1
|
Sun Y, Zhang L, Huang JQ, Su J, Cui LG. Non-invasive diagnosis of pancreatic steatosis with ultrasound images using deep learning network. Heliyon 2024; 10:e37580. [PMID: 39296003 PMCID: PMC11409133 DOI: 10.1016/j.heliyon.2024.e37580] [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: 06/04/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/21/2024] Open
Abstract
Objective This study aimed to verify whether pancreatic steatosis (PS) is an independent risk factor for type 2 diabetes mellitus (T2DM). We also developed and validated a deep learning model for the diagnosis of PS using ultrasonography (US) images based on histological classifications. Methods In this retrospective study, we analysed data from 139 patients who underwent US imaging of the pancreas followed by pancreatic resection at our medical institution. Logistic regression analysis was employed to ascertain the independent predictors of T2DM. The diagnostic efficacy of the deep learning model for PS was assessed using receiver operating characteristic curve analysis and compared with traditional visual assessment methodology in US imaging. Results The incidence rate of PS in the study cohort was 64.7 %. Logistic regression analysis revealed that age (P = 0.003) and the presence of PS (P = 0.048) were independent factors associated with T2DM. The deep learning model demonstrated robust diagnostic capabilities for PS, with areas under the curve of 0.901 and 0.837, sensitivities of 0.895 and 0.920, specificities of 0.700 and 0.765, accuracies of 0.814 and 0.857, and F1-scores of 0.850 and 0.885 for the training and validation cohorts, respectively. These metrics significantly outperformed those of conventional US imaging (P < 0.001 and P = 0.045, respectively). Conclusion The deep learning model significantly enhanced the diagnostic accuracy of conventional ultrasound for PS detection. Its high sensitivity could facilitate widespread screening for PS in large populations, aiding in the early identification of individuals at an elevated risk for T2DM in routine clinical practice.
Collapse
Affiliation(s)
- Yang Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| | - Li Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| | - Jian-Qiu Huang
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| | - Jing Su
- Department of Pathology, School of Basic Medical Science, Peking University Health Science Center, Beijing, China
| | - Li-Gang Cui
- Department of Ultrasound, Peking University Third Hospital, Beijing, China
| |
Collapse
|
2
|
Ookura R, Usuki N, Miki Y. Correlation between Pancreatic Fat Deposition and Metabolic Syndrome: Relationships with Location in the Pancreas and Sex. Intern Med 2024; 63:2113-2123. [PMID: 38171856 PMCID: PMC11358728 DOI: 10.2169/internalmedicine.2450-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024] Open
Abstract
Objective To investigate the correlation between pancreatic fat deposition and metabolic syndrome (MetS) parameters, focusing on the locations of fat deposition in the pancreas and sex differences. Methods Degrees of fat deposition in the head, body, and tail of the pancreas were evaluated using computed tomography (CT). We examined the relationships between pancreatic fat deposition and the age, body mass index (BMI), visceral and subcutaneous fat, serum lipid profiles, hepatic steatosis, diabetes mellitus (DM), and hypertension (HTN). Results In this retrospective study, greater fat deposition was associated with a higher BMI, visceral and subcutaneous fat accumulation, and hepatic steatosis, with the pancreatic head showing the strongest correlation. Correlations of pancreatic fat deposition with the BMI and visceral and subcutaneous fat accumulation were stronger in females than in males, while correlations with hepatic steatosis were stronger in males than in females. In addition, a multivariate analysis did not suggest a direct causal relationship between pancreatic fat deposition and DM and HTN, but there was a significant correlation between pancreatic fat deposition in the pancreatic head and visceral fat area. Conclusion Pancreatic fat deposition, as evaluated by CT, especially in the part of the pancreatic head adjacent to the ampulla of Vater, is a sensitive indicator of MetS. The correlations between pancreatic fat deposition and MetS parameters tended to be stronger in females than in males. These results may help further elucidate the pathophysiology of MetS and provide opportunities for its diagnosis.
Collapse
Affiliation(s)
- Ryusuke Ookura
- Department of Diagnostic and Interventional Radiology, Japan Community Healthcare Organization Osaka Hospital, Japan
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, Japan
| | - Noriaki Usuki
- Department of Diagnostic and Interventional Radiology, Japan Community Healthcare Organization Osaka Hospital, Japan
| | - Yukio Miki
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, Japan
| |
Collapse
|
3
|
Gjela M, Askeland A, Mellergaard M, Drewes AM, Handberg A, Frøkjær JB. Intra-pancreatic fat deposition and its relation to obesity: a magnetic resonance imaging study. Scand J Gastroenterol 2024; 59:742-748. [PMID: 38557425 DOI: 10.1080/00365521.2024.2333365] [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: 01/04/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES Intra-pancreatic fat deposition (IPFD) is suspected to be associated with various medical conditions. This study aimed to assess pancreatic fat content in lean and obese individuals, characterize obese individuals with and without IPFD, and explore the underlying mechanisms. MATERIALS AND METHODS Sixty-two obese individuals without diabetes and 35 lean controls underwent magnetic resonance imaging (MRI) using proton density fat fraction (PDFF) maps to evaluate pancreatic and hepatic fat content, and visceral adipose tissue (VAT) content. Pancreatic fibrosis was explored by T1 relaxation time and MR elastography (MRE) measurements. Associations between pancreatic fat, measures of obesity and metabolic syndrome were examined using uni- and multivariate regression analyses. RESULTS Pancreatic PDFF was higher in obese than in lean controls (median 8.0%, interquartile range (6.1;13.3) % vs 2.6(1.7;3.9)%, p < 0.001). Obese individuals with IPFD (PDFF ≥6.2%) had higher waist circumference (114.0 ± 12.5 cm vs 105.2 ± 8.7 cm, p = 0.007) and VAT (224.9(142.1; 316.1) cm2 vs 168.2(103.4; 195.3) cm2, p < 0.001) than those without. In univariate analysis, pancreatic PDFF in obese individuals correlated with BMI (r = 0.27, p = 0.03), waist circumference (r = 0.44, p < 0.001), VAT (r = 0.37, p = 0.004), hepatic PDFF (r = 0.25, p = 0.046) and diastolic blood pressure (r = 0.32, p = 0.01). However, in multivariate analysis, only VAT was associated to pancreatic fat content. MRI measures of pancreatic fibrosis indicated no evident fibrosis in relation to increased pancreatic fat content. CONCLUSIONS Pancreatic fat content was increased in obese individuals compared with lean controls and predominantly correlated with the amount of visceral adipose tissue. Pancreatic fat content was not clearly linked to measures of pancreatic fibrosis.
Collapse
Affiliation(s)
- Mimoza Gjela
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Anders Askeland
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Maiken Mellergaard
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Asbjørn Mohr Drewes
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
| | - Aase Handberg
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Jens Brøndum Frøkjær
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
4
|
Yang T, Feng J, Yao R, Feng Q, Shen J. CT-based pancreatic radiomics predicts secondary loss of response to infliximab in biologically naïve patients with Crohn's disease. Insights Imaging 2024; 15:69. [PMID: 38472447 DOI: 10.1186/s13244-024-01637-4] [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: 11/13/2023] [Accepted: 01/27/2024] [Indexed: 03/14/2024] Open
Abstract
OBJECTIVES Predicting secondary loss of response (SLR) to infliximab (IFX) is paramount for tailoring personalized management regimens. Concurrent pancreatic manifestations in patients with Crohn's disease (CD) may correlate with SLR to anti-tumor necrosis factor treatment. This work aimed to evaluate the potential of pancreatic radiomics to predict SLR to IFX in biologic-naive individuals with CD. METHODS Three models were developed by logistic regression analyses to identify high-risk subgroup prone to SLR. The area under the curve (AUC), calibration curve, decision curve analysis (DCA), and integrated discrimination improvement (IDI) were applied for the verification of model performance. A quantitative nomogram was proposed based on the optimal prediction model, and its reliability was substantiated by 10-fold cross-validation. RESULTS In total, 184 CD patients were enrolled in the period January 2016 to February 2022. The clinical model incorporated age of onset, disease duration, disease location, and disease behavior, whereas the radiomics model consisted of five texture features. These clinical parameters and the radiomics score calculated by selected texture features were applied to build the combined model. Compared to other two models, combined model achieved favorable, significantly improved discrimination power (AUCcombined vs clinical 0.851 vs 0.694, p = 0.02; AUCcombined vs radiomics 0.851 vs 0.740, p = 0.04) and superior clinical usefulness, which was further converted into reliable nomogram with an accuracy of 0.860 and AUC of 0.872. CONCLUSIONS The first proposed pancreatic-related nomogram represents a credible, noninvasive predictive instrument to assist clinicians in accurately identifying SLR and non-SLR in CD patients. CRITICAL RELEVANCE STATEMENT This study first built a visual nomogram incorporating pancreatic texture features and clinical factors, which could facilitate clinicians to make personalized treatment decisions and optimize cost-effectiveness ratio for patients with CD. KEY POINTS • The first proposed pancreatic-related model predicts secondary loss of response for infliximab in Crohn's disease. • The model achieved satisfactory predictive accuracy, calibration ability, and clinical value. • The model-based nomogram has the potential to identify long-term failure in advance and tailor personalized management regimens.
Collapse
Affiliation(s)
- Tian Yang
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai, 200127, China
- NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), Shanghai, China
| | - Jing Feng
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai, 200127, China
- NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), Shanghai, China
| | - Ruchen Yao
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai, 200127, China
- NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), Shanghai, China
| | - Qi Feng
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pu Jian Road, Shanghai, 200127, China.
| | - Jun Shen
- Renji Hospital, School of Medicine, Shanghai Jiao Tong University; Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai Institute of Digestive Disease, 160# Pu Jian Ave, Shanghai, 200127, China.
- NHC Key Laboratory of Digestive Diseases (Renji Hospital, Shanghai Jiaotong University School of Medicine), Shanghai, China.
| |
Collapse
|
5
|
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
|
6
|
Pagkali A, Makris A, Brofidi K, Agouridis AP, Filippatos TD. Pathophysiological Mechanisms and Clinical Associations of Non-Alcoholic Fatty Pancreas Disease. Diabetes Metab Syndr Obes 2024; 17:283-294. [PMID: 38283640 PMCID: PMC10813232 DOI: 10.2147/dmso.s397643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Non-Alcoholic Fatty Pancreas disease (NAFPD), characterized by fat accumulation in pancreatic tissue, is an emerging clinical entity. However, the clinical associations, the underlying molecular drivers, and the pathophysiological mechanisms of NAFPD have not yet been characterized in detail. The NAFPD spectrum not only includes infiltration and accumulation of fat within and between pancreatic cells but also involves several inflammatory processes, dysregulation of physiological metabolic pathways, and hormonal defects. A deeper understanding of the underlying molecular mechanisms is key to correlate NAFPD with clinical entities including non-alcoholic fatty liver disease, metabolic syndrome, diabetes mellitus, atherosclerosis, as well as pancreatic cancer and pancreatitis. The aim of this review is to examine the pathophysiological mechanisms of NAFPD and to assess the possible causative/predictive risk factors of NAFPD-related clinical syndromes.
Collapse
Affiliation(s)
- Antonia Pagkali
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Makris
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kalliopi Brofidi
- Department of Internal Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Aris P Agouridis
- School of Medicine, European University Cyprus, Nicosia, Cyprus
- Department of Internal Medicine, German Oncology Center, Limassol, Cyprus
| | | |
Collapse
|
7
|
Ting L, Liyun W, Zheng W, Cao Z. PANCREATIC FAT CONTENT PLAYS AN IMPORTANT ROLE IN THE DEVELOPMENT OF TYPE 2 DIABETES MELLITUS SIMILAR TO THAT OF LIVER FAT CONTENT. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2023; 19:421-425. [PMID: 38933258 PMCID: PMC11197827 DOI: 10.4183/aeb.2023.421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Objective Type 2 diabetes mellitus (T2DM) is a major health problem worldwide. Earlier studies have reported that pancreatic fat content (PFC) and liver fat content (LFC) are risk factors for T2DM. The aim of the present study was to demonstrate the relationship between PFC, LFC and T2DM. Methods A total of 70 T2DM subjects and 30 non-diabetic volunteers who underwent Dixon-based magnetic resonance imaging (MRI) method at Yixing People's Hospital between December 2018 to December 2020 were included in the study. The three-point Dixon (3p-Dixon) method was used to measure the fat content in the pancreas and liver. Clinical indices including gender, age, body mass index (BMI), total cholesterol, triglyceride, glucose and C peptide levels were collected. The association between PFC, LFC, and OGTT-derived parameters was examined by Pearson and Spearman correlation analyses. Results T2DM subjects had higher PFC and LFC than those measured in the non-diabetic subjects (p <0.05). PFC and LFC were associated positively with OGTT-derived parameters such as insulin secretion, insulin resistance, and early- and late-phase insulin secretion in the male T2DM subjects(p <0.05), but not in the non-diabetic and female T2DM subjects. The relationship between PFC and OGTT-derived parameters was also more obvious than that for LFC in overweight and obese male patients with T2DM whose BMI was >24 kg/m2. Conclusion PFC and LFC were both associated with β-cell dysfunction and insulin resistance in males with T2DM. The relationship between PFC and β-cell dysfunction and insulin resistance was more obvious than that observed for LFC in overweight and obese male T2DM patients. More attention should therefore be paid to PFC in clinical settings.
