/mrm.29590] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/23/2022] [Accepted: 01/01/2023] [Indexed: 01/20/2023]
Abstract
PURPOSE Quantitative volumetric T1 mapping in the liver has the potential to aid in the detection, diagnosis, and quantification of liver fibrosis, inflammation, and spatially resolved liver function. However, accurate measurement of hepatic T1 is confounded by the presence of fat and inhomogeneous B 1 + $$ {B}_1^{+} $$ excitation. Furthermore, scan time constraints related to respiratory motion require tradeoffs of reduced volumetric coverage and/or increased acquisition time. This work presents a novel 3D acquisition and estimation method for confounder-corrected T1 measurement over the entire liver within a single breath-hold through simultaneous estimation of T1 , fat and B 1 + $$ {B}_1^{+} $$ . THEORY AND METHODS The proposed method combines chemical shift encoded MRI and variable flip angle MRI with a B 1 + $$ {B}_1^{+} $$ mapping technique to enable confounder-corrected T1 mapping. The method was evaluated theoretically and demonstrated in both phantom and in vivo acquisitions at 1.5 and 3.0T. At 1.5T, the method was evaluated both pre- and post- contrast enhancement in healthy volunteers. RESULTS The proposed method demonstrated excellent linear agreement with reference inversion-recovery spin-echo based T1 in phantom acquisitions at both 1.5 and 3.0T, with minimal bias (5.2 and 45 ms, respectively) over T1 ranging from 200-1200 ms. In vivo results were in general agreement with reference saturation-recovery based 2D T1 maps (SMART1 Map, GE Healthcare). CONCLUSION The proposed 3D T1 mapping method accounts for fat and B 1 + $$ {B}_1^{+} $$ confounders through simultaneous estimation of T1 , B 1 + $$ {B}_1^{+} $$ , PDFF and R 2 * $$ {R}_2^{\ast } $$ . It demonstrates strong linear agreement with reference T1 measurements, with low bias and high precision, and can achieve full liver coverage in a single breath-hold.
Collapse
Affiliation(s)
- Nathan T Roberts
- Department of Radiology, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Electrical and Computer Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Daiki Tamada
- Department of Radiology, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Yavuz Muslu
- Department of Radiology, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Diego Hernando
- Department of Radiology, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Electrical and Computer Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Medical Physics, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Scott B Reeder
- Department of Radiology, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Medical Physics, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Medicine, University of Wisconsin - Madison, Madison, Wisconsin, USA.,Department of Emergency Medicine, University of Wisconsin - Madison, Madison, Wisconsin, USA
| |
Collapse
|
32
|
McCracken C, Raisi-Estabragh Z, Veldsman M, Raman B, Dennis A, Husain M, Nichols TE, Petersen SE, Neubauer S. Multi-organ imaging demonstrates the heart-brain-liver axis in UK Biobank participants. Nat Commun 2022; 13:7839. [PMID: 36543768 PMCID: PMC9772225 DOI: 10.1038/s41467-022-35321-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Medical imaging provides numerous insights into the subclinical changes that precede serious diseases such as heart disease and dementia. However, most imaging research either describes a single organ system or draws on clinical cohorts with small sample sizes. In this study, we use state-of-the-art multi-organ magnetic resonance imaging phenotypes to investigate cross-sectional relationships across the heart-brain-liver axis in 30,444 UK Biobank participants. Despite controlling for an extensive range of demographic and clinical covariates, we find significant associations between imaging-derived phenotypes of the heart (left ventricular structure, function and aortic distensibility), brain (brain volumes, white matter hyperintensities and white matter microstructure), and liver (liver fat, liver iron and fibroinflammation). Simultaneous three-organ modelling identifies differentially important pathways across the heart-brain-liver axis with evidence of both direct and indirect associations. This study describes a potentially cumulative burden of multiple-organ dysfunction and provides essential insight into multi-organ disease prevention.
Collapse
Affiliation(s)
- Celeste McCracken
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 9DU, UK
| | - Zahra Raisi-Estabragh
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, EC1A 7BE, UK.
| | - Michele Veldsman
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), University of Oxford, Oxford, UK
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Betty Raman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 9DU, UK
| | - Andrea Dennis
- Perspectum Ltd, Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Masud Husain
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), University of Oxford, Oxford, UK
- Department of Experimental Psychology, University of Oxford, Oxford, UK
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Thomas E Nichols
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Oxford Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Steffen E Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, EC1A 7BE, UK
- Health Data Research UK, London, UK
- The Alan Turing Institute, London, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 9DU, UK
| |
Collapse
|
33
|
Wang L, Yang JD, Yoo CC, Lai KKY, Braun J, McGovern DPB, Xie Y, Pandol SJ, Lu SC, Li D. Magnetic resonance imaging for characterization of hepatocellular carcinoma metabolism. Front Physiol 2022; 13:1056511. [PMID: 36589457 PMCID: PMC9800006 DOI: 10.3389/fphys.2022.1056511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/14/2022] [Indexed: 12/23/2022] Open
Abstract
With a better understanding of the pathophysiological and metabolic changes in hepatocellular carcinoma (HCC), multiparametric and novel functional magnetic resonance (MR) and positron emission tomography (PET) techniques have received wide interest and are increasingly being applied in preclinical and clinical research. These techniques not only allow for non-invasive detection of structural, functional, and metabolic changes in malignant tumor cells but also characterize the tumor microenvironment (TME) and the interactions of malignant tumor cells with the TME, which has hypoxia and low pH, resulting from the Warburg effect and accumulation of metabolites produced by tumor cells and other cellular components. The heterogeneity and complexity of the TME require a combination of images with various parameters and modalities to characterize tumors and guide therapy. This review focuses on the value of multiparametric magnetic resonance imaging and PET/MR in evaluating the structural and functional changes of HCC and in detecting metabolites formed owing to HCC and the TME.
Collapse
Affiliation(s)
- Lixia Wang
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ju Dong Yang
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States,Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States,Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Charles C. Yoo
- Office of the Medical Director 1st MRI, Los Angeles, CA, United States
| | - Keane K. Y. Lai
- Department of Molecular Medicine, Beckman Research Institute of City of Hope and City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Jonathan Braun
- F. Widjaja Inflammatory Bowel Disease Institute, Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Dermot P. B. McGovern
- F. Widjaja Inflammatory Bowel Disease Institute, Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Yibin Xie
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Stephen J. Pandol
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Debiao Li
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States,Department of Bioengineering, University of California, Los Angeles, CA, United States,*Correspondence: Debiao Li,
| |
Collapse
|
34
|
Akbari P, Sosina OA, Bovijn J, Landheer K, Nielsen JB, Kim M, Aykul S, De T, Haas ME, Hindy G, Lin N, Dinsmore IR, Luo JZ, Hectors S, Geraghty B, Germino M, Panagis L, Parasoglou P, Walls JR, Halasz G, Atwal GS, Regeneron Genetics Center Della GattaGiusy1, DiscovEHR Collaboration, Jones M, LeBlanc MG, Still CD, Carey DJ, Giontella A, Orho-Melander M, Berumen J, Kuri-Morales P, Alegre-Díaz J, Torres JM, Emberson JR, Collins R, Rader DJ, Zambrowicz B, Murphy AJ, Balasubramanian S, Overton JD, Reid JG, Shuldiner AR, Cantor M, Abecasis GR, Ferreira MAR, Sleeman MW, Gusarova V, Altarejos J, Harris C, Economides AN, Idone V, Karalis K, Della Gatta G, Mirshahi T, Yancopoulos GD, Melander O, Marchini J, Tapia-Conyer R, Locke AE, Baras A, Verweij N, Lotta LA. Multiancestry exome sequencing reveals INHBE mutations associated with favorable fat distribution and protection from diabetes. Nat Commun 2022; 13:4844. [PMID: 35999217 PMCID: PMC9399235 DOI: 10.1038/s41467-022-32398-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 07/28/2022] [Indexed: 12/13/2022] Open
Abstract
Body fat distribution is a major, heritable risk factor for cardiometabolic disease, independent of overall adiposity. Using exome-sequencing in 618,375 individuals (including 160,058 non-Europeans) from the UK, Sweden and Mexico, we identify 16 genes associated with fat distribution at exome-wide significance. We show 6-fold larger effect for fat-distribution associated rare coding variants compared with fine-mapped common alleles, enrichment for genes expressed in adipose tissue and causal genes for partial lipodystrophies, and evidence of sex-dimorphism. We describe an association with favorable fat distribution (p = 1.8 × 10-09), favorable metabolic profile and protection from type 2 diabetes (~28% lower odds; p = 0.004) for heterozygous protein-truncating mutations in INHBE, which encodes a circulating growth factor of the activin family, highly and specifically expressed in hepatocytes. Our results suggest that inhibin βE is a liver-expressed negative regulator of adipose storage whose blockade may be beneficial in fat distribution-associated metabolic disease.
Collapse
Affiliation(s)
- Parsa Akbari
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Olukayode A. Sosina
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Jonas Bovijn
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Karl Landheer
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Jonas B. Nielsen
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Minhee Kim
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Senem Aykul
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Tanima De
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Mary E. Haas
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - George Hindy
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Nan Lin
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Ian R. Dinsmore
- grid.280776.c0000 0004 0394 1447Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA USA
| | - Jonathan Z. Luo
- grid.280776.c0000 0004 0394 1447Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA USA
| | - Stefanie Hectors
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Benjamin Geraghty
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Mary Germino
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Lampros Panagis
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Prodromos Parasoglou
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Johnathon R. Walls
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Gabor Halasz
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Gurinder S. Atwal
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | | | | | - Marcus Jones
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Michelle G. LeBlanc
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Christopher D. Still
- grid.280776.c0000 0004 0394 1447Geisinger Obesity Institute, Geisinger Health System, Danville, PA USA
| | - David J. Carey
- grid.280776.c0000 0004 0394 1447Geisinger Obesity Institute, Geisinger Health System, Danville, PA USA
| | - Alice Giontella
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden ,grid.5611.30000 0004 1763 1124Department of Medicine, University of Verona, Verona, Italy
| | - Marju Orho-Melander
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Jaime Berumen
- grid.9486.30000 0001 2159 0001Unidad de Medicina Experimental de la Facultad de Medicina de la Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Pablo Kuri-Morales
- grid.9486.30000 0001 2159 0001Unidad de Medicina Experimental de la Facultad de Medicina de la Universidad Nacional Autónoma de México, Mexico City, Mexico ,grid.419886.a0000 0001 2203 4701Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, Mexico
| | - Jesus Alegre-Díaz
- grid.9486.30000 0001 2159 0001Unidad de Medicina Experimental de la Facultad de Medicina de la Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jason M. Torres
- grid.4991.50000 0004 1936 8948MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK ,grid.4991.50000 0004 1936 8948Clinical Trial Service Unit & Epidemiological Studies Unit Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jonathan R. Emberson
- grid.4991.50000 0004 1936 8948MRC Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK ,grid.4991.50000 0004 1936 8948Clinical Trial Service Unit & Epidemiological Studies Unit Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Rory Collins
- grid.4991.50000 0004 1936 8948Clinical Trial Service Unit & Epidemiological Studies Unit Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel J. Rader
- grid.25879.310000 0004 1936 8972Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Brian Zambrowicz
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Andrew J. Murphy
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Suganthi Balasubramanian
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - John D. Overton
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Jeffrey G. Reid
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Alan R. Shuldiner
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Michael Cantor
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Goncalo R. Abecasis
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Manuel A. R. Ferreira
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Mark W. Sleeman
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Viktoria Gusarova
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Judith Altarejos
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Charles Harris
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Aris N. Economides
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA ,grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Vincent Idone
- grid.418961.30000 0004 0472 2713Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Katia Karalis
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Giusy Della Gatta
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Tooraj Mirshahi
- grid.280776.c0000 0004 0394 1447Geisinger Obesity Institute, Geisinger Health System, Danville, PA USA
| | | | - Olle Melander
- grid.4514.40000 0001 0930 2361Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden ,grid.411843.b0000 0004 0623 9987Department of Emergency and Internal Medicine, Skåne University Hospital, Malmö, Sweden
| | - Jonathan Marchini
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Roberto Tapia-Conyer
- grid.419886.a0000 0001 2203 4701Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, Mexico
| | - Adam E. Locke
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Aris Baras
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY, USA.
| | - Niek Verweij
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| | - Luca A. Lotta
- grid.418961.30000 0004 0472 2713Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY USA
| |
Collapse
|
35
|
Topiwala A, Wang C, Ebmeier KP, Burgess S, Bell S, Levey DF, Zhou H, McCracken C, Roca-Fernández A, Petersen SE, Raman B, Husain M, Gelernter J, Miller KL, Smith SM, Nichols TE. Associations between moderate alcohol consumption, brain iron, and cognition in UK Biobank participants: Observational and mendelian randomization analyses. PLoS Med 2022; 19:e1004039. [PMID: 35834561 PMCID: PMC9282660 DOI: 10.1371/journal.pmed.1004039] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 06/01/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Brain iron deposition has been linked to several neurodegenerative conditions and reported in alcohol dependence. Whether iron accumulation occurs in moderate drinkers is unknown. Our objectives were to investigate evidence in support of causal relationships between alcohol consumption and brain iron levels and to examine whether higher brain iron represents a potential pathway to alcohol-related cognitive deficits. METHODS AND FINDINGS Observational associations between brain iron markers and alcohol consumption (n = 20,729 UK Biobank participants) were compared with associations with genetically predicted alcohol intake and alcohol use disorder from 2-sample mendelian randomization (MR). Alcohol intake was self-reported via a touchscreen questionnaire at baseline (2006 to 2010). Participants with complete data were included. Multiorgan susceptibility-weighted magnetic resonance imaging (9.60 ± 1.10 years after baseline) was used to ascertain iron content of each brain region (quantitative susceptibility mapping (QSM) and T2*) and liver tissues (T2*), a marker of systemic iron. Main outcomes were susceptibility (χ) and T2*, measures used as indices of iron deposition. Brain regions of interest included putamen, caudate, hippocampi, thalami, and substantia nigra. Potential pathways to alcohol-related iron brain accumulation through elevated systemic iron stores (liver) were explored in causal mediation analysis. Cognition was assessed at the scan and in online follow-up (5.82 ± 0.86 years after baseline). Executive function was assessed with the trail-making test, fluid intelligence with puzzle tasks, and reaction time by a task based on the "Snap" card game. Mean age was 54.8 ± 7.4 years and 48.6% were female. Weekly alcohol consumption was 17.7 ± 15.9 units and never drinkers comprised 2.7% of the sample. Alcohol consumption was associated with markers of higher iron (χ) in putamen (β = 0.08 standard deviation (SD) [95% confidence interval (CI) 0.06 to 0.09], p < 0.001), caudate (β = 0.05 [0.04 to 0.07], p < 0.001), and substantia nigra (β = 0.03 [0.02 to 0.05], p < 0.001) and lower iron in the thalami (β = -0.06 [-0.07 to -0.04], p < 0.001). Quintile-based analyses found these associations in those consuming >7 units (56 g) alcohol weekly. MR analyses provided weak evidence these relationships are causal. Genetically predicted alcoholic drinks weekly positively associated with putamen and hippocampus susceptibility; however, these associations did not survive multiple testing corrections. Weak evidence for a causal relationship between genetically predicted alcohol use disorder and higher putamen susceptibility was observed; however, this was not robust to multiple comparisons correction. Genetically predicted alcohol use disorder was associated with serum iron and transferrin saturation. Elevated liver iron was observed at just >11 units (88 g) alcohol weekly c.f. <7 units (56 g). Systemic iron levels partially mediated associations of alcohol intake with brain iron. Markers of higher basal ganglia iron associated with slower executive function, lower fluid intelligence, and slower reaction times. The main limitations of the study include that χ and T2* can reflect changes in myelin as well as iron, alcohol use was self-reported, and MR estimates can be influenced by genetic pleiotropy. CONCLUSIONS To the best of our knowledge, this study represents the largest investigation of moderate alcohol consumption and iron homeostasis to date. Alcohol consumption above 7 units weekly associated with higher brain iron. Iron accumulation represents a potential mechanism for alcohol-related cognitive decline.