Collapse
Affiliation(s)
- L. Ting
- Yixing People’s Hospital, Department of Endocrinology, Yixing, Jiangsu Province, China
| | - W. Liyun
- Yixing People’s Hospital, Department of Endocrinology, Yixing, Jiangsu Province, China
| | - W. Zheng
- Yixing People’s Hospital, Department of Endocrinology, Yixing, Jiangsu Province, China
| | - Z. Cao
- Yixing People’s Hospital, Department of Endocrinology, Yixing, Jiangsu Province, China
| |
Collapse
|
8
|
Janssens LP, Takahashi H, Nagayama H, Nugen F, Bamlet WR, Oberg AL, Fuemmeler E, Goenka AH, Erickson BJ, Takahashi N, Majumder S. Artificial intelligence assisted whole organ pancreatic fat estimation on magnetic resonance imaging and correlation with pancreas attenuation on computed tomography. Pancreatology 2023; 23:556-562. [PMID: 37193618 DOI: 10.1016/j.pan.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Fatty pancreas is associated with inflammatory and neoplastic pancreatic diseases. Magnetic resonance imaging (MRI) is the diagnostic modality of choice for measuring pancreatic fat. Measurements typically use regions of interest limited by sampling and variability. We have previously described an artificial intelligence (AI)-aided approach for whole pancreas fat estimation on computed tomography (CT). In this study, we aimed to assess the correlation between whole pancreas MRI proton-density fat fraction (MR-PDFF) and CT attenuation. METHODS We identified patients without pancreatic disease who underwent both MRI and CT between January 1, 2015 and June 1, 2020. 158 paired MRI and CT scans were available for pancreas segmentation using an iteratively trained convolutional neural network (CNN) with manual correction. Boxplots were generated to visualize slice-by-slice variability in 2D-axial slice MR-PDFF. Correlation between whole pancreas MR-PDFF and age, BMI, hepatic fat and pancreas CT-Hounsfield Unit (CT-HU) was assessed. RESULTS Mean pancreatic MR-PDFF showed a strong inverse correlation (Spearman -0.755) with mean CT-HU. MR-PDFF was higher in males (25.22 vs 20.87; p = 0.0015) and in subjects with diabetes mellitus (25.95 vs 22.17; p = 0.0324), and was positively correlated with age and BMI. The pancreatic 2D-axial slice-to-slice MR-PDFF variability increased with increasing mean whole pancreas MR-PDFF (Spearman 0.51; p < 0.0001). CONCLUSION Our study demonstrates a strong inverse correlation between whole pancreas MR-PDFF and CT-HU, indicating that both imaging modalities can be used to assess pancreatic fat. 2D-axial pancreas MR-PDFF is variable across slices, underscoring the need for AI-aided whole-organ measurements for objective and reproducible estimation of pancreatic fat.
Collapse
Affiliation(s)
- Laurens P Janssens
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Fred Nugen
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - William R Bamlet
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Ann L Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Eric Fuemmeler
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Ajit H Goenka
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Shounak Majumder
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Abbasoğlu A, Karçaaltıncaba M, Karaosmanoğlu AD, Özmen MN, Akata D, İdilman İS. Associations Between Hepatic and Pancreatic Steatosis with Lumbar Spinal Bone Marrow Fat: A Single-Center Magnetic Resonance Imaging Study. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2023; 34:618-625. [PMID: 37303245 PMCID: PMC10441167 DOI: 10.5152/tjg.2023.22225] [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: 03/27/2022] [Accepted: 09/12/2022] [Indexed: 06/13/2023]
Abstract
BACKGROUND To evaluate the associations between hepatic, pancreatic steatosis, and lumbar spinal bone marrow fat determined by magnetic resonance imaging-proton density fat fraction in patients with no known or suspected liver disease. METHODS A total of 200 patients who were referred to our radiology department for upper abdominal magnetic resonance imaging between November 2015 and November 2017 were included in this study. All patients underwent a magnetic resonance imaging-proton density fat fraction on a 1.5-T magnetic resonance imaging system. RESULTS The mean liver, pancreas, and lumbar magnetic resonance imaging-proton density fat fraction were 7.52 ± 4.82%, 5.25 ± 5.44%, and 46.85 ± 10.38% in the study population. There were significant correlations between liver and pancreas (rs = 0.180, P = .036), liver and lumbar (rs = 0.317, P < .001), and pancreas and lumbar magnetic resonance imaging-proton density fat fraction (rs = 0.215, P = .012) in female patients. A weak correlation was observed between liver and lumbar magnetic resonance imaging-proton density fat fraction (rs = 0.174, P = .014) in the total population. The prevalence of hepatic and pancreatic steatosis was 42.5% and 29%, respectively. The prevalence of pancreatic steatosis (42.9% vs. 22.8%, P = .004) was higher in male patients compared to female patients. In subgroup analysis, in patients with hepatic steatosis, there were higher pancreas magnetic resonance imaging-proton density fat fraction (6.07 ± 6.42% vs. 4.66 ± 4.53%, P = .036) and lumbar magnetic resonance imaging-proton density fat fraction (48.81 ± 10.01% vs. 45.40 ± 10.46%, P =.029) compared to patients without hepatic steatosis. In patients with pancreatic steatosis, there were higher liver (9.07 ± 6.08 vs. 6.87 ± 4.06, P = .009) and lumbar magnetic resonance imaging-proton density fat fraction (49.31 ± 9.13% vs.45.83 ± 10.76%, P = .032) in comparison with patients without pancreatic steatosis. CONCLUSION Based on the results of the present study, fat accumulation in liver, pancreas, and lumbar vertebra have associations with more evident in females.
Collapse
Affiliation(s)
- Akın Abbasoğlu
- Department of Radiology, Hacettepe University Faculty of Medicine, Liver Imaging Team, Ankara, Turkey
| | - Musturay Karçaaltıncaba
- Department of Radiology, Hacettepe University Faculty of Medicine, Liver Imaging Team, Ankara, Turkey
| | - Ali Devrim Karaosmanoğlu
- Department of Radiology, Hacettepe University Faculty of Medicine, Liver Imaging Team, Ankara, Turkey
| | - Mustafa Nasuh Özmen
- Department of Radiology, Hacettepe University Faculty of Medicine, Liver Imaging Team, Ankara, Turkey
| | - Deniz Akata
- Department of Radiology, Hacettepe University Faculty of Medicine, Liver Imaging Team, Ankara, Turkey
| | - İlkay S. İdilman
- Department of Radiology, Hacettepe University Faculty of Medicine, Liver Imaging Team, Ankara, Turkey
| |
Collapse
|
10
|
Story JD, Ghahremani S, Kafali SG, Shih SF, Kuwahara KJ, Calkins KL, Wu HH. Using Free-Breathing MRI to Quantify Pancreatic Fat and Investigate Spatial Heterogeneity in Children. J Magn Reson Imaging 2023; 57:508-518. [PMID: 35778376 PMCID: PMC9805469 DOI: 10.1002/jmri.28337] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND MRI acquisition for pediatric pancreatic fat quantification is limited by breath-holds (BH). Full segmentation (FS) or small region of interest (ROI) analysis methods may not account for pancreatic fat spatial heterogeneity, which may limit accuracy. PURPOSE To improve MRI acquisition and analysis for quantifying pancreatic proton-density fat fraction (pPDFF) in children by investigating free-breathing (FB)-MRI, characterizing pPDFF spatial heterogeneity, and relating pPDFF to clinical markers. STUDY TYPE Prospective. POPULATION A total of 34 children, including healthy (N = 16, 8 female) and overweight (N = 18, 5 female) subjects. FIELD STRENGTH AND SEQUENCES 3 T; multiecho gradient-echo three-dimensional (3D) stack-of-stars FB-MRI, multiecho gradient-echo 3D Cartesian BH-MRI. ASSESSMENT A radiologist measured FS- and ROI-based pPDFF on FB-MRI and BH-MRI PDFF maps, with anatomical images as references. Regional pPDFF in the pancreatic head, body, and tail were measured on FB-MRI. FS-pPDFF, ROI-pPDFF, and regional pPDFF were compared, and related to clinical markers, including hemoglobin A1c. STATISTICAL TESTS T-test, Bland-Altman analysis, Lin's concordance correlation coefficient (CCC), one-way analysis of variance, and Spearman's rank correlation coefficient were used. P < 0.05 was considered significant. RESULTS FS-pPDFF and ROI-pPDFF from FB-MRI and BH-MRI had mean difference = 0.4%; CCC was 0.95 for FS-pPDFF and 0.62 for ROI-pPDFF. FS-pPDFF was higher than ROI-pPDFF (10.4% ± 6.4% vs. 4.2% ± 2.8%). Tail-pPDFF (11.6% ± 8.1%) was higher than body-pPDFF (8.9% ± 6.3%) and head-pPDFF (8.7% ± 5.2%). Head-pPDFF and body-pPDFF positively correlated with hemoglobin A1c. DATA CONCLUSION FB-MRI pPDFF is comparable to BH-MRI. Spatial heterogeneity affects pPDFF quantification. Regional measurements of pPDFF in the head and body were correlated with hemoglobin A1c, a marker of insulin sensitivity. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jacob D. Story
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Shahnaz Ghahremani
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Sevgi Gokce Kafali
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, United States
| | - Shu-Fu Shih
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, United States
| | - Kelsey J. Kuwahara
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Kara L. Calkins
- Department of Pediatrics, Division of Neonatology and Developmental Biology, and the UCLA Children’s Discovery and Innovation Institute, University of California Los Angeles, Los Angeles, CA, United States
| | - Holden H. Wu
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
11
|
An Adapted Deep Convolutional Neural Network for Automatic Measurement of Pancreatic Fat and Pancreatic Volume in Clinical Multi-Protocol Magnetic Resonance Images: A Retrospective Study with Multi-Ethnic External Validation. Biomedicines 2022; 10:biomedicines10112991. [PMID: 36428558 PMCID: PMC9687882 DOI: 10.3390/biomedicines10112991] [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: 10/08/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Pancreatic volume and fat fraction are critical prognoses for metabolic diseases like type 2 diabetes (T2D). Magnetic Resonance Imaging (MRI) is a required non-invasive quantification method for the pancreatic fat fraction. The dramatic development of deep learning has enabled the automatic measurement of MR images. Therefore, based on MRI, we intend to develop a deep convolutional neural network (DCNN) that can accurately segment and measure pancreatic volume and fat fraction. This retrospective study involved abdominal MR images from 148 diabetic patients and 246 healthy normoglycemic participants. We randomly separated them into training and testing sets according to the proportion of 80:20. There were 2364 recognizable pancreas images labeled and pre-treated by an upgraded superpixel algorithm for a discernible pancreatic boundary. We then applied them to the novel DCNN model, mimicking the most accurate and latest manual pancreatic segmentation process. Fat phantom and erosion algorithms were employed to increase the accuracy. The results were evaluated by dice similarity coefficient (DSC). External validation datasets included 240 MR images from 10 additional patients. We assessed the pancreas and pancreatic fat volume using the DCNN and compared them with those of specialists. This DCNN employed the cutting-edge idea of manual pancreas segmentation and achieved the highest DSC (91.2%) compared with any reported models. It is the first framework to measure intra-pancreatic fat volume and fat deposition. Performance validation reflected by regression R2 value between manual operation and trained DCNN segmentation on the pancreas and pancreatic fat volume were 0.9764 and 0.9675, respectively. The performance of the novel DCNN enables accurate pancreas segmentation, pancreatic fat volume, fraction measurement, and calculation. It achieves the same segmentation level of experts. With further training, it may well surpass any expert and provide accurate measurements, which may have significant clinical relevance.