Collapse
Affiliation(s)
- Anya Topiwala
- Nuffield Department Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
| | - Chaoyue Wang
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
| | - Klaus P. Ebmeier
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, United Kingdom
| | - Stephen Burgess
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Steven Bell
- Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Daniel F. Levey
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States of America
| | - Hang Zhou
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States of America
| | - Celeste McCracken
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | | | - Steffen E. Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kingdom
- Health Data Research UK, London, United Kingdom
- Alan Turing Institute, London, United Kingdom
| | - Betty Raman
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Masud Husain
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
- Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, United Kingdom
- Division of Clinical Neurology, John Radcliffe Hospital, Oxford University Hospitals Trust, Oxford, United Kingdom
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Psychiatry, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, United States of America
| | - Karla L. Miller
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
| | - Stephen M. Smith
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
| | - Thomas E. Nichols
- Nuffield Department Population Health, Big Data Institute, University of Oxford, Oxford, United Kingdom
- Wellcome Centre for Integrative Neuroimaging (WIN FMRIB), Oxford University, Oxford, United Kingdom
| |
Collapse
|
36
|
Waddell T, Bagur A, Cunha D, Thomaides‐Brears H, Banerjee R, Cuthbertson DJ, Brown E, Cusi K, Després J, Brady M. Greater ectopic fat deposition and liver fibroinflammation and lower skeletal muscle mass in people with type 2 diabetes. Obesity (Silver Spring) 2022; 30:1231-1238. [PMID: 35475573 PMCID: PMC9321120 DOI: 10.1002/oby.23425] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/27/2022] [Accepted: 02/16/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Type 2 diabetes (T2D) is associated with significant end-organ damage and ectopic fat accumulation. Multiparametric magnetic resonance imaging (MRI) can provide a rapid, noninvasive assessment of multiorgan and body composition. The primary objective of this study was to investigate differences in visceral adiposity, ectopic fat accumulation, body composition, and relevant biomarkers between people with and without T2D. METHODS Participant demographics, routine biochemistry, and multiparametric MRI scans of the liver, pancreas, visceral and subcutaneous adipose tissue, and skeletal muscle were analyzed from 266 participants (131 with T2D and 135 without T2D) who were matched for age, gender, and BMI. Wilcoxon and χ2 tests were performed to calculate differences between groups. RESULTS Participants with T2D had significantly elevated liver fat (7.4% vs. 5.3%, p = 0.011) and fibroinflammation (as assessed by corrected T1 [cT1]; 730 milliseconds vs. 709 milliseconds, p = 0.019), despite there being no differences in liver biochemistry, serum aspartate aminotransferase (p = 0.35), or alanine transaminase concentration (p = 0.11). Significantly lower measures of skeletal muscle index (45.2 cm2 /m2 vs. 50.6 cm2 /m2 , p = 0.003) and high-density lipoprotein cholesterol (1.1 mmol/L vs. 1.3 mmol/L, p < 0.0001) were observed in participants with T2D. CONCLUSIONS Multiparametric MRI revealed significantly elevated liver fat and fibroinflammation in participants with T2D, despite normal liver biochemistry. This study corroborates findings of significantly lower measures of skeletal muscle and high-density lipoprotein cholesterol in participants with T2D versus those without T2D.
Collapse
Affiliation(s)
- Tom Waddell
- Department of Engineering ScienceThe University of OxfordOxfordUK
- Perspectum Ltd.OxfordUK
| | - Alexandre Bagur
- Department of Engineering ScienceThe University of OxfordOxfordUK
- Perspectum Ltd.OxfordUK
| | | | | | | | - Daniel J. Cuthbertson
- Department of Cardiovascular and Metabolic MedicineInstitute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
- Liverpool University Hospitals NHS Foundation TrustLiverpoolUK
| | - Emily Brown
- Department of Cardiovascular and Metabolic MedicineInstitute of Life Course and Medical SciencesUniversity of LiverpoolLiverpoolUK
- Liverpool University Hospitals NHS Foundation TrustLiverpoolUK
| | | | | | | |
Collapse
|
37
|
Seidelin AS, Nordestgaard BG, Tybjærg-Hansen A, Yaghootkar H, Stender S. A rare genetic variant in the manganese transporter SLC30A10 and elevated liver enzymes in the general population. Hepatol Int 2022; 16:702-711. [PMID: 35397106 DOI: 10.1007/s12072-022-10331-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/14/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND A genetic variant in the manganese transporter SLC30A10 (rs188273166, p.Thr95Ile) was associated with increased plasma alanine transaminase (ALT) in a recent genome-wide association study in the UK Biobank (UKB). The aims of the present study were to test the association of rs188273166 with ALT in an independent cohort, and to begin to assess the clinical, hepatic, and biochemical phenotypes associated with the variant. METHODS We included n = 334,886 white participants from UKB, including 14,462 with hepatic magnetic resonance imaging (MRI), and n = 113,612 individuals from the Copenhagen City Heart Study and the Copenhagen General Population Study combined. RESULTS Genotyping SLC30A10 p.Thr95Ile identified 816 heterozygotes in the UKB and 111 heterozygotes in the Copenhagen cohort. Compared to noncarriers, heterozygotes had 4 and 5 U/L higher levels of ALT in the UKB and Copenhagen cohort, respectively, and 3 U/L higher plasma aspartate transaminase and gamma-glutamyl transferase in the UKB. Heterozygotes also had higher corrected T1 on liver MRI, a marker of hepatic inflammation (p = 4 × 10-7), but no change in MRI-quantified steatosis (p = 0.57). Plasma manganese was within the normal range in nine heterozygotes that provided new blood samples. SLC30A10 p.Thr95Ile heterozygotes had an eightfold increased risk of biliary tract cancer in UKB (p = 4 × 10-7), but this association was not replicated in the Copenhagen cohort. CONCLUSIONS SLC30A10 p.Thr95Ile was associated with elevated liver enzymes in two large general population cohorts, and with MRI-quantified hepatic inflammation. A rare genetic variant (p.Thr95Ile) in the manganese transporter SLC30A10 is associated with elevated plasma alanine transaminase (ALT) and higher corrected T1 on liver MRI, markers of liver inflammation. These data support that the variant may increase the risk of liver disease.
Collapse
Affiliation(s)
- Anne-Sofie Seidelin
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Børge Grønne Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hanieh Yaghootkar
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine (CIRTM), Brunel University London, Uxbridge, UK
- Research Centre for Optimal Health, School of Life Sciences, University of Westminster, London, UK
| | - Stefan Stender
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, 2100, Copenhagen, Denmark.
- Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
38
|
Ghavamian A, Liu C, Kang B, Yuan X, Wang X, Gao L, Zhao X. Liver T1 relaxation time of the 'normal liver' in healthy Asians: measurement with MOLLI and B 1-corrected VFA methods at 3T. Br J Radiol 2022; 95:20211008. [PMID: 35324344 PMCID: PMC10993984 DOI: 10.1259/bjr.20211008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/15/2022] [Accepted: 02/02/2022] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES Liver T1 is a potential magnetic resonance imaging biomarker for liver diseases. This study aimed to determine the T1 relaxation time of the normal liver (PDFF<5%) in healthy Asian volunteers using modified look-locker inversion recovery (MOLLI) and B1 inhomogeneity-corrected variable flip angle (B1-corrected VFA). METHODS 60 healthy Asian volunteers without focal or diffuse liver disease underwent a liver scan at 3T magnetic resonance. Proton density fat fraction (PDFF) and liver stiffness measurements were applied for the quantification of liver fat and fibrosis. T1 mapping was performed with MOLLI and B1-corrected VFA sequences. Bland-Altman, linear regression, Student t-test, and one-way analysis of variance were used for statistical analysis. RESULTS The mean T1 relaxation times of the whole liver were 901 ± 34 ms by MOLLI, and 948 ± 29 ms by B1-corrected VFA in healthy volunteers. There was a strong correlation (r = 0.86, p < 0.0001) for liver T1 between two T1 mapping methods. There were significant differences between the right and left lobes in liver T1 relaxation times using both methods (p < 0.05). Gender and Asian ethnic disparities had no impact on liver T1 relaxation times. CONCLUSION T1 relaxation times of the normal liver (PDFF<5%) in healthy volunteers were established by MOLLI and B1-corrected VFA T1 mapping methods at 3T. It may provide suitable and robust baseline values for the assessment of liver diseases. ADVANCES IN KNOWLEDGE Gender and Asian ethnic disparities do not impact liver T1 relaxation time measurements.
Collapse
Affiliation(s)
- Armin Ghavamian
- Department of Radiology, Shandong Provincial Hospital, Cheeloo
College of Medicine, Shandong University,
Shandong, China
| | - Cuihong Liu
- Department of Radiology, Shandong Provincial Hospital, Cheeloo
College of Medicine, Shandong University,
Shandong, China
- Shandong Provincial Hospital Affiliated to Shandong First
Medical University, Shandong University,
Shandong, China
| | - Bing Kang
- Shandong Provincial Hospital Affiliated to Shandong First
Medical University, Shandong University,
Shandong, China
| | - Xianshun Yuan
- Shandong Provincial Hospital Affiliated to Shandong First
Medical University, Shandong University,
Shandong, China
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital, Cheeloo
College of Medicine, Shandong University,
Shandong, China
- Shandong Provincial Hospital Affiliated to Shandong First
Medical University, Shandong University,
Shandong, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital
affiliated to Shandong University, Shandong Clinical Medical Center of
Endocrinology and Metabolism, Institute of Endocrinology and Metabolism,
Shandong Academy of Clinical Medicine,
Shandong, China
| | - Xinya Zhao
- Department of Radiology, Shandong Provincial Hospital, Cheeloo
College of Medicine, Shandong University,
Shandong, China
- Shandong Provincial Hospital Affiliated to Shandong First
Medical University, Shandong University,
Shandong, China
| |
Collapse
|
39
|
Duan T, Jiang HY, Ling WW, Song B. Noninvasive imaging of hepatic dysfunction: A state-of-the-art review. World J Gastroenterol 2022; 28:1625-1640. [PMID: 35581963 PMCID: PMC9048786 DOI: 10.3748/wjg.v28.i16.1625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/17/2021] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
Hepatic dysfunction represents a wide spectrum of pathological changes, which can be frequently found in hepatitis, cholestasis, metabolic diseases, and focal liver lesions. As hepatic dysfunction is often clinically silent until advanced stages, there remains an unmet need to identify affected patients at early stages to enable individualized intervention which can improve prognosis. Passive liver function tests include biochemical parameters and clinical grading systems (e.g., the Child-Pugh score and Model for End-Stage Liver Disease score). Despite widely used and readily available, these approaches provide indirect and limited information regarding hepatic function. Dynamic quantitative tests of liver function are based on clearance capacity tests such as the indocyanine green (ICG) clearance test. However, controversial results have been reported for the ICG clearance test in relation with clinical outcome and the accuracy is easily affected by various factors. Imaging techniques, including ultrasound, computed tomography, and magnetic resonance imaging, allow morphological and functional assessment of the entire hepatobiliary system, hence demonstrating great potential in evaluating hepatic dysfunction noninvasively. In this article, we provide a state-of-the-art summary of noninvasive imaging modalities for hepatic dysfunction assessment along the pathophysiological track, with special emphasis on the imaging modality comparison and selection for each clinical scenario.
Collapse
Affiliation(s)
- Ting Duan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Han-Yu Jiang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wen-Wu Ling
- Department of Medical Ultrasound, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
40
|
Rijnberg FM, Westenberg JJM, van Assen HC, Juffermans JF, Kroft LJM, van den Boogaard PJ, Terol Espinosa de Los Monteros C, Warmerdam EG, Leiner T, Grotenhuis HB, Jongbloed MRM, Hazekamp MG, Roest AAW, Lamb HJ. 4D flow cardiovascular magnetic resonance derived energetics in the Fontan circulation correlate with exercise capacity and CMR-derived liver fibrosis/congestion. J Cardiovasc Magn Reson 2022; 24:21. [PMID: 35346249 PMCID: PMC8962091 DOI: 10.1186/s12968-022-00854-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 03/15/2022] [Indexed: 12/12/2022] Open
Abstract
AIM This study explores the relationship between in vivo 4D flow cardiovascular magnetic resonance (CMR) derived blood flow energetics in the total cavopulmonary connection (TCPC), exercise capacity and CMR-derived liver fibrosis/congestion. BACKGROUND The Fontan circulation, in which both caval veins are directly connected with the pulmonary arteries (i.e. the TCPC) is the palliative approach for single ventricle patients. Blood flow efficiency in the TCPC has been associated with exercise capacity and liver fibrosis using computational fluid dynamic modelling. 4D flow CMR allows for assessment of in vivo blood flow energetics, including kinetic energy (KE) and viscous energy loss rate (EL). METHODS Fontan patients were prospectively evaluated between 2018 and 2021 using a comprehensive cardiovascular and liver CMR protocol, including 4D flow imaging of the TCPC. Peak oxygen consumption (VO2) was determined using cardiopulmonary exercise testing (CPET). Iron-corrected whole liver T1 (cT1) mapping was performed as a marker of liver fibrosis/congestion. KE and EL in the TCPC were computed from 4D flow CMR and normalized for inflow. Furthermore, blood flow energetics were compared between standardized segments of the TCPC. RESULTS Sixty-two Fontan patients were included (53% male, 17.3 ± 5.1 years). Maximal effort CPET was obtained in 50 patients (peak VO2 27.1 ± 6.2 ml/kg/min, 56 ± 12% of predicted). Both KE and EL in the entire TCPC (n = 28) were significantly correlated with cT1 (r = 0.50, p = 0.006 and r = 0.39, p = 0.04, respectively), peak VO2 (r = - 0.61, p = 0.003 and r = - 0.54, p = 0.009, respectively) and % predicted peak VO2 (r = - 0.44, p = 0.04 and r = - 0.46, p = 0.03, respectively). Segmental analysis indicated that the most adverse flow energetics were found in the Fontan tunnel and left pulmonary artery. CONCLUSIONS Adverse 4D flow CMR derived KE and EL in the TCPC correlate with decreased exercise capacity and increased levels of liver fibrosis/congestion. 4D flow CMR is promising as a non-invasive screening tool for identification of patients with adverse TCPC flow efficiency.