Collapse
|
12
|
Bi Y, Lin HY, Li ML, Zhou J, Sun XL. The Association Between Pancreatic Steatosis and Metabolic Syndrome: A 5-Year Follow-up Study Among a General Chinese Population. Pancreas 2022; 51:1000-1006. [PMID: 36607946 DOI: 10.1097/mpa.0000000000002138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVES To date, the complete natural history of pancreatic steatosis is unknown. This study aimed to investigate the association of fatty pancreas (FP) in the incidence of metabolic syndrome and its components among Chinese patients with a 5-year follow-up. METHODS Three independent cross-sectional surveys were carried out in 2013, 2015, and 2018. Fatty pancreas was diagnosed via transabdominal sonography. Logistic regression analysis was used to estimate the correlation between FP and metabolic syndrome. New cases of metabolic syndrome and its components were estimated by Cox proportional hazards models. RESULTS At baseline, 12,551 individuals classified into FP (n = 1010) and non-FP (n = 11,541) groups were finally enrolled. In cross-sectional analyses, odds ratio of FP was 2.378 (95% confidence interval [CI], 2.085-2.713; P < 0.001). In longitudinal analyses, FP was associated with the occurrence of metabolic syndrome (hazard ratio [HR], 3.179; 95% CI, 2.197-4.6; P < 0.001), type 2 diabetes mellitus (HR, 13.99; 95% CI, 7.865-24.883; P < 0.001), nonalcoholic fatty liver disease (HR, 31.843; 95% CI, 7.73-131.171; P < 0.001), and hypertension (HR, 12.801; 95% CI, 7.323-22.38; P < 0.001). CONCLUSIONS Pancreatic steatosis is strongly associated with the occurrence of metabolic syndrome and its components such as hypertension and diabetes.
Collapse
Affiliation(s)
- Ye Bi
- From the Department of Geriatric Endocrinology
| | - Hai-Yan Lin
- Physical Examination Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | | | - Jie Zhou
- From the Department of Geriatric Endocrinology
| | | |
Collapse
|
13
|
Fatty Pancreas-Centered Metabolic Basis of Pancreatic Adenocarcinoma: From Obesity, Diabetes and Pancreatitis to Oncogenesis. Biomedicines 2022; 10:biomedicines10030692. [PMID: 35327494 PMCID: PMC8945032 DOI: 10.3390/biomedicines10030692] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest types of cancer, and it is currently the third most common cause of cancer death in the U.S.A. Progress in the fight against PDAC has been hampered by an inability to detect it early in the overwhelming majority of patients, and also by the reduced oxygen levels and nutrient perfusion caused by new matrix formation through the activation of stromal cells in the context of desmoplasia. One harbinger of PDAC is excess intrapancreatic fat deposition, namely, fatty pancreas, which specifically affects the tumor macro- and microenvironment in the organ. Over half of PDAC patients have diabetes mellitus (DM) at the time of diagnosis, and fatty pancreas is associated with subsequent DM development. Moreover, there is a strong association between fatty pancreas and fatty liver through obesity, and a higher intrapancreatic fat percentage has been noted in acute pancreatitis patients with DM than in those without DM. All these findings suggest that the link between fatty pancreas and PDAC might occur through metabolic alterations, either DM-related or non-DM-related. Based on clinical, in vivo and in vitro evidence, the current review highlights the etiologies of fatty pancreas (including fatty infiltration and replacement) and the fatty pancreas-associated metabolic alterations involved in oncogenesis to provide crucial targets to prevent, detect, and/or effectively treat PDAC.
Collapse
|
14
|
Ware MA, Kaar JL, Behn CD, Bartlette K, Carreau AM, Lopez-Paniagua D, Scherzinger A, Xie D, Rahat H, Garcia-Reyes Y, Nadeau KJ, Cree-Green M. Pancreatic fat relates to fasting insulin and postprandial lipids but not polycystic ovary syndrome in adolescents with obesity. Obesity (Silver Spring) 2022; 30:191-200. [PMID: 34932884 PMCID: PMC10786704 DOI: 10.1002/oby.23317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/23/2021] [Accepted: 09/17/2021] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Adolescents with polycystic ovary syndrome (PCOS) and obesity can have insulin resistance, dysglycemia, and hepatic steatosis. Excess pancreatic fat may disturb insulin secretion and relate to hepatic fat. Associations between pancreatic fat fraction (PFF) and metabolic measures in PCOS were unknown. METHODS This secondary analysis included 113 sedentary, nondiabetic adolescent girls (age = 15.4 [1.9] years), with or without PCOS and BMI ≥ 90th percentile. Participants underwent fasting labs, oral glucose tolerance tests, and magnetic resonance imaging for hepatic fat fraction (HFF) and PFF. Groups were categorized by PFF (above or below the median of 2.18%) and compared. RESULTS Visceral fat and HFF were elevated in individuals with PCOS versus control individuals, but PFF was similar. PFF did not correlate with serum androgens. Higher and lower PFF groups had similar HFF, with no correlation between PFF and HFF, although hepatic steatosis was more common in those with higher PFF (≥5.0% HFF; 60% vs. 36%; p = 0.014). The higher PFF group had higher fasting insulin (p = 0.026), fasting insulin resistance (homeostatic model assessment of insulin resistance, p = 0.032; 1/fasting insulin, p = 0.028), free fatty acids (p = 0.034), and triglycerides (p = 0.004) compared with those with lower PFF. β-Cell function and insulin sensitivity were similar between groups. CONCLUSIONS Neither PCOS status nor androgens related to PFF. However, fasting insulin and postprandial lipids were worse with higher PFF.
Collapse
Affiliation(s)
- Meredith A. Ware
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Modern Human Anatomy, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Vista University College of Osteopathic Medicine, Parker, Colorado, USA
| | - Jill L. Kaar
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cecilia Diniz Behn
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, Colorado, USA
| | - Kai Bartlette
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, Colorado, USA
| | - Anne-Marie Carreau
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Department of Medicine, School of Medicine, Québec CHU Research Center, Laval University, Québec City, Québec, Canada
| | - Dan Lopez-Paniagua
- Department of Radiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ann Scherzinger
- Department of Radiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Danielle Xie
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Haseeb Rahat
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Rocky Vista University College of Osteopathic Medicine, Parker, Colorado, USA
| | - Yesenia Garcia-Reyes
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kristen J. Nadeau
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Ludeman Family Center for Women’s Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Melanie Cree-Green
- Division of Pediatric Endocrinology, Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Children’s Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Ludeman Family Center for Women’s Health Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
15
|
Filippatos TD, Alexakis K, Mavrikaki V, Mikhailidis DP. Nonalcoholic Fatty Pancreas Disease: Role in Metabolic Syndrome, "Prediabetes," Diabetes and Atherosclerosis. Dig Dis Sci 2022; 67:26-41. [PMID: 33469809 DOI: 10.1007/s10620-021-06824-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023]
Abstract
Fat accumulation in the pancreas associated with obesity and the metabolic syndrome (MetS) has been defined as "non-alcoholic fatty pancreas disease" (NAFPD). The aim of this review is to describe the association of NAFPD with obesity, MetS, type 2 diabetes mellitus (T2DM) and atherosclerosis and also increase awareness regarding NAFPD. Various methods are used for the detection and quantification of pancreatic fat accumulation that may play a significant role in the differences that have been observed in the prevalence of NAFPD. Endoscopic ultrasound provides detailed images of the pancreas and its use is expected to increase in the future. Obesity and MetS have been recognized as NAFPD risk factors. NAFPD is strongly associated with non-alcoholic fatty liver disease (NAFLD) and it seems that the presence of both may be related with aggravation of NAFLD. A role of NAFPD in the development of "prediabetes" and T2DM has also been suggested by most human studies. Accumulation of fat in pancreatic tissue possibly initiates a vicious cycle of beta-cell deterioration and further pancreatic fat accumulation. Additionally, some evidence indicates a correlation between NAFPD and atherosclerotic markers (e.g., carotid intima-media thickness). Weight loss and bariatric surgery decreases pancreatic triglyceride content but pharmacologic treatments for NAFPD have not been evaluated in specifically designed studies. Hence, NAFPD is a marker of local fat accumulation possibly associated with beta-cell function impairment, carbohydrate metabolism disorders and atherosclerosis.
Collapse
Affiliation(s)
- T D Filippatos
- Metabolic Diseases Research Unit, Internal Medicine Laboratory, School of Medicine, Faculty of Medicine, University of Crete, P.O. Box 2208, Heraklion, Crete, Greece.
| | - K Alexakis
- Metabolic Diseases Research Unit, Internal Medicine Laboratory, School of Medicine, Faculty of Medicine, University of Crete, P.O. Box 2208, Heraklion, Crete, Greece
| | - V Mavrikaki
- Metabolic Diseases Research Unit, Internal Medicine Laboratory, School of Medicine, Faculty of Medicine, University of Crete, P.O. Box 2208, Heraklion, Crete, Greece
| | - D P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), London, NW3 2QG, UK.,Mohammed Bin Rashid University (MBRU) of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
16
|
Lee MS, Lee JS, Kim BS, Kim DR, Kang KS. Quantitative Analysis of Pancreatic Fat in Children with Obesity Using Magnetic Resonance Imaging and Ultrasonography. Pediatr Gastroenterol Hepatol Nutr 2021; 24:555-563. [PMID: 34796100 PMCID: PMC8593362 DOI: 10.5223/pghn.2021.24.6.555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/19/2021] [Accepted: 08/04/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE The aim of this study was to evaluate the pancreatic fat fraction (PFF) using magnetic resonance imaging (MRI) in children with and without obesity and to correlate PFF with body mass index (BMI) z-score, hepatic fat fraction (HFF), and ultrasonography-derived pancreato-perihepatic fat index (PPHFI). METHODS This prospective study included 45 children with obesity and 19 without obesity (control group). PFF and HFF were quantitatively assessed using the abdominal multi-echo Dixon method for MRI. The PPHFI was assessed using transabdominal ultrasonography. Anthropometric, MRI, and ultrasonographic characteristics were compared between the two groups. Correlations between PFF, HFF, PPHFI, and BMI z-scores in each group were also analyzed. RESULTS The PFF, HFF, PPHFI, and BMI z-score were higher in the group with obesity than in the control group (PFF: 6.65±3.42 vs. 1.78±0.55, HFF: 19.5±13.0 vs. 2.31±1, PPHFI: 3.65 ±1.63 vs. 0.94±0.31, BMI z-score: 2.27±0.56 vs. 0.42±0.54, p<0.01, respectively). PFF was correlated with BMI z-scores, PPHFI, and HFF in the obesity group, and multivariate analysis showed that PFF was strongly correlated with BMI z-score and PPHFI (p<0.05). The BMI z-score was strongly correlated with PFF in the control group (p<0.01). CONCLUSION These results suggest that MRI-derived PFF measures are associated with childhood obesity. PFF and PPHFI were also highly correlated in the obesity group. Therefore, PFF may be an objective index of pancreatic fat content and has the potential for clinical utility as a non-invasive biomarker for the assessment of childhood obesity.
Collapse
Affiliation(s)
- Mu Sook Lee
- Department of Radiology, Keimyung University Dongsan Hospital, Daegu, Korea
| | - Jeong Sub Lee
- Department of Radiology, Jeju National University Hospital, Jeju National University College of Medicine, Jeju, Korea
| | - Bong Soo Kim
- Department of Radiology, Jeju National University Hospital, Jeju National University College of Medicine, Jeju, Korea
| | - Doo Ri Kim
- Department of Radiology, Jeju National University Hospital, Jeju National University College of Medicine, Jeju, Korea
| | - Ki Soo Kang
- Department of Pediatrics, Jeju National University Hospital, Jeju National University College of Medicine, Jeju, Korea
| |
Collapse
|
17
|
Koç U, Taydaş O. Evaluation of pancreatic steatosis prevalence and anthropometric measurements using non-contrast computed tomography. TURKISH JOURNAL OF GASTROENTEROLOGY 2021; 31:640-648. [PMID: 33090101 DOI: 10.5152/tjg.2020.19434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIMS Pancreatic steatosis (PS) is a subject of current interest and its prevalence has been reported to range from 16.1% to 30.7% using various radiological methods. This study aimed to evaluate PS prevalence with non-contrast computed tomography (CT). MATERIALS AND METHODS The non-contrast CT scans taken in 2016 and 2017 in our hospital were retrospectively screened. A total of 637 cases (320 males, 317 females) were included in the study. CT number measurements were performed from three anatomic regions of the pancreas using regions of interest (ROI) of approximately 1 cm2. The cases with a <0.7 ratio of the pancreatic over splenic CT number were accepted as quantitatively steatosis-positive. Anthropometric evaluations were undertaken by determining various parameters defined on CT. RESULTS PS was determined visually in 30.6% of the males and 29% of the females, and quantitatively in 32.8% and 30.6%, respectively. A positive agreement was determined between the quantitative and visual evaluations of steatosis (Cohen's kappa coefficient=0.587, p<0.001). Although PS was seen to be mostly diffuse, the tail region of the pancreas was determined to be the area with most steatosis. CONCLUSION PS is usually overlooked in radiology practice but it has a clinical presentation with non-insignificant prevalence. Current radiological methods are adequate in the evaluation of PS. The determination of the cut-off values for various criteria on non-contrast CT can provide more objective evaluations.