Collapse
Affiliation(s)
- Friso M Rijnberg
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333ZA, Leiden, The Netherlands.
| | - Jos J M Westenberg
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans C van Assen
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Joe F Juffermans
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lucia J M Kroft
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | - Tim Leiner
- Department of Radiology, Utrecht Medical Center, Utrecht, The Netherlands
| | - Heynric B Grotenhuis
- Department of Pediatric Cardiology, Utrecht Medical Center, Utrecht, The Netherlands
| | - Monique R M Jongbloed
- Department of Cardiology and Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mark G Hazekamp
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333ZA, Leiden, The Netherlands
| | - Arno A W Roest
- Department of Pediatric Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hildo J Lamb
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
41
|
Mojtahed A, Núñez L, Connell J, Fichera A, Nicholls R, Barone A, Marieiro M, Puddu A, Arya Z, Ferreira C, Ridgway G, Kelly M, Lamb HJ, Caseiro-Alves F, Brady JM, Banerjee R. Repeatability and reproducibility of deep-learning-based liver volume and Couinaud segment volume measurement tool. Abdom Radiol (NY) 2022; 47:143-151. [PMID: 34605963 PMCID: PMC8776724 DOI: 10.1007/s00261-021-03262-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 12/27/2022]
Abstract
Purpose Volumetric and health assessment of the liver is crucial to avoid poor post-operative outcomes following liver resection surgery. No current methods allow for concurrent and accurate measurement of both Couinaud segmental volumes for future liver remnant estimation and liver health using non-invasive imaging. In this study, we demonstrate the accuracy and precision of segmental volume measurements using new medical software, Hepatica™. Methods MRI scans from 48 volunteers from three previous studies were used in this analysis. Measurements obtained from Hepatica™ were compared with OsiriX. Time required per case with each software was also compared. The performance of technicians and experienced radiologists as well as the repeatability and reproducibility were compared using Bland–Altman plots and limits of agreement. Results High levels of agreement and lower inter-operator variability for liver volume measurements were shown between Hepatica™ and existing methods for liver volumetry (mean Dice score 0.947 ± 0.010). A high consistency between technicians and experienced radiologists using the device for volumetry was shown (± 3.5% of total liver volume) as well as low inter-observer and intra-observer variability. Tight limits of agreement were shown between repeated Couinaud segment volume (+ 3.4% of whole liver), segmental liver fibroinflammation and segmental liver fat measurements in the same participant on the same scanner and between different scanners. An underestimation of whole-liver volume was observed between three non-reference scanners. Conclusion Hepatica™ produces accurate and precise whole-liver and Couinaud segment volume and liver tissue characteristic measurements. Measurements are consistent between trained technicians and experienced radiologists. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s00261-021-03262-x.
Collapse
Affiliation(s)
- Amirkasra Mojtahed
- Division of Abdominal Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Luis Núñez
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - John Connell
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK.
| | | | - Rowan Nicholls
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Angela Barone
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Mariana Marieiro
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Anthony Puddu
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Zobair Arya
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Carlos Ferreira
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Ged Ridgway
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Matt Kelly
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Hildo J Lamb
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - J Michael Brady
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| | - Rajarshi Banerjee
- Perspectum Ltd., Gemini One, 5520 John Smith Drive, Oxford, OX4 2LL, UK
| |
Collapse
|
42
|
Simchick G, Zhao R, Hamilton G, Reeder SB, Hernando D. Spectroscopy-based multi-parametric quantification in subjects with liver iron overload at 1.5T and 3T. Magn Reson Med 2021; 87:597-613. [PMID: 34554595 DOI: 10.1002/mrm.29021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/13/2021] [Accepted: 09/07/2021] [Indexed: 01/02/2023]
Abstract
PURPOSE To evaluate the precision profile (repeatability and reproducibility) of quantitative STEAM-MRS and to determine the relationships between multiple MR biomarkers of chronic liver disease in subjects with iron overload at both 1.5 Tesla (T) and 3T. METHODS MRS data were acquired in patients with known or suspected liver iron overload. Two STEAM-MRS sequences (multi-TE and multi-TE-TR) were acquired at both 1.5T and 3T (same day), including test-retest acquisition. Each acquisition enabled estimation of R1, R2, and FWHM (each separately for water and fat); and proton density fat fraction. The test-retest repeatability and reproducibility across acquisition modes (multi-TE vs. multi-TE-TR) of the estimates were evaluated using intraclass correlation coefficients, linear regression, and Bland-Altman analyses. Multi-parametric relationships between parameters at each field strength, across field strengths, and with liver iron concentration were also evaluated using linear and nonlinear regression. RESULTS Fifty-six (n = 56) subjects (10 to 73 years, 37 males/19 females) were successfully recruited. Both STEAM-MRS sequences demonstrated good-to-excellent precision (intraclass correlation coefficient ≥ 0.81) for the quantification of R1water , R2water , FWHMwater , and proton density fat fraction at both 1.5T and 3T. Additionally, several moderate (R2 = 0.50 to 0.69) to high (R2 ≥ 0.70) correlations were observed between biomarkers, across field strengths, and with liver iron concentration. CONCLUSIONS Over a broad range of liver iron concentration, STEAM-MRS enables rapid and precise measurement of multiple biomarkers of chronic liver disease. By evaluating the multi-parametric relationships between biomarkers, this work may advance the comprehensive MRS-based assessment of chronic liver disease and may help establish biomarkers of chronic liver disease.
Collapse
Affiliation(s)
- Gregory Simchick
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ruiyang Zhao
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Gavin Hamilton
- Department of Radiology, University of California, San Diego, California, USA
| | - Scott B Reeder
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Emergency Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Diego Hernando
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
43
|
Arndtz K, Shumbayawonda E, Hodson J, Eddowes PJ, Dennis A, Thomaides-Brears H, Mouchti S, Kelly MD, Banerjee R, Neubauer S, Hirschfield GM. Multiparametric Magnetic Resonance Imaging, Autoimmune Hepatitis, and Prediction of Disease Activity. Hepatol Commun 2021; 5:1009-1020. [PMID: 34141986 PMCID: PMC8183180 DOI: 10.1002/hep4.1687] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/05/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Noninvasive monitoring of disease activity in autoimmune hepatitis (AIH) has potential advantages for patients for whom liver biopsy is invasive and with risk. We sought to understand the association of multiparametric magnetic resonance imaging (mpMRI) with clinical course of patients with AIH. We prospectively recruited 62 patients (median age, 55 years; 82% women) with clinically confirmed AIH. At recruitment, patients underwent mpMRI with LiverMultiScan alongside clinical investigations, which were repeated after 12-18 months. Associations between iron-corrected T1 (cT1) and other markers of disease were investigated at baseline and at follow-up. Discriminative performance of cT1, liver stiffness, and enhanced liver fibrosis (ELF) to identify those who failed to maintain remission over follow-up was investigated using the areas under the receiver operating characteristic curves (AUCs). Baseline cT1 correlated with alanine aminotransferase (Spearman's correlation coefficient [r S] = 0.28, P = 0.028), aspartate aminotransferase (r S = 0.26, P = 0.038), international normalized ratio (r S = 0.35 P = 0.005), Model for End-Stage Liver Disease (r S = 0.32, P = 0.020), ELF (r S = 0.29, P = 0.022), and liver stiffness r S = 0.51, P < 0.001). After excluding those not in remission at baseline (n = 12), 32% of the remainder failed to maintain remission during follow-up. Failure to maintain remission was associated with significant increases in cT1 over follow-up (AUC, 0.71; 95% confidence interval [CI], 0.52-0.90; P = 0.035) but not with changes in liver stiffness (AUC, 0.68; 95% CI, 0.49-0.87; P = 0.067) or ELF (AUC, 0.57; 95% CI, 0.37-0.78; P = 0.502). cT1 measured at baseline was a significant predictor of future loss of biochemical remission (AUC, 0.68; 95% CI, 0.53-0.83; P = 0.042); neither liver stiffness (AUC, 0.53; 95% CI, 0.34-0.71; P = 0.749) nor ELF (AUC, 0.52; 95% CI, 0.33-0.70; P = 0.843) were significant predictors of loss of biochemical remission. Conclusion: Noninvasive mpMRI has potential to contribute to risk stratification in patients with AIH.