Collapse
Affiliation(s)
- Ural Koç
- Department of Radiology, Erzincan Mengucek Gazi Training and Research Hospital, Erzincan, Turkey
| | - Onur Taydaş
- Department of Radiology, Erzincan Mengucek Gazi Training and Research Hospital, Erzincan, Turkey
| |
Collapse
|
18
|
Lee EH, Kim JY, Yang HR. Association between ectopic pancreatic and hepatic fat and metabolic risk factors in children with non-alcoholic fatty liver disease. Pediatr Obes 2021; 16:e12793. [PMID: 33942524 DOI: 10.1111/ijpo.12793] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/03/2021] [Accepted: 03/13/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Few studies have reported an association between ectopic pancreatic and hepatic fat and metabolic factors in children with non-alcoholic fatty liver disease (NAFLD). OBJECTIVES We investigated this association and also the factors associated with pancreatic and hepatic fat deposition in children with NAFLD. METHODS This cross-sectional study investigated 65 children with NAFLD (49 boys, 13.0 ± 3.2 years, mean body mass index z-score 2.5 ± 1.2), who underwent liver biopsy and magnetic resonance imaging-based proton density fat fraction, as well as anthropometry, laboratory tests, body composition analysis, and hepatic fat fraction (HFF) and pancreatic fat fraction (PFF) measurements. RESULTS HFF and PFF were 4.2%-49.9% (median 24.3) and 0.4%-26.9% (median 3.8), respectively. HFF was not significantly correlated with PFF. HFF was correlated with total body fat% (r = 0.329, p = 0.010) and γ-glutamyl transpeptidase (GGT) (r = 0.260, p = 0.040), while PFF was correlated with the diastolic blood pressure (r = 0.253, p = 0.045), GGT (r = 0.335, p = 0.007) and fasting plasma glucose (r = 0.417, p = 0.001). Multiple linear regression analysis showed that HFF was significantly associated with sex, age, body fat% and GGT, whereas PFF was associated with hypertension and fasting plasma glucose levels but not insulin resistance. CONCLUSIONS HFF was associated with sex, age and body fat in children with NAFLD, while PFF was associated with hypertension and increased fasting plasma glucose, which suggests that the pathophysiology of ectopic fat accumulation varies across organs in children with NAFLD.
Collapse
Affiliation(s)
- Eun Hye Lee
- Department of Pediatrics, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, South Korea.,Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Ji Young Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hye Ran Yang
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, South Korea.,Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
19
|
Virostko J, Craddock RC, Williams JM, Triolo TM, Hilmes MA, Kang H, Du L, Wright JJ, Kinney M, Maki JH, Medved M, Waibel M, Kay TWH, Thomas HE, Greeley SAW, Steck AK, Moore DJ, Powers AC. Development of a standardized MRI protocol for pancreas assessment in humans. PLoS One 2021; 16:e0256029. [PMID: 34428220 PMCID: PMC8384163 DOI: 10.1371/journal.pone.0256029] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/29/2021] [Indexed: 11/26/2022] Open
Abstract
Magnetic resonance imaging (MRI) has detected changes in pancreas volume and other characteristics in type 1 and type 2 diabetes. However, differences in MRI technology and approaches across locations currently limit the incorporation of pancreas imaging into multisite trials. The purpose of this study was to develop a standardized MRI protocol for pancreas imaging and to define the reproducibility of these measurements. Calibrated phantoms with known MRI properties were imaged at five sites with differing MRI hardware and software to develop a harmonized MRI imaging protocol. Subsequently, five healthy volunteers underwent MRI at four sites using the harmonized protocol to assess pancreas size, shape, apparent diffusion coefficient (ADC), longitudinal relaxation time (T1), magnetization transfer ratio (MTR), and pancreas and hepatic fat fraction. Following harmonization, pancreas size, surface area to volume ratio, diffusion, and longitudinal relaxation time were reproducible, with coefficients of variation less than 10%. In contrast, non-standardized image processing led to greater variation in MRI measurements. By using a standardized MRI image acquisition and processing protocol, quantitative MRI of the pancreas performed at multiple locations can be incorporated into clinical trials comparing pancreas imaging measures and metabolic state in individuals with type 1 or type 2 diabetes.
Collapse
Affiliation(s)
- John Virostko
- Department of Diagnostic Medicine, University of Texas at Austin, Austin, Texas, United States of America
- Livestrong Cancer Institutes, University of Texas at Austin, Austin, Texas, United States of America
- Department of Oncology, University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| | - Richard C. Craddock
- Department of Diagnostic Medicine, University of Texas at Austin, Austin, Texas, United States of America
| | - Jonathan M. Williams
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Taylor M. Triolo
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Melissa A. Hilmes
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Hakmook Kang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Liping Du
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jordan J. Wright
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Mara Kinney
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Jeffrey H. Maki
- Department of Radiology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Milica Medved
- Department of Radiology, University of Chicago, Chicago, IL, United States of America
| | - Michaela Waibel
- Immunology and Diabetes Unit, St Vincent’s Institute, Fitzroy, Victoria, Australia
| | - Thomas W. H. Kay
- Immunology and Diabetes Unit, St Vincent’s Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St. Vincent’s Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| | - Helen E. Thomas
- Immunology and Diabetes Unit, St Vincent’s Institute, Fitzroy, Victoria, Australia
- Department of Medicine, St. Vincent’s Hospital, The University of Melbourne, Fitzroy, Victoria, Australia
| | - Siri Atma W. Greeley
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, Kovler Diabetes Center, University of Chicago, Chicago, IL, United States of America
| | - Andrea K. Steck
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Daniel J. Moore
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Pathology, Immunology, and Microbiology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Alvin C. Powers
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States of America
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, United States of America
| |
Collapse
|
20
|
Swislocki A. Fatty Pancreas: An Underappreciated Intersection of the Metabolic Profile and Pancreatic Adenocarcinoma. Metab Syndr Relat Disord 2021; 19:317-324. [PMID: 33656378 DOI: 10.1089/met.2020.0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Although the prevalence of pancreatic cancer is increasing, treatment strategies remain limited, and success is rare. A growing body of evidence links pancreatic cancer to pre-existing metabolic disorders, including, but not limited to, type 2 diabetes mellitus and obesity. An infrequently described finding, fatty pancreas, initially described in the context of obesity in the early 20th century, appears to be at the crossroads of type 2 diabetes and obesity on the one hand, and the development of pancreatic cancer on the other. Similarly, other conditions of the pancreas, such as intrapancreatic mucinous neoplasms, also seem to be related to diabetes while increasing the subsequent risk of pancreatic cancer. In this review, the author explores the diagnostic criteria for, and prevalence of, fatty pancreas and the potential link to other pancreatic conditions, including pancreatic cancer. Diagnostic limitations, and areas of controversy are also addressed, as are potential therapeutic approaches to fatty pancreas intended to reduce the subsequent risk of pancreatic cancer.
Collapse
Affiliation(s)
- Arthur Swislocki
- Medical Service (612/111), Veterans Affairs Northern California Health Care System (VANCHCS), Martinez, California, USA
- Department of Medicine, UC Davis School of Medicine, Sacramento, California, USA
| |
Collapse
|
21
|
Kim J, Albakheet SS, Han K, Yoon H, Lee MJ, Koh H, Kim S, Suh J, Han SJ, Ihn K, Shin HJ. Quantitative MRI Assessment of Pancreatic Steatosis Using Proton Density Fat Fraction in Pediatric Obesity. Korean J Radiol 2021; 22:1886-1893. [PMID: 34269534 PMCID: PMC8546128 DOI: 10.3348/kjr.2020.1343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/14/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022] Open
Abstract
Objective To assess the feasibility of quantitatively assessing pancreatic steatosis using magnetic resonance imaging (MRI) and its correlation with obesity and metabolic risk factors in pediatric patients. Materials and Methods Pediatric patients (≤ 18 years) who underwent liver fat quantification MRI between January 2016 and June 2019 were retrospectively included and divided into the obesity and control groups. Pancreatic proton density fat fraction (P-PDFF) was measured as the average value for three circular regions of interest (ROIs) drawn in the pancreatic head, body, and tail. Age, weight, laboratory results, and mean liver MRI values including liver PDFF (L-PDFF), stiffness on MR elastography, and T2* values were assessed for their correlation with P-PDFF using linear regression analysis. The associations between P-PDFF and metabolic risk factors, including obesity, hypertension, diabetes mellitus (DM), and dyslipidemia, were assessed using logistic regression analysis. Results A total of 172 patients (male:female = 125:47; mean ± standard deviation [SD], 13.2 ± 3.1 years) were included. The mean P-PDFF was significantly higher in the obesity group than in the control group (mean ± SD, 4.2 ± 2.5% vs. 3.4 ± 2.4%; p = 0.037). L-PDFF and liver stiffness values showed no significant correlation with P-PDFF (p = 0.235 and p = 0.567, respectively). P-PDFF was significantly associated with obesity (odds ratio 1.146, 95% confidence interval 1.006–1.307, p = 0.041), but there was no significant association with hypertension, DM, and dyslipidemia. Conclusion MRI can be used to quantitatively measure pancreatic steatosis in children. P-PDFF is significantly associated with obesity in pediatric patients.
Collapse
Affiliation(s)
- Jisoo Kim
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Korea
| | - Salman S Albakheet
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Radiology, King Faisal General Hospital, Al-Hofuf, Saudi Arabia
| | - Kyunghwa Han
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Korea
| | - Haesung Yoon
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Korea.,Severance Pediatric Liver Disease Research Group, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Jung Lee
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Korea.,Severance Pediatric Liver Disease Research Group, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hong Koh
- Severance Pediatric Liver Disease Research Group, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Pediatric Gastroenterology, Hepatology and Nutrition, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Kim
- Severance Pediatric Liver Disease Research Group, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Pediatric Gastroenterology, Hepatology and Nutrition, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Junghwan Suh
- Department of Pediatric Endocrinology, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seok Joo Han
- Severance Pediatric Liver Disease Research Group, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Pediatric Surgery, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyong Ihn
- Severance Pediatric Liver Disease Research Group, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Pediatric Surgery, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun Joo Shin
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University College of Medicine, Seoul, Korea.,Severance Pediatric Liver Disease Research Group, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Radiology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea.
| |
Collapse
|
22
|
Ajmera VH, Cachay ER, Ramers CB, Bassirian S, Singh S, Bettencourt R, Richards L, Hamilton G, Middleton M, Fowler K, Sirlin C, Loomba R. Optimal Threshold of Controlled Attenuation Parameter for Detection of HIV-Associated NAFLD With Magnetic Resonance Imaging as the Reference Standard. Clin Infect Dis 2021; 72:2124-2131. [PMID: 32975278 DOI: 10.1093/cid/ciaa429] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Controlled attenuation parameter (CAP) is an ultrasound-based point-of-care method to quantify liver fat; however, the optimal threshold for CAP to detect pathologic liver fat among persons living with human immunodeficiency virus (HIV; PLWH) is unknown. Therefore, we aimed to identify the diagnostic accuracy and optimal threshold of CAP for the detection of liver-fat among PLWH with magnetic resonance imaging proton-density fat fraction (MRI-PDFF) as the reference standard. METHODS Patients from a prospective single-center cohort of PLWH at risk for HIV-associated nonalcoholic fatty liver disease (NAFLD) who underwent contemporaneous MRI-PDFF and CAP assessment were included. Subjects with other forms of liver disease including viral hepatitis and excessive alcohol intake were excluded. Receiver operatic characteristic (ROC) curve analysis were performed to identify the optimal threshold for the detection of HIV-associated NAFLD (liver fat ≥ 5%). RESULTS Seventy PLWH (90% men) at risk for NAFLD were included. The mean (± standard deviation) age and body mass index were 48.6 (±10.2) years and 30 (± 5.3) kg/m2, respectively. The prevalence of HIV-associated NAFLD (MRI-PDFF ≥ 5%) was 80%. The M and XL probes were used for 56% and 44% of patients, respectively. The area under the ROC curve of CAP for the detection of MRI-PDFF ≥ 5% was 0.82 (0.69-0.95) at the cut-point of 285 dB/m. The positive predictive value of CAP ≥ 285 dB/m was 93.2% in this cohort with sensitivity of 73% and specificity of 78.6%. CONCLUSIONS The optimal cut-point of CAP to correctly identify HIV-associated NAFLD was 285 dB/m, is similar to previously published cut-point for primary NAFLD and may be incorporated into routine care to identify patients at risk of HIV-associated NAFLD.
Collapse
Affiliation(s)
- Veeral H Ajmera
- NAFLD Research Center, Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Edward R Cachay
- Division of Infectious Diseases, Owen Clinic, University of California San Diego, San Diego, California, USA
| | - Christian B Ramers
- Laura Rodriguez Research Institute, Family Health Centers of San Diego, San Diego, California, USA
| | - Shirin Bassirian
- NAFLD Research Center, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Seema Singh
- NAFLD Research Center, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Richele Bettencourt
- NAFLD Research Center, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Lisa Richards
- NAFLD Research Center, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Gavin Hamilton
- Liver Imaging Group, University of California, San Diego, La Jolla, California, USA
| | - Michael Middleton
- Liver Imaging Group, University of California, San Diego, La Jolla, California, USA
| | - Katie Fowler
- Liver Imaging Group, University of California, San Diego, La Jolla, California, USA
| | - Claude Sirlin
- Liver Imaging Group, University of California, San Diego, La Jolla, California, USA
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Gastroenterology, Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Epidemiology, Department of Family and Preventive Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
23
|
Idilman IS, Low HM, Gidener T, Philbrick K, Mounajjed T, Li J, Allen AM, Yin M, Venkatesh SK. Association between Visceral Adipose Tissue and Non-Alcoholic Steatohepatitis Histology in Patients with Known or Suspected Non-Alcoholic Fatty Liver Disease. J Clin Med 2021; 10:jcm10122565. [PMID: 34200525 PMCID: PMC8228492 DOI: 10.3390/jcm10122565] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 01/29/2023] Open
Abstract
(1) Purpose: To determine the association between visceral adipose tissue (VAT) and proton density fat fraction (PDFF) with magnetic resonance imaging (MRI), and hepatic steatosis (HS), non-alcoholic steatohepatitis (NASH) and hepatic fibrosis (HF) in patients with known or suspected non-alcoholic fatty liver disease (NAFLD). (2) Methods: 135 subjects that had a liver biopsy performed within 3 months (bariatric cohort) or 1 month (NAFLD cohort) of an MRI exam formed the study group. VAT volume was quantified at L2-L3 level on opposed-phase images with signal intensity-based painting using a semi-quantitative software. Liver PDFF and pancreas PDFF were calculated on fat fraction maps. Liver volume (Lvol) and spleen volume (Svol) were also calculated using a semi-automated 3D volume tool available on PACS. A histological analysis was performed by an expert hepatopathologist blinded to imaging findings. (3) Results: The mean Lvol, Svol, liver PDFF, pancreas PDFF and VAT of the study population were 2492.2 mL, 381.6 mL, 13.2%, 12.7% and 120.6 mL, respectively. VAT showed moderate correlation with liver PDFF (r = 0.41, p < 0.001) and weak correlation with Lvol (r = 0.38, p < 0.001), Svol (r = 0.20, p = 0.025) and pancreas PDFF (rs = 0.29, p = 0.001). VAT, Lvol and liver PDFF were significantly higher in patients with HS (p < 0.001), NASH (p < 0.05) and HF (p < 0.05). VAT was also significantly higher in the presence of lobular inflammation (p = 0.019) and hepatocyte ballooning (p = 0.001). The cut-off VAT volumes for predicting HS, NASH and HF were 101.8 mL (AUC, 0.7), 111.8 mL (AUC, 0.64) and 111.6 mL (AUC, 0.66), respectively. (4) Conclusion: The MRI determined VAT can be used for predicting the presence of HS, NASH and HF in patients with known or suspected NAFLD.
Collapse
Affiliation(s)
- Ilkay S. Idilman
- Division of Abdominal Imaging, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (I.S.I.); (H.M.L.); (T.G.); (K.P.); (J.L.); (M.Y.)
- Department of Radiology, School of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Hsien Min Low
- Division of Abdominal Imaging, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (I.S.I.); (H.M.L.); (T.G.); (K.P.); (J.L.); (M.Y.)
- Department of Radiology, Tan Tock Seng Hospital, Singapore 308433, Singapore
| | - Tolga Gidener
- Division of Abdominal Imaging, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (I.S.I.); (H.M.L.); (T.G.); (K.P.); (J.L.); (M.Y.)
| | - Kenneth Philbrick
- Division of Abdominal Imaging, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (I.S.I.); (H.M.L.); (T.G.); (K.P.); (J.L.); (M.Y.)
| | - Taofic Mounajjed
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Jiahui Li
- Division of Abdominal Imaging, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (I.S.I.); (H.M.L.); (T.G.); (K.P.); (J.L.); (M.Y.)
| | - Alina M. Allen
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Meng Yin
- Division of Abdominal Imaging, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (I.S.I.); (H.M.L.); (T.G.); (K.P.); (J.L.); (M.Y.)
| | - Sudhakar K. Venkatesh
- Division of Abdominal Imaging, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (I.S.I.); (H.M.L.); (T.G.); (K.P.); (J.L.); (M.Y.)
- Correspondence:
| |
Collapse
|
24
|
Pietrobon CB, Lisboa PC, Bertasso IM, Peixoto TC, Soares PN, de Oliveira E, Rabelo K, de Carvalho JJ, Manhães AC, de Moura EG. Pancreatic steatosis in adult rats induced by nicotine exposure during breastfeeding. Endocrine 2021; 72:104-115. [PMID: 33420949 DOI: 10.1007/s12020-020-02579-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Maternal nicotine exposure negatively impacts offspring's health and metabolism, leading to obesity and insulin resistance. Here we investigated the pancreatic islet function, glycemic homeostasis, and insulin signaling in adult rat offspring that were nicotine-exposed during breastfeeding. METHODS For this, lactating Wistar rat dams were divided into two groups: Nicotine (implanted with osmotic minipumps containing 6 mg/Kg, NIC) and Control (saline, CON). Solutions were released from postnatal (PN) day 2-16. At PN110 and PN170, 10 offspring per litter/sex/group were submitted to the oral glucose tolerance test (OGTT). PN180 offspring were killed and glycemia, insulinemia, adiponectinemia, pancreas morphology as well as pancreatic islet protein expression (related to insulin secretion) and skeletal muscle (related to insulin action) were evaluated. Males and females were compared to their respective controls. RESULTS Adult NIC offspring of both sexes showed glucose intolerance in the OGTT. Despite normoglycemia, NIC males showed hyperinsulinemia while females, although normoinsulinemic, had hyperglycemia. Both sexes showed increased IRI, reduced adiponectin/visceral fat mass ratio and higher ectopic deposition of lipids in the pancreatic tissue adipocytes. In pancreatic islets, NIC males showed lower PDX-1 expression while females had higher PDX-1 and GLUT2 expressions plus lower α2 adrenergic receptor. In the muscle, NIC offspring of both sexes showed reduction of GLUT4 expression; NIC males also had lower insulin receptor and pAKT expressions. CONCLUSIONS Thus, glycemic homeostasis and peripheral insulin signaling in adult offspring of both sexes are affected by nicotine exposure through the milk, increasing the risk for type 2 diabetes development.
Collapse
Affiliation(s)
- Carla Bruna Pietrobon
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Cristina Lisboa
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Iala Milene Bertasso
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thamara Cherem Peixoto
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Novaes Soares
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elaine de Oliveira
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kíssila Rabelo
- Laboratory of Ultrastructure and Tissue Biology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge José de Carvalho
- Laboratory of Ultrastructure and Tissue Biology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alex Christian Manhães
- Neurophysiology Laboratory, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
25
|
Lee EH, Kim JY, Yang HR. Relationship Between Histological Features of Non-alcoholic Fatty Liver Disease and Ectopic Fat on Magnetic Resonance Imaging in Children and Adolescents. Front Pediatr 2021; 9:685795. [PMID: 34178902 PMCID: PMC8222518 DOI: 10.3389/fped.2021.685795] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/14/2021] [Indexed: 12/25/2022] Open
Abstract
Objectives: To investigate the association between ectopic fat content in the liver and pancreas, obesity-related metabolic components, and histological findings of non-alcoholic fatty liver disease (NAFLD) in children. Methods: This cross-sectional study investigated 63 children with biopsy-proven NAFLD who underwent magnetic resonance imaging (MRI), anthropometry, laboratory tests, and body composition analysis. Clinical and metabolic parameters, MRI-measured hepatic fat fraction (HFF) and pancreatic fat fraction (PFF), and histological findings were analyzed. Results: In a total of 63 children (48 boys, median age 12.6 years, median body mass index z-score 2.54), HFF was associated with histological steatosis [10.4, 23.7, and 31.1% in each steatosis grade, P < 0.001; Spearman's rho coefficient (rs) = 0.676; P < 0.001] and NAFLD activity score (rs = 0.470, P < 0.001), but not with lobular inflammation, hepatocyte ballooning, and hepatic fibrosis. PFF was not associated with any histological features of the liver. Waist circumference-to-height ratio and body fat percentage were associated with the steatosis grade (P = 0.006 and P = 0.004, respectively). Alanine aminotransferase was not associated with steatosis but was associated with lobular inflammation (P = 0.008). Lobular inflammation was also associated with high total cholesterol and low-density lipoprotein cholesterol and metabolic syndrome (P = 0.015, P = 0.036, and P = 0.038, respectively). Conclusions: Hepatic steatosis on MRI was only associated with the histological steatosis grade, while elevated serum levels of liver enzymes and lipids were related to the severity of lobular inflammation. Therefore, MRI should be interpreted in conjunction with the anthropometric and laboratory findings in pediatric patients.
Collapse
Affiliation(s)
- Eun Hye Lee
- Department of Pediatrics, Nowon Eulji Medical Center, Eulji University, Daejeon, South Korea
| | - Ji Young Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hye Ran Yang
- Department of Pediatrics, Seoul National University Bundang Hospital, Seongnam, South Korea.,College of Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
26
|
A Universal Gut-Microbiome-Derived Signature Predicts Cirrhosis. Cell Metab 2020; 32:878-888.e6. [PMID: 32610095 PMCID: PMC7822714 DOI: 10.1016/j.cmet.2020.06.005] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/20/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022]
Abstract
Dysregulation of the gut microbiome has been implicated in the progression of non-alcoholic fatty liver disease (NAFLD) to advanced fibrosis and cirrhosis. To determine the diagnostic capacity of this association, we compared stool microbiomes across 163 well-characterized participants encompassing non-NAFLD controls, NAFLD-cirrhosis patients, and their first-degree relatives. Interrogation of shotgun metagenomic and untargeted metabolomic profiles by using the random forest machine learning algorithm and differential abundance analysis identified discrete metagenomic and metabolomic signatures that were similarly effective in detecting cirrhosis (diagnostic accuracy 0.91, area under curve [AUC]). Combining the metagenomic signature with age and serum albumin levels accurately distinguished cirrhosis in etiologically and genetically distinct cohorts from geographically separated regions. Additional inclusion of serum aspartate aminotransferase levels, which are increased in cirrhosis patients, enabled discrimination of cirrhosis from earlier stages of fibrosis. These findings demonstrate that a core set of gut microbiome species might offer universal utility as a non-invasive diagnostic test for cirrhosis.