Collapse
Affiliation(s)
- Katherine Arndtz
- Centre for Liver and Gastrointestinal ResearchNational Institute for Health Research (NIHR) Birmingham Liver Biomedical Research CentreBirminghamUnited Kingdom.,University Hospitals Birmingham National Health Service (NHS) Foundation TrustBirminghamUnited Kingdom
| | | | - James Hodson
- University Hospitals Birmingham National Health Service (NHS) Foundation TrustBirminghamUnited Kingdom
| | - Peter J Eddowes
- Centre for Liver and Gastrointestinal ResearchNational Institute for Health Research (NIHR) Birmingham Liver Biomedical Research CentreBirminghamUnited Kingdom.,NIHR Nottingham Biomedical Research CentreNottingham University Hospitals NHS Trust and University of NottinghamNottinghamUnited Kingdom
| | | | | | | | | | | | | | - Gideon M Hirschfield
- Centre for Liver and Gastrointestinal ResearchNational Institute for Health Research (NIHR) Birmingham Liver Biomedical Research CentreBirminghamUnited Kingdom.,Toronto Centre for Liver DiseaseUniversity Health NetworkTorontoONCanada
| |
Collapse
|
44
|
Breit HC, Block KT, Winkel DJ, Gehweiler JE, Henkel MJ, Weikert T, Stieltjes B, Boll DT, Heye TJ. Evaluation of liver fibrosis and cirrhosis on the basis of quantitative T1 mapping: Are acute inflammation, age and liver volume confounding factors? Eur J Radiol 2021; 141:109789. [PMID: 34051684 DOI: 10.1016/j.ejrad.2021.109789] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/28/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE To evaluate potential confounding factors in the quantitative assessment of liver fibrosis and cirrhosis using T1 relaxation times. METHODS The study population is based on a radiology-information-system database search for abdominal MRI performed from July 2018 to April 2019 at our institution. After applying exclusion criteria 200 (59 ± 16 yrs) remaining patients were retrospectively included. 93 patients were defined as liver-healthy, 40 patients without known fibrosis or cirrhosis, and 67 subjects had a clinically or biopsy-proven liver fibrosis or cirrhosis. T1 mapping was performed using a slice based look-locker approach. A ROI based analysis of the left and the right liver was performed. Fat fraction, R2*, liver volume, laboratory parameters, sex, and age were evaluated as potential confounding factors. RESULTS T1 values were significantly lower in healthy subjects without known fibrotic changes (1.5 T MRI: 575 ± 56 ms; 3 T MRI: 857 ± 128 ms) compared to patients with acute liver disease (1.5 T MRI: 657 ± 73 ms, p < 0.0001; 3 T MRI: 952 ± 37 ms, p = 0.028) or known fibrosis or cirrhosis (1.5 T MRI: 644 ± 83 ms, p < 0.0001; 3 T MRI: 995 ± 150 ms, p = 0.018). T1 values correlated moderately with the Child-Pugh stage at 1.5 T (p = 0.01, ρ = 0.35). CONCLUSION T1 mapping is a capable predictor for detection of liver fibrosis and cirrhosis. Especially age is not a confounding factor and, hence, age-independent thresholds can be defined. Acute liver diseases are confounding factors and should be ruled out before employing T1-relaxometry based thresholds to screen for patients with liver fibrosis or cirrhosis.
Collapse
Affiliation(s)
- Hanns C Breit
- Department of Radiology, University Hospital Basel, Basel, Switzerland.
| | - Kai T Block
- NYU Langone Medical Center, New York City, United States
| | - David J Winkel
- Department of Radiology, University Hospital Basel, Basel, Switzerland
| | | | - Maurice J Henkel
- Department of Radiology, University Hospital Basel, Basel, Switzerland
| | - Thomas Weikert
- Department of Radiology, University Hospital Basel, Basel, Switzerland
| | - Bram Stieltjes
- Department of Radiology, University Hospital Basel, Basel, Switzerland
| | - Daniel T Boll
- Department of Radiology, University Hospital Basel, Basel, Switzerland
| | - Tobias J Heye
- Department of Radiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
45
|
Waterton JC. Survey of water proton longitudinal relaxation in liver in vivo. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2021; 34:779-789. [PMID: 33978944 PMCID: PMC8578172 DOI: 10.1007/s10334-021-00928-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/05/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022]
Abstract
Objective To determine the variability, and preferred values, for normal liver longitudinal water proton relaxation rate R1 in the published literature. Methods Values of mean R1 and between-subject variance were obtained from literature searching. Weighted means were fitted to a heuristic and to a model. Results After exclusions, 116 publications (143 studies) remained, representing apparently normal liver in 3392 humans, 99 mice and 249 rats. Seventeen field strengths were included between 0.04 T and 9.4 T. Older studies tended to report higher between-subject coefficients of variation (CoV), but for studies published since 1992, the median between-subject CoV was 7.4%, and in half of those studies, measured R1 deviated from model by 8.0% or less. Discussion The within-study between-subject CoV incorporates repeatability error and true between-subject variation. Between-study variation also incorporates between-population variation, together with bias from interactions between methodology and physiology. While quantitative relaxometry ultimately requires validation with phantoms and analysis of propagation of errors, this survey allows investigators to compare their own R1 and variability values with the range of existing literature. Supplementary Information The online version contains supplementary material available at 10.1007/s10334-021-00928-x.
Collapse
Affiliation(s)
- John Charles Waterton
- Centre for Imaging Sciences, Division of Informatics Imaging and Data Sciences, School of Health Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester, M13 9PL, UK. .,Bioxydyn Ltd, Rutherford House, Manchester Science Park, Pencroft Way, Manchester, M15 6SZ, UK.
| |
Collapse
|
46
|
Imajo K, Tetlow L, Dennis A, Shumbayawonda E, Mouchti S, Kendall TJ, Fryer E, Yamanaka S, Honda Y, Kessoku T, Ogawa Y, Yoneda M, Saito S, Kelly C, Kelly MD, Banerjee R, Nakajima A. Quantitative multiparametric magnetic resonance imaging can aid non-alcoholic steatohepatitis diagnosis in a Japanese cohort. World J Gastroenterol 2021; 27:609-623. [PMID: 33642832 PMCID: PMC7901049 DOI: 10.3748/wjg.v27.i7.609] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/17/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Non-invasive assessment of non-alcoholic steatohepatitis (NASH) is increasing in desirability due to the invasive nature and costs associated with the current form of assessment; liver biopsy. Quantitative multiparametric magnetic resonance imaging (mpMRI) to measure liver fat (proton density fat fraction) and fibroinflammatory disease [iron-corrected T1 (cT1)], as well as elastography techniques [vibration-controlled transient elastography (VCTE) liver stiffness measure], magnetic resonance elastography (MRE) and 2D Shear-Wave elastography (SWE) to measure stiffness and fat (controlled attenuated parameter, CAP) are emerging alternatives which could be utilised as safe surrogates to liver biopsy.