Collapse
|
27
|
Kuchay MS, Krishan S, Mishra SK, Choudhary NS, Singh MK, Wasir JS, Kaur P, Gill HK, Bano T, Farooqui KJ, Mithal A. Effect of dulaglutide on liver fat in patients with type 2 diabetes and NAFLD: randomised controlled trial (D-LIFT trial). Diabetologia 2020; 63:2434-2445. [PMID: 32865597 DOI: 10.1007/s00125-020-05265-7] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 07/22/2020] [Indexed: 02/08/2023]
Abstract
AIMS/HYPOTHESIS Liraglutide, a daily injectable glucagon-like peptide-1 receptor (GLP-1r) agonist, has been shown to reduce liver fat content (LFC) in humans. Data regarding the effect of dulaglutide, a once-weekly GLP-1r agonist, on human LFC are scarce. This study examined the effect of dulaglutide on LFC in individuals with type 2 diabetes and non-alcoholic fatty liver disease (NAFLD). METHODS Effect of dulaglutide on liver fat (D-LIFT) was a 24 week, open-label, parallel-group, randomised controlled trial to determine the effect of dulaglutide on liver fat at a tertiary care centre in India. Adults (n = 64), who had type 2 diabetes and MRI-derived proton density fat fraction-assessed LFC of ≥6.0% at baseline, were randomly assigned to receive dulaglutide weekly for 24 weeks (add-on to usual care) or usual care, based on a predefined computer-generated number with a 1:1 allocation that was concealed using serially numbered, opaque, sealed envelopes. The primary endpoint was the difference of the change in LFC from 0 (baseline) to 24 weeks between groups. The secondary outcome measures included the difference of the change in pancreatic fat content (PFC), change in liver stiffness measurement (LSM in kPa) measured by vibration-controlled transient elastography, and change in liver enzymes. RESULTS Eighty-eight patients were screened; 32 were randomly assigned to the dulaglutide group and 32 to the control group. Overall, 52 participants were included for per-protocol analysis: those who had MRI-PDFF data at baseline and week 24. Dulaglutide treatment resulted in a control-corrected absolute change in LFC of -3.5% (95% CI -6.6, -0.4; p = 0.025) and relative change of -26.4% (-44.2, -8.6; p = 0.004), corresponding to a 2.6-fold greater reduction. Dulaglutide-treated participants also showed a significant reduction in γ-glutamyl transpeptidase (GGT) levels (mean between-group difference -13.1 U/l [95% CI -24.4, -1.8]; p = 0.025) and non-significant reductions in aspartate aminotransferase (AST) (-9.3 U/l [-19.5, 1.0]; p = 0.075) and alanine aminotransferase (ALT) levels (-13.1 U/l [-24.4, 2.5]; p = 0.10). Absolute changes in PFC (-1.4% [-3.2, 0.3]; p = 0.106) and LSM (-1.31 kPa [-2.99, 0.37]; p = 0.123) were not significant when comparing the two groups. There were no serious drug-related adverse events. CONCLUSIONS/INTERPRETATION When included in the standard treatment for type 2 diabetes, dulaglutide significantly reduces LFC and improves GGT levels in participants with NAFLD. There were non-significant reductions in PFC, liver stiffness, serum AST and serum ALT levels. Dulaglutide could be considered for the early treatment of NAFLD in patients with type 2 diabetes. TRIAL REGISTRATION ClinicalTrials.gov NCT03590626 FUNDING: The current study was supported by an investigator-initiated study grant from Medanta-The Medicity's departmental research fund and a grant from the Endocrine and Diabetes Foundation (EDF), India. Graphical abstract.
Collapse
Affiliation(s)
- Mohammad S Kuchay
- Division of Endocrinology and Diabetes, Medanta-The Medicity Hospital, Haryana, India.
| | - Sonal Krishan
- Department of Radiology, Medanta-The Medicity Hospital, Haryana, India
| | - Sunil K Mishra
- Division of Endocrinology and Diabetes, Medanta-The Medicity Hospital, Haryana, India
| | - Narendra S Choudhary
- Institute of Digestive and Hepatobiliary Sciences, Medanta-The Medicity Hospital, Haryana, India
| | - Manish K Singh
- Department of Clinical Research and Studies, Medanta-The Medicity Hospital, Haryana, India
| | - Jasjeet S Wasir
- Division of Endocrinology and Diabetes, Medanta-The Medicity Hospital, Haryana, India
| | - Parjeet Kaur
- Division of Endocrinology and Diabetes, Medanta-The Medicity Hospital, Haryana, India
| | - Harmandeep K Gill
- Division of Endocrinology and Diabetes, Medanta-The Medicity Hospital, Haryana, India
| | - Tarannum Bano
- Division of Endocrinology and Diabetes, Medanta-The Medicity Hospital, Haryana, India
| | - Khalid J Farooqui
- Division of Endocrinology and Diabetes, Medanta-The Medicity Hospital, Haryana, India
| | - Ambrish Mithal
- Division of Endocrinology and Diabetes, Medanta-The Medicity Hospital, Haryana, India
| |
Collapse
|
28
|
Crabtree CD, LaFountain RA, Hyde PN, Chen C, Pan Y, Lamba N, Sapper TN, Short JA, Kackley ML, Buga A, Miller VJ, Scandling D, Andersson I, Barker S, Hu HH, Volek JS, Simonetti OP. Quantification of Human Central Adipose Tissue Depots: An Anatomically Matched Comparison Between DXA and MRI. ACTA ACUST UNITED AC 2020; 5:358-366. [PMID: 31893234 PMCID: PMC6935994 DOI: 10.18383/j.tom.2019.00018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Excess visceral adipose tissue (VAT) and VAT volume relative to subcutaneous adipose tissue (SAT) are associated with elevated health risks. This study compares fat measurements by dual-energy X-ray absorptiometry (DXA) and magnetic resonance imaging (MRI). In total, 21 control subjects (Control) and 16 individuals with metabolic syndrome (MetSyn) were scanned by DXA and MRI. The region measured by MRI was matched to the android region defined by DXA, and MRI reproducibility was also evaluated. In addition, liver fat fraction was quantified via MRI and whole-body fat by DXA. VAT measurements are interchangeable between DXA and MRI in the Control (R = 0.946), MetSyn (R = 0.968), and combined cohort (R = 0.983). VAT/SAT ratio did not differ in the Control group (P = .10), but VAT/SAT ratio measured by DXA was significantly higher in the MetSyn group (P < .01) and the combined (P = .03) cohort. Intraobserver (ICC = 0.998) and interobserver (ICC = 0.977) reproducibility of MRI VAT measurements was excellent. Liver fat fraction by MRI was higher (P = .001) in MetSyn (12.4% ± 7.6%) than in controls (2.6% ± 2.2%), as was whole-body fat percentage by DXA (P = .001) between the MetSyn (42.0% ± 8.1%) and Control groups (26.7% ± 6.9%). DXA and MRI VAT are interchangeable when measured over an anatomically matched region of the abdomen, while SAT and VAT/SAT ratio differ between the 2 modalities.
Collapse
Affiliation(s)
| | | | | | | | - Yue Pan
- Dorothy M. Davis Heart & Lung Research Institute, and
| | | | | | | | | | | | | | | | - Irma Andersson
- Department of Radiology, The Ohio State University, Columbus, OH
| | - Samantha Barker
- Department of Radiology, The Ohio State University, Columbus, OH
| | - Houchun H Hu
- Department of Radiology, Nationwide Children's Hospital, Columbus, OH; and
| | | | - Orlando P Simonetti
- Dorothy M. Davis Heart & Lung Research Institute, and.,Department of Radiology, The Ohio State University, Columbus, OH.,Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH
| |
Collapse
|
29
|
Use of Biomarkers and Imaging for Early Detection of Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12071965. [PMID: 32707720 PMCID: PMC7409286 DOI: 10.3390/cancers12071965] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer remains one of the deadliest cancers worldwide, and it is typically diagnosed late, with a poor prognosis. Early detection is the most important underlying factor for improving the prognosis of pancreatic cancer patients. One of the most effective strategies for detecting cancers at an early stage is screening of the general population. However, because of the low incidence of pancreatic cancer in the general population, the stratification of subjects who need to undergo further examinations by invasive and expensive modalities is important. Therefore, minimally invasive modalities involving biomarkers and imaging techniques that would facilitate the early detection of pancreatic cancer are highly needed. Multiple types of new blood biomarkers have recently been developed, including unique post-translational modifications of circulating proteins, circulating exosomes, microRNAs, and circulating tumor DNA. We previously reported that circulating apolipoprotein A2 undergoes unique processing in the bloodstream of patients with pancreatic cancer and its precancerous lesions. Additionally, we recently demonstrated a new method for measuring pancreatic proton density in the fat fraction using a fat–water magnetic resonance imaging technique that reflects pancreatic steatosis. In this review, we describe recent developments in potential biomarkers and imaging modalities for the early detection and risk stratification of pancreatic cancer, and we discuss current strategies for implementing screening programs for pancreatic cancer.
Collapse
|
30
|
Noninvasive assessment of abdominal adipose tissues and quantification of hepatic and pancreatic fat fractions in type 2 diabetes mellitus. Magn Reson Imaging 2020; 72:95-102. [PMID: 32668273 DOI: 10.1016/j.mri.2020.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/01/2020] [Accepted: 07/01/2020] [Indexed: 11/23/2022]
Abstract
The purpose of this study was to evaluate adipose tissue distributions and hepatic and pancreatic fat contents using a 6-point Dixon MRI technique in type 2 diabetes mellitus (T2DM), and to assess associations between fat distributions and biochemical markers of insulin resistance. Intra-abdominal MRI was investigated in 14 T2DM patients, 13 age- and sex-matched healthy controls (HC) and 11 young HC using a 3 T Prisma MRI scanner. All T2DM subjects completed a fasting comprehensive metabolic panel, and demographic measurements were taken according to standardized methodologies. We observed excellent correlation (R2 = 0.94) between hepatic fat fraction quantified using 6-point Dixon MRI and gold standard MRS, establishing the accuracy and reliability of the Dixon technique. Significantly increased visceral adipose tissue (VAT) volumes were found in T2DM patients compared to age-matched HC (1569.81 ± 670.62 cm3 vs. 1106.60 ± 566.85 cm3, p = .04). We also observed a trend of increasing subcutaneous adipose tissues (SAT), and total abdominal fat (TAT) volumes in T2DM compared to age-matched HC. Hepatic fat fraction percentage (HFF%) was 44.6% higher in T2DM compared to age-matched HC and 64.4% higher compared to young HC. Pancreatic fat fractions in the head and body/tail were higher in T2DM patients compared to both healthy cohorts. We also observed correlations between fat contents of the liver and pancreas in T2DM patients, and association between biochemical markers of T2DM with HFF, indicating a risk for non-alcoholic fatty liver disease among T2DM. In summary, this study provides evidence of T2DM patients having increased liver and pancreatic fat, as well as increased adipose tissues.
Collapse
|
31
|
Liver and pancreas: 'Castor and Pollux' regarding the relationship between hepatic steatosis and pancreas exocrine insufficiency. Pancreatology 2020; 20:880-886. [PMID: 32475757 DOI: 10.1016/j.pan.2020.04.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 04/07/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic exocrine insufficiency (PEI) is found in 30-50% of diabetes mellitus (DM). Insulin resistance is triggering factor in both DM and nonalcoholic fatty liver disease (NAFLD). Therefore, we aimed to investigate frequency of PEI in NAFLD, and relationship of fecal pancreatic elastase (PE) levels with liver histology and pancreatic fat. METHODS Ninety-seven biopsy proven NAFLD patients and 50 controls were enrolled. Pancreas exocrine functions were measured by PE. Magnetic resonance imaging-estimated proton density fat fraction (MRI-PDFF) was used to quantify fat. RESULTS NAFLD patients had significantly lower PE levels than controls (297 [204-517] vs. 500 [298-678] μg/g, p < 0.01). PEI (PE < 200 μg/g) ratio of NAFLD patients (22.7%, n = 22) was higher than PEI ratio of controls (6%, n = 3) (p = 0.011). Among diabetic (n = 35) NAFLD patients, 9 (25.7%) exhibited PEI, compared to 13 (21%) of non-diabetics. There was no significant difference in patients with and without DM in terms of PEI (p = 0.592). Among NASH (n = 68) patients 16 (23.5%) exhibited PEI, compared to (20.7%) of non-NASH (p = 0.76). Multiple analysis revealed NAFLD as a predictor of PEI independent of age, sex and DM (OR = 4.892, p = 0,021). Mean pancreas MRI-PDFF was significantly higher in diabetics (13.7% ± 3.6% vs. 8.7% ± 5.1%, p = 0.001). There was no significant pancreas MRI-PDFF difference between NASH and non-NASH (P = 0.95). Mean pancreas MRI-PDFF was significantly higher in patients with PEI (13.7% ± 3.4% vs. 8.9% ± 5.2%, P < 0.01). CONCLUSION This is the first study demonstrating the high frequency of PEI in NAFLD independent of DM. Moreover, increasing pancreatic steatosis appears to be associated with higher frequency of PEI in NAFLD.
Collapse
|
32
|
Paul J, Shihaz AVH. PANCREATIC STEATOSIS: A NEW DIAGNOSIS AND THERAPEUTIC CHALLENGE IN GASTROENTEROLOGY. ARQUIVOS DE GASTROENTEROLOGIA 2020; 57:216-220. [PMID: 32490903 DOI: 10.1590/s0004-2803.202000000-27] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/20/2020] [Indexed: 02/08/2023]
Abstract
Fat infiltration in the pancreas is called pancreatic steatosis and it has several synonyms such as pancreatic lipomatosis, non-alcoholic fatty pancreatic disease, lipomatous pseudohypertrophy, fatty replacement, fatty pancreas and fatty infiltration. Pancreatic steatosis describes a disease ranging from infiltration of fat in the pancreas to pancreatic inflammation, and development of pancreatic fibrosis. There are multiple aetiologies of this condition, such as metabolic syndrome, alcohol intake, viral infections, toxins, congenital syndromes, etc. Pancreatic steatosis is usually diagnosed by trans-abdominal ultrasound, computed tomography scan and magnetic resonance imaging. Fatty infiltration in pancreas may lead to pancreatitis, diabetes mellitus and may be a predisposing cause of pancreatic cancer. Now a day, pancreatic steatosis is a common incidental finding during abdominal ultrasonography for other reasons and is a new challenge in Gastroenterology. But there is no guideline for pancreatic steatosis till now. In this review article, we are trying to give an overall idea (aetiologies, diagnosis, management, clinical significances) on pancreatic steatosis.
Collapse
Affiliation(s)
- Jayanta Paul
- Desun Hospital and Heart Institute, Department of Gastroenterology, Kolkata, India
| | | |
Collapse
|
33
|
Vieira J, Amorim J, Martí-Bonmatí L, Alberich-Bayarri Á, França M. Quantifying steatosis in the liver and pancreas with MRI in patient with chronic liver disease. RADIOLOGIA 2020. [DOI: 10.1016/j.rxeng.2019.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Tremmel DM, Feeney AK, Mitchell SA, Chlebeck PJ, Raglin S, Fernandez LA, Odorico JS, Sackett SD. Hypertension, but not body mass index, is predictive of increased pancreatic lipid content and islet dysfunction. Am J Transplant 2020; 20:1105-1115. [PMID: 31715064 PMCID: PMC7103563 DOI: 10.1111/ajt.15698] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/15/2019] [Accepted: 11/04/2019] [Indexed: 01/25/2023]
Abstract
Pancreatic steatosis is thought to be a negative risk factor for pancreas transplant outcomes. Despite considering donor body mass index (BMI) and the visualization of intercalated fat as indicators of donor pancreas lipid content, transplant surgeons do not use a quantitative method to directly measure steatosis when deciding to transplant a pancreas. In this study, we used nondiabetic human pancreata donated for research to measure the pancreatic and islet-specific lipid content to determine which clinical markers correlate best with lipid content. Interestingly, we found that BMI and age correlate with increased pancreatic lipid content (Panc-LC) in men, but not women. Our findings further suggest that total Panc-LC correlates with an increase in islet lipid content for both men and women. We noted that pancreata donated from individuals with a history of hypertension have increased Panc-LC independent of donor BMI or sex. Moreover, we identify hypertension as a risk factor for reduced islet function after islet isolation. Together, our findings emphasize differences in pancreas graft quality related to pancreatic and islet lipid content, which may not be predicted by assessing BMI alone but may be influenced by a donor history of hypertension.
Collapse
Affiliation(s)
- Daniel M. Tremmel
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA.,Co-first authors
| | - Austin K. Feeney
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA.,Co-first authors
| | - Samantha A. Mitchell
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Peter J. Chlebeck
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Sierra Raglin
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Luis A. Fernandez
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Jon S. Odorico
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| | - Sara D. Sackett
- Division of Transplantation, Department of Surgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, 53705, USA
| |
Collapse
|
35
|
Ballester-Vallés C, Flores-Méndez J, Delgado-Moraleda J, Ballesteros Martín-Portugués A, Merino-Torres J, Fornés-Ferrer V, Martí-Bonmatí L. Hepatic and pancreatic fat as imaging biomarkers of metabolic syndrome. RADIOLOGIA 2020. [DOI: 10.1016/j.rxeng.2019.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Koç U, Ocakoğlu G, Alğin O. The efficacy of the 3-dimensional vibe-caipirinha-dixon technique in the evaluation of pancreatic steatosis. Turk J Med Sci 2020; 50:184-194. [PMID: 31865664 PMCID: PMC7080364 DOI: 10.3906/sag-1909-83] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022] Open
Abstract
Background/aim CAIPIRINHA is a new technique in abdominal imaging. Pancreatic steatosis (PS) is a subject of increasing scientific interest. The aim of this study was to investigate the efficacy of the isotropic 3D-VIBE- CAIPIRINHA -DIXON technique on a new generation 3-tesla MR unit in the evaluation of PS. Materials and methods In this retrospective study, the imaging findings of 49 patients with PS and 41 control subjects were examined. The pancreas-to-spleen ratio (PSR), pancreas-to-muscle ratio (PMR), and pancreatic signal intensity index (PSII) were defined as 3 new parameters and these indexes were calculated from the in-phase/out of phase 3D-VIBE- CAIPIRINHA-DIXON images. Results The PSR, PMR, and PSII values were significantly different between the patient and control groups (P = 0.001, P = 0.009, P < 0.001, respectively). Statistically significant differences were observed between patient and control groups for ROI measurements of fatty areas on these sequences/images: subtraction (in-out) (P < 0.001), T2W HASTE (P < 0.001), DIXON-fat (P < 0.001), fat-suppressed T1W (P = 0.002), and subtraction (out-in) (P = 0.010). Conclusion Evaluation of PS with the 3D-VIBE-CAIPIRINHA-DIXON technique can be made rapidly and effectively.
Collapse
Affiliation(s)
- Ural Koç
- Section of Radiology, Ankara Sehit Ahmet Ozsoy State Hospital, Ankara, Turkey
| | - Gökhan Ocakoğlu
- Department of Biostatistics, School of Medicine, Uludag University, Bursa, Turkey
| | - Oktay Alğin
- Department of Radiology, School of Medicine, Yıldırım Beyazıt University, Ankara, Turkey
| |
Collapse
|
37
|
Vieira J, Amorim J, Martí-Bonmatí L, Alberich-Bayarri Á, França M. Quantifying steatosis in the liver and pancreas with MRI in patient with chronic liver disease. RADIOLOGIA 2020; 62:222-228. [PMID: 31932016 DOI: 10.1016/j.rx.2019.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/08/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022]
Abstract
AIM To compare pancreatic and hepatic steatosis quantified by proton density fat fraction (PDFF) on magnetic resonance imaging (MRI) in patients with chronic liver disease. MATERIAL AND METHODS This cross-sectional study included 46 adult patients who underwent liver biopsy for chronic viral hepatitis (n=19) or other chronic non-alcoholic liver diseases (NALD) (n=27). Liver biopsy was used as the gold standard for diagnosing and grading hepatic steatosis. All patients underwent clinical evaluation and MRI with a multi-echo chemical shift-encoded (MECSE) gradient-echo sequence for liver and pancreas PDFF quantification. We used Spearman's correlation coefficient to determine the degree of association between hepatic PDFF and steatosis grade, and between pancreatic PDFF and steatosis grade and hepatic PDFF. To compare the chronic viral hepatitis group and the NALD group, we used t-tests for continuous or ordinal variables and chi-square tests for categorical variables. RESULTS Hepatic PDFF measurements correlated with steatosis grades (RS=0.875, p<0.001). Pancreatic PDFF correlated with hepatic steatosis grades (RS=0.573, p<0.001) and hepatic PDFF measurements (RS=0.536, p<0.001). In the subgroup of patients with chronic NALD, the correlations remained significant between pancreatic PDFF and hepatic PDFF (RS=0.632, p<0.001) and between pancreatic PDFF and liver steatosis (RS=0.608, p<0.001); however, in the subgroup of patients with viral hepatitis these correlations were no longer significant. CONCLUSION Pancreatic fat deposition correlates with hepatic steatosis in patients with chronic NALD, but not in those with chronic viral hepatitis.
Collapse
Affiliation(s)
- J Vieira
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidad de Oporto, Oporto, Portugal.
| | - J Amorim
- Departamento de Diagnóstico por la Imagen, Centro Hospitalar do Porto, Oporto, Portugal; Escola de Medicina, Universidade do Minho, Braga, Portugal; ICVS/3B's, Instituto de Investigación de Ciencias de la Vida y la Salud, Universidade do Minho, Braga, Portugal
| | - L Martí-Bonmatí
- Departamento de Radiología y Grupo de Investigación Biomédica en Imagen GIBI2(30). Hospital Universitario y Politécnico La Fe e Instituto de Investigación Sanitaria La Fe, Valencia, España
| | - Á Alberich-Bayarri
- Departamento de Radiología y Grupo de Investigación Biomédica en Imagen GIBI2(30). Hospital Universitario y Politécnico La Fe e Instituto de Investigación Sanitaria La Fe, Valencia, España; Quantitative Imaging Biomarkers in Medicine (QUIBIM), Valencia, España
| | - M França
- Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidad de Oporto, Oporto, Portugal; Departamento de Diagnóstico por la Imagen, Centro Hospitalar do Porto, Oporto, Portugal; i3S, Instituto de Investigacão e Inovação em Saúde, IBMC, Instituto de Biología Molecular y Celular, Oporto, Portugal
| |
Collapse
|
38
|
Virostko J. Quantitative Magnetic Resonance Imaging of the Pancreas of Individuals With Diabetes. Front Endocrinol (Lausanne) 2020; 11:592349. [PMID: 33343509 PMCID: PMC7747766 DOI: 10.3389/fendo.2020.592349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
Magnetic resonance imaging (MRI) has the potential to improve our understanding of diabetes and improve both diagnosis and monitoring of the disease. Although the spatial resolution of MRI is insufficient to directly image the endocrine pancreas in people, the increasing awareness that the exocrine pancreas is also involved in diabetes pathogenesis has spurred new MRI applications. These techniques build upon studies of exocrine pancreatic diseases, for which MRI has already developed into a routine clinical tool for diagnosis and monitoring of pancreatic cancer and pancreatitis. By adjusting the imaging contrast and carefully controlling image acquisition and processing, MRI can quantify a variety of tissue pathologies. This review introduces a number of quantitative MRI techniques that have been applied to study the diabetic pancreas, summarizes progress in validating and standardizing each technique, and discusses the need for image analyses that account for spatial heterogeneity in the pancreas.
Collapse
Affiliation(s)
- John Virostko
- Department of Diagnostic Medicine, University of Texas at Austin, Austin, TX, United States
- Livestrong Cancer Institutes, University of Texas at Austin, Austin, TX, United States
- Department of Oncology, University of Texas at Austin, Austin, TX, United States
- *Correspondence: John Virostko,
| |
Collapse
|
39
|
Wang YS, Ye J, Cao YH, Zhang R, Han XF, Zou LL, Kuang L, Zhang J, Lian H, Xia JX, Zhang Q, Dai W. Association of [ 1H]-MRS quantified liver fat content with glucose metabolism status. Diabetol Metab Syndr 2020; 12:51. [PMID: 32528557 PMCID: PMC7282165 DOI: 10.1186/s13098-020-00558-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/30/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Previous literatures have implied that the liver fat deposition plays a crucial role in the development and progression of insulin resistance. In the present study, we aimed to investigate the association of liver fat content (LFC) with glucose metabolism status in the population of newly diagnosed type 2 diabetes mellitus (nT2DM), prediabetes mellitus (PDM) and normal controls (NC), and assessing if the LFC could as an indicator for the prediction of T2DM. METHODS A total of 242 subjects (including 141 nT2DM patients, 48 PDM subjects and 53 NC) were enrolled. The levels of LFC were quantified by using the proton magnetic resonance spectroscopy ([1H]-MRS) technique. Clinical and laboratory parameters of study subjects were collected by medical records and biochemical detection. One-way ANOVA or nonparametric test (Kruskal-Wallis) was applied for intergroup comparisons; intergroup comparison was performed in using of Bonferroni multiple-significance-test correction. RESULTS There were significantly increased LFC levels in nT2DM (14.72% ± 6.37%) than in PDM (9.62% ± 4.41%) and that of NC groups (5.11% ± 3.66%) (all p < 0.001). The prevalence of nonalcoholic fatty liver disease (NAFLD) was also found to be increased in nT2DM (91.48%) than in PDM (85.41%) and that of NC (32.07%) groups. Correlation analysis revealed that the increase of LFC positively associated with fast plasma glucose (FPG), 2 h plasma glucose (PG), Delta G30 and homeostatic model assessment of insulin resistance (HOMA-IR), negatively associated with Delta Ins30, Delta C30, Ins30/G30 AUC, CP30/G30 AUC, Ins AUC/G AUC, CP AUC/G AUC, homeostatic model assessment for β-cell function index (HOMA-β) and matsuda insulin sensitivity index (Matsuda ISI). Multilinear regression analysis showed that LFC, body mass index (BMI) and diastolic blood pressure (DBP) contributed for the prediction of HOMA-IR, and total cholesterol (TC), age, waist circumference (WC) and low-density lipoprotein cholesterol (LDL-C) were the significant contributors for HOMA-β. CONCLUSIONS Our study revealed an increased LFC level and prevalence of NAFLD in nT2DM than in PDM and that of NC groups, the increase of LFC was closely associated with insulin resistance and impaired glucose metabolism status, may be regarded as potential indicator contributing to the development and progression of T2DM.