AIM To evaluate the agreement of non-invasive imaging modalities with liver biopsy, and their subsequent diagnostic accuracy for identifying NASH patients.
METHODS From January 2019 to February 2020, Japanese patients suspected of NASH were recruited onto a prospective, observational study and were screened using non-invasive imaging techniques; mpMRI with LiverMultiScan®, VCTE, MRE and 2D-SWE. Patients were subsequently biopsied, and samples were scored by three independent pathologists. The diagnostic performances of the non-invasive imaging modalities were assessed using area under receiver operating characteristic curve (AUC) with the median of the histology scores as the gold standard diagnoses. Concordance between all three independent pathologists was further explored using Krippendorff’s alpha (a) from weighted kappa statistics.
RESULTS N = 145 patients with mean age of 60 (SD: 13 years.), 39% females, and 40% with body mass index ≥ 30 kg/m2 were included in the analysis. For identifying patients with NASH, MR liver fat and cT1 were the strongest performing individual measures (AUC: 0.80 and 0.75 respectively), and the mpMRI metrics combined (cT1 and MR liver fat) were the overall best non-invasive test (AUC: 0.83). For identifying fibrosis ≥ 1, MRE performed best (AUC: 0.97), compared to VCTE-liver stiffness measure (AUC: 0.94) and 2D-SWE (AUC: 0.94). For assessment of steatosis ≥ 1, MR liver fat was the best performing non-invasive test (AUC: 0.92), compared to controlled attenuated parameter (AUC: 0.75). Assessment of the agreement between pathologists showed that concordance was best for steatosis (a = 0.58), moderate for ballooning (a = 0.40) and fibrosis (a = 0.40), and worst for lobular inflammation (a = 0.11).
CONCLUSION Quantitative mpMRI is an effective alternative to liver biopsy for diagnosing NASH and non-alcoholic fatty liver, and thus may offer clinical utility in patient management.
Collapse
Affiliation(s)
- Kento Imajo
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Louise Tetlow
- Innovation, Perspectum, Oxford OX4 2LL, United Kingdom
| | - Andrea Dennis
- Innovation, Perspectum, Oxford OX4 2LL, United Kingdom
| | | | - Sofia Mouchti
- Innovation, Perspectum, Oxford OX4 2LL, United Kingdom
| | - Timothy J Kendall
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom, Edinburgh EH16 4TJ, United Kingdom
| | - Eve Fryer
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, United Kingdom
| | - Shogi Yamanaka
- Anatomic and Clinical Pathology Department, Yokohoma City University Hospital, Yokohoma 236-0004, Japan
| | - Yasushi Honda
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Takaomi Kessoku
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Yuji Ogawa
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Masato Yoneda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Satoru Saito
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | | | - Matt D Kelly
- Innovation, Perspectum, Oxford OX4 2LL, United Kingdom
| | | | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
47
|
Sethi P, Thavanesan N, Welsh FK, Connell J, Pickles E, Kelly M, Fallowfield JA, Kendall TJ, Mole DJ, Rees M. Quantitative multiparametric MRI allows safe surgical planning in patients undergoing liver resection for colorectal liver metastases: report of two patients. BJR Case Rep 2021; 7:20200172. [PMID: 34131498 PMCID: PMC8171142 DOI: 10.1259/bjrcr.20200172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/16/2020] [Accepted: 12/26/2020] [Indexed: 01/11/2023] Open
Abstract
It is not uncommon for clinicians to encounter varying degrees of hepatic steatosis in patients undergoing resection for colorectal liver metastases (CRLM). Magnetic resonance imaging is currently the preferred investigation for identification and pre-operative planning of these patients. An objective assessment of liver quality and degree of steatosis is paramount for planning a safe resection, which is seldom provided by routine MRI sequences. We studied two patients who underwent an additional pre-operative multiparametric MRI scan (LiverMultiScanTM) as a part of an observational clinical trial (HepaT1ca, NCT03213314) to assess the quality of liver. Outcome was assessed in the form of post-hepatectomy liver failure. Both patients (Patient 1 and 2) had comparable pre-operative characteristics. Both patients were planned for an extended right hepatectomy with an estimated future liver remnant of approximately 30%. Conventional preoperative contrast MRI showed mild liver steatosis in both patients. Patient one developed post-hepatectomy liver failure leading to prolonged hospital stay compared to patient two who had uneventful post-operative course. Retrospective evaluation of multiparametric MRI scan revealed findings consistent with fibro-inflammatory disease and steatosis (cT1 829 ms, PDFF 14%) for patient 1 whereas patient two had normal parameters (cT1 735 ms, PDFF 2.4%). These findings corresponded with the resection specimen histology. Multiparametric MRI can objectively evaluate future liver health and volume which may help refine surgical decision-making and improve patient outcomes.
Collapse
Affiliation(s)
- Pulkit Sethi
- Department of Hepatobiliary Surgery, Basingstoke and North Hampshire Hospital, Basingstoke, Hampshire, United Kingdom
| | - Navamayooran Thavanesan
- Department of Hepatobiliary Surgery, Basingstoke and North Hampshire Hospital, Basingstoke, Hampshire, United Kingdom
| | - Fenella Ks Welsh
- Department of Hepatobiliary Surgery, Basingstoke and North Hampshire Hospital, Basingstoke, Hampshire, United Kingdom
| | | | | | - Matt Kelly
- Perspectum, Gemini One, Oxford, United Kingdom
| | - Jonathan A Fallowfield
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Timothy J Kendall
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Myrddin Rees
- Department of Hepatobiliary Surgery, Basingstoke and North Hampshire Hospital, Basingstoke, Hampshire, United Kingdom
| |
Collapse
|
48
|
Mole DJ, Fallowfield JA, Sherif AE, Kendall T, Semple S, Kelly M, Ridgway G, Connell JJ, McGonigle J, Banerjee R, Brady JM, Zheng X, Hughes M, Neyton L, McClintock J, Tucker G, Nailon H, Patel D, Wackett A, Steven M, Welsh F, Rees M, the HepaT1ca Study Group. Quantitative magnetic resonance imaging predicts individual future liver performance after liver resection for cancer. PLoS One 2020; 15:e0238568. [PMID: 33264327 PMCID: PMC7710097 DOI: 10.1371/journal.pone.0238568] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022] Open
Abstract
The risk of poor post-operative outcome and the benefits of surgical resection as a curative therapy require careful assessment by the clinical care team for patients with primary and secondary liver cancer. Advances in surgical techniques have improved patient outcomes but identifying which individual patients are at greatest risk of poor post-operative liver performance remains a challenge. Here we report results from a multicentre observational clinical trial (ClinicalTrials.gov NCT03213314) which aimed to inform personalised pre-operative risk assessment in liver cancer surgery by evaluating liver health using quantitative multiparametric magnetic resonance imaging (MRI). We combined estimation of future liver remnant (FLR) volume with corrected T1 (cT1) of the liver parenchyma as a representation of liver health in 143 patients prior to treatment. Patients with an elevated preoperative liver cT1, indicative of fibroinflammation, had a longer post-operative hospital stay compared to those with a cT1 within the normal range (6.5 vs 5 days; p = 0.0053). A composite score combining FLR and cT1 predicted poor liver performance in the 5 days immediately following surgery (AUROC = 0.78). Furthermore, this composite score correlated with the regenerative performance of the liver in the 3 months following resection. This study highlights the utility of quantitative MRI for identifying patients at increased risk of poor post-operative liver performance and a longer stay in hospital. This approach has the potential to inform the assessment of individualised patient risk as part of the clinical decision-making process for liver cancer surgery.