Collapse
Affiliation(s)
- Yun-Sheng Wang
- Department of Endocrinology, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Jun Ye
- Department of Endocrinology, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Yong-Hong Cao
- Department of Endocrinology, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Rong Zhang
- Department of Endocrinology, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Xiao-Fang Han
- Department of Endocrinology, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Ling-Ling Zou
- Department of Endocrinology, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Lei Kuang
- Department of Endocrinology, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Ji Zhang
- Department of Magnetic Resonance Imaging, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Hu Lian
- Department of Magnetic Resonance Imaging, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Jin-Xiang Xia
- Department of Magnetic Resonance Imaging, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| | - Qiu Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui China
| | - Wu Dai
- Department of Endocrinology, The Second People’s Hospital of Hefei, The Affiliated Hefei Hospital of Anhui Medical University, Hefei, 230011 Anhui China
| |
Collapse
|
40
|
Aliyari Ghasabeh M, Shaghaghi M, Khoshpouri P, pan L, Pandy A, Pandy P, Zhong X, Kannengiesser S, Kamel IR. Correlation between incidental fat deposition in the liver and pancreas in asymptomatic individuals. Abdom Radiol (NY) 2020; 45:203-210. [PMID: 31482380 DOI: 10.1007/s00261-019-02206-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE To explore the utility of two different fat quantification methods in the liver and pancreas and to test the accuracy of multi-echo Dixon as a single sequence in detecting early stage of fat deposition. METHODS 58 healthy potential liver donors underwent abdominal 3T MRI, prospectively. Single-voxel MR Spectroscopy (MRS), dual-echo Dixon, and multi-echo Dixon were performed. Two independent readers obtained proton density fat fraction (PDFF) of the liver and pancreas by placing ROIs on the 2 Dixon sequences. Correlation between the two PDFF measurements was assessed in the liver and pancreas. Values in the liver were also compared to those obtained by MRS. RESULTS PDFF in the liver was 6.3 ± 4.2%, 5.5 ± 3.9%, and 5.1 ± 4.1% by MRS, dual-echo Dixon, and multi-echo Dixon, respectively. Dual-echo Dixon and multi-echo Dixon showed good correlation in PDFF quantification of the liver (r = 0.82, p < 0.0005). Multi-echo Dixon showed a good correlation (r = 0.72, p = 0.0005) between the fat measured in the liver and in the pancreas. To differentiate between normal (PDFF ≤ 6%) and mild fat deposition (PDFF: 6-33%) in the liver, analysis showed sensitivity, specificity, and accuracy of 74%, 81%, and 80% for dual-echo Dixon and 85%, 96%, and 89% for multi-echo Dixon, respectively. Mean PDFF in the pancreas was 7.2 ± 2.8% and 6.7 ± 3.3%, by dual-echo and multi-echo Dixon, respectively. Dual-echo Dixon and multi-echo Dixon showed good correlation in PDFF quantification of the pancreas (r = 0.58, p < 0.0005). CONCLUSION Multi-echo Dixon in liver has high accuracy in distinguishing between subjects with normal liver fat and those with mildly elevated liver fat. Multi-echo Dixon can be used to screen for early fat deposition in the liver and pancreas.
Collapse
|
41
|
Three-dimensional analysis of pancreatic fat by fat-water magnetic resonance imaging provides detailed characterization of pancreatic steatosis with improved reproducibility. PLoS One 2019; 14:e0224921. [PMID: 31790429 PMCID: PMC6886808 DOI: 10.1371/journal.pone.0224921] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Since pancreatic steatosis is reported as a possible risk factor for pancreatic cancer, the development of a non-invasive method to quantify pancreatic steatosis is needed. Proton density fat fraction (PDFF) measurement is a magnetic resonance imaging (MRI) based method for quantitatively assessing the steatosis of a region of interest (ROI). Although it is commonly used for quantification of hepatic steatosis, pancreatic PDFF can greatly vary depending on the ROI's location because of the patchy nature of pancreatic fat accumulation. In this study, we attempted to quantify pancreatic steatosis by fat-water MRI with improved reproducibility. METHODS Using the MRI images of 159 patients with nonalcoholic fatty liver disease, we attempted to calculate the average PDFF of whole pancreas. We set ROIs covering the entire area of the pancreas appearing in every slice and calculated the average PDFF from all the voxels included in the pancreas. We named this average value as whole-pancreatic PDFF and evaluated the reproducibility of the measured values. In addition to whole-pancreatic PDFF, we measured the average PDFF of the pancreatic head (head-PDFF) and that of the pancreatic body plus tail separately and analyzed their correlation with the clinical characteristics of the patients. RESULTS The mean inter-examiner coefficient of variation of the whole-pancreatic PDFF was 11.39%. The whole-pancreatic PDFF was correlated with age (p = 0.039), body mass index (p = 0.0093) and presence/absence of diabetes (p = 0.0055). The serum level of low-density lipoprotein cholesterol was inversely correlated with the head-PDFF. CONCLUSION We developed a new measurement method of the pancreatic PDFF with greater reproducibility. Using this method, we characterized pancreatic steatosis in detail. This novel measurement method allows accurate estimation of the severity of pancreatic steatosis and is therefore useful for the detailed characterization of pancreatic steatosis.
Collapse
|
42
|
Ajmera VH, Cachay E, Ramers C, Vodkin I, Bassirian S, Singh S, Mangla N, Bettencourt R, Aldous JL, Park D, Lee D, Blanchard J, Mamidipalli A, Boehringer A, Aslam S, Leinhard OD, Richards L, Sirlin C, Loomba R. MRI Assessment of Treatment Response in HIV-associated NAFLD: A Randomized Trial of a Stearoyl-Coenzyme-A-Desaturase-1 Inhibitor (ARRIVE Trial). Hepatology 2019; 70:1531-1545. [PMID: 31013363 PMCID: PMC7164416 DOI: 10.1002/hep.30674] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 04/01/2019] [Indexed: 12/12/2022]
Abstract
Aramchol, an oral stearoyl-coenzyme-A-desaturase-1 inhibitor, has been shown to reduce hepatic fat content in patients with primary nonalcoholic fatty liver disease (NAFLD); however, its effect in patients with human immunodeficiency virus (HIV)-associated NAFLD is unknown. The aramchol for HIV-associated NAFLD and lipodystrophy (ARRIVE) trial was a double-blind, randomized, investigator-initiated, placebo-controlled trial to test the efficacy of 12 weeks of treatment with aramchol versus placebo in HIV-associated NAFLD. Fifty patients with HIV-associated NAFLD, defined by magnetic resonance imaging (MRI)-proton density fat fraction (PDFF) ≥5%, were randomized to receive either aramchol 600 mg daily (n = 25) or placebo (n = 25) for 12 weeks. The primary endpoint was a change in hepatic fat as measured by MRI-PDFF in colocalized regions of interest. Secondary endpoints included changes in liver stiffness using magnetic resonance elastography (MRE) and vibration-controlled transient elastography (VCTE), and exploratory endpoints included changes in total-body fat and muscle depots on dual-energy X-ray absorptiometry (DXA), whole-body MRI, and cardiac MRI. The mean (± standard deviation) of age and body mass index were 48.2 ± 10.3 years and 30.7 ± 4.6 kg/m2 , respectively. There was no difference in the reduction in mean MRI-PDFF between the aramchol group at -1.3% (baseline MRI-PDFF 15.6% versus end-of-treatment MRI-PDFF 14.4%, P = 0.24) and the placebo group at -1.4% (baseline MRI-PDFF 13.3% versus end-of-treatment MRI-PDFF 11.9%, P = 0.26). There was no difference in the relative decline in mean MRI-PDFF between the aramchol and placebo groups (6.8% versus 1.1%, P = 0.68). There were no differences in MRE-derived and VCTE-derived liver stiffness and whole-body (fat and muscle) composition analysis by MRI or DXA. Compared to baseline, end-of-treatment aminotransferases were lower in the aramchol group but not in the placebo arm. There were no significant adverse events. Conclusion: Aramchol, over a 12-week period, did not reduce hepatic fat or change body fat and muscle composition by using MRI-based assessment in patients with HIV-associated NAFLD (clinicaltrials.gov ID:NCT02684591).
Collapse
Affiliation(s)
- Veeral H. Ajmera
- NAFLD Research Center, Department of Medicine, La Jolla, California,Division of Gastroenterology, Department of Medicine, La Jolla, California
| | - Edward Cachay
- Division of Infectious Diseases, Owen Clinic, University of California San Diego, San Diego, California
| | | | - Irine Vodkin
- Division of Gastroenterology, Department of Medicine, La Jolla, California
| | - Shirin Bassirian
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Seema Singh
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Neeraj Mangla
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | | | | | | | - Daniel Lee
- Division of Infectious Diseases, Owen Clinic, University of California San Diego, San Diego, California
| | - Jennifer Blanchard
- Division of Infectious Diseases, Owen Clinic, University of California San Diego, San Diego, California
| | - Adrija Mamidipalli
- Liver Imaging Group, University of California, San Diego, La Jolla, California
| | - Andrew Boehringer
- Liver Imaging Group, University of California, San Diego, La Jolla, California
| | - Saima Aslam
- Division of Infectious Diseases and Global Public Health, University of California, San Diego, San Diego California
| | - Olof Dahlqvist Leinhard
- AMRA Medical AB, Linkoping Sweden,Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Lisa Richards
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Claude Sirlin
- Liver Imaging Group, University of California, San Diego, La Jolla, California
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, La Jolla, California,Division of Gastroenterology, Department of Medicine, La Jolla, California,Division of Epidemiology, Department of Family and Preventive Medicine, University of California at San Diego, La Jolla, California
| |
Collapse
|
43
|
Milovanovic T, Dragasevic S, Stojkovic Lalosevic M, Zgradic S, Milicic B, Dumic I, Kmezic S, Saponjski D, Antic A, Markovic V, Popovic D. Ultrasonographic Evaluation of Fatty Pancreas in Serbian Patients with Non Alcoholic Fatty Liver Disease-A Cross Sectional Study. ACTA ACUST UNITED AC 2019; 55:medicina55100697. [PMID: 31627351 PMCID: PMC6843527 DOI: 10.3390/medicina55100697] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022]
Abstract
Background and Objectives: The aim of the study was to determine the association between presences of fatty pancreas (FP) with the features of metabolic syndrome (MeS) in patients with non–alcoholic fatty liver disease (NAFLD) and to establish a new noninvasive scoring system for the prediction of FP in patients with NAFLD. Material and Methods: 143 patients with NAFLD were classified according to FP severity grade into the two groups and evaluated for diagnostic criteria of MeS. All patients underwent sonographic examination with adiposity measurements and the liver biopsy. Liver fibrosis was evaluated semi-quantitatively according to the METAVIR scoring system and using non-invasive markers of hepatic fibrosis. Results: Waist circumference (WC) was predictive for increased risk of FP in NAFLD patients. Elevated fasting plasma glucose, total cholesterol, serum amylase and lipase levels were associated with presence of severe FP (p value = 0.052, p value = 0.007, p value = 0.014; p value = 0.024, respectively). Presence of increased amounts of mesenteric fat was associated with severe FP (p value = 0.013). The results of this study demonstrated highly significant association between NAFLD and presence of FP. The model for predicting the presence of FP was designed with probability value above 6.5. Conclusion: Pancreatic fat accumulation leads to worsening of pancreatic function which in turns exacerbates severity of metabolic syndrome associated with both, NAFLD and NAFPD.
Collapse
Affiliation(s)
- Tamara Milovanovic
- School of Medicine, University of Belgrade, 11 000 Belgrade, Serbia.
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, 11 000 Belgrade, Serbia.
| | - Sanja Dragasevic
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, 11 000 Belgrade, Serbia.
| | - Milica Stojkovic Lalosevic
- School of Medicine, University of Belgrade, 11 000 Belgrade, Serbia.
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, 11 000 Belgrade, Serbia.
| | - Sanja Zgradic
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, 11 000 Belgrade, Serbia.
| | - Biljana Milicic
- Institute for Medical Informatics and Biostatistics, School of Dentistry, University of Belgrade, 11 000 Belgrade, Serbia.
| | - Igor Dumic
- Division of Hospital Medicine, Mayo Clinic Health System, Eau Claire