Collapse
Affiliation(s)
- Damian J. Mole
- Clinical Surgery, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Jonathan A. Fallowfield
- Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Ahmed E. Sherif
- Clinical Surgery, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, United Kingdom
- Department of HPB Surgery, National Liver Institute, Menoufia University, Shibin Elkom, Egypt
| | - Timothy Kendall
- Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
- Department of Pathology, NHS Lothian, Edinburgh, United Kingdom
| | - Scott Semple
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Matt Kelly
- Perspectum, Gemini One, Oxford, United Kingdom
| | | | | | | | | | | | - Xiaozhong Zheng
- Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Michael Hughes
- Clinical Surgery, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, United Kingdom
| | - Lucile Neyton
- Centre for Inflammation Research, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | | | - Garry Tucker
- Clinical Research Facility, NHS Lothian, Edinburgh, United Kingdom
| | - Hilary Nailon
- Clinical Research Facility, NHS Lothian, Edinburgh, United Kingdom
| | - Dilip Patel
- Clinical Radiology, NHS Lothian, Edinburgh, United Kingdom
| | | | | | - Fenella Welsh
- Hampshire Hospitals Foundation Trust, Basingstoke, United Kingdom
| | - Myrddin Rees
- Hampshire Hospitals Foundation Trust, Basingstoke, United Kingdom
| | | |
Collapse
|
49
|
Jayaswal ANA, Levick C, Selvaraj EA, Dennis A, Booth JC, Collier J, Cobbold J, Tunnicliffe EM, Kelly M, Barnes E, Neubauer S, Banerjee R, Pavlides M. Prognostic value of multiparametric magnetic resonance imaging, transient elastography and blood-based fibrosis markers in patients with chronic liver disease. Liver Int 2020; 40:3071-3082. [PMID: 32730664 DOI: 10.1111/liv.14625] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/03/2020] [Accepted: 07/22/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND & AIMS Liver cT1 , liver T1 , transient elastography (TE) and blood-based biomarkers have independently been shown to predict clinical outcomes but have not been directly compared in a single cohort of patients. Our aim was to compare these tests' prognostic value in a cohort of patients with compensated chronic liver disease. METHODS Patients with unselected compensated liver disease aetiologies had baseline assessments and were followed up for development of clinical outcomes, blinded to the imaging results. The prognostic value of non-invasive liver tests at prespecified thresholds was assessed for a combined clinical endpoint comprising ascites, variceal bleeding, hepatic encephalopathy, hepatocellular carcinoma, liver transplantation and mortality. RESULTS One hundred and ninety-seven patients (61% male) with median age of 54 years were followed up for 693 patient-years (median (IQR) 43 (26-58) months). The main diagnoses were NAFLD (41%), viral hepatitis (VH, 25%) and alcohol-related liver disease (ArLD; 14%). During follow-up 14 new clinical events, and 11 deaths occurred. Clinical outcomes were predicted by liver cT1 > 825ms with HR 9.9 (95% CI: 1.29-76.4, P = .007), TE > 8kPa with HR 7.8 (95% CI: 0.97-62.3, P = .02) and FIB-4 > 1.45 with HR 4.09 (95% CI: 0.90-18.4, P = .05). In analysis taking into account technical failure and unreliability, liver cT1 > 825 ms could predict clinical outcomes (P = .03), but TE > 8kPa could not (P = .4). CONCLUSIONS We provide further evidence that liver cT1 , TE and serum-based biomarkers can predict clinical outcomes, but when taking into account technical failure/unreliability, TE cut-offs perform worse than those of cT1 and blood biomarkers.
Collapse
Affiliation(s)
- Arjun N A Jayaswal
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Christina Levick
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Emmanuel A Selvaraj
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.,Translational Gastroenterology Unit, University of Oxford, Oxford, UK.,Oxford NIHR Biomedical Research Centre, Oxford, UK
| | | | | | - Jane Collier
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Jeremy Cobbold
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Elizabeth M Tunnicliffe
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | - Eleanor Barnes
- Peter Medawar Building, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | - Michael Pavlides
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.,Translational Gastroenterology Unit, University of Oxford, Oxford, UK.,Oxford NIHR Biomedical Research Centre, Oxford, UK
| |
Collapse
|
50
|
Alenaini W, Parkinson JRC, McCarthy JP, Goldstone AP, Wilman HR, Banerjee R, Yaghootkar H, Bell JD, Thomas EL. Ethnic Differences in Body Fat Deposition and Liver Fat Content in Two UK-Based Cohorts. Obesity (Silver Spring) 2020; 28:2142-2152. [PMID: 32939982 DOI: 10.1002/oby.22948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Differences in the content and distribution of body fat and ectopic lipids may be responsible for ethnic variations in metabolic disease susceptibility. The aim of this study was to examine the ethnic distribution of body fat in two separate UK-based populations. METHODS Anthropometry and body composition were assessed in two separate UK cohorts: the Hammersmith cohort and the UK Biobank, both comprising individuals of South Asian descent (SA), individuals of Afro-Caribbean descent (AC), and individuals of European descent (EUR). Regional adipose tissue stores and liver fat were measured by magnetic resonance techniques. RESULTS The Hammersmith cohort (n = 747) had a mean (SD) age of 41.1 (14.5) years (EUR: 374 men, 240 women; SA: 68 men, 22 women; AC: 14 men, 29 women), and the UK Biobank (n = 9,533) had a mean (SD) age of 55.5 (7.5) years (EUR: 4,483 men, 4,873 women; SA: 80 men, 43 women, AC: 31 men, 25 women). Following adjustment for age and BMI, no significant differences in visceral adipose tissue or liver fat were observed between SA and EUR individuals in the either cohort. CONCLUSIONS Our data, consistent across two independent UK-based cohorts, present a limited number of ethnic differences in the distribution of body fat depots associated with metabolic disease. These results suggest that the ethnic variation in susceptibility to features of the metabolic syndrome may not arise from differences in body fat.
Collapse
Affiliation(s)
- Wareed Alenaini
- Research Centre for Optimal Health, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, UK
| | - James R C Parkinson
- Research Centre for Optimal Health, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, UK
| | - John P McCarthy
- School of Healthcare Practice, University of Bedfordshire, Luton, Bedfordshire, UK
| | - Anthony P Goldstone
- PsychoNeuroEndocrinology Research Group, Neuro-psychopharmacology Unit, Centre for Psychiatry, Division of Brain Sciences, Imperial College London-Hammersmith Hospital, London, UK
| | - Henry R Wilman
- Research Centre for Optimal Health, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, UK
- Perspectum Diagnostics, Oxford, UK
| | | | - Hanieh Yaghootkar
- Research Centre for Optimal Health, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, UK
- Genetics of Complex Traits, Medical School, University of Exeter-Royal Devon & Exeter Hospital, Exeter, UK
- Division of Medical Sciences, Department of Health Sciences, Luleå University of Technology, Luleå, Sweden
| | - Jimmy D Bell
- Research Centre for Optimal Health, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, UK
| | - E Louise Thomas
- Research Centre for Optimal Health, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, UK
| |
Collapse
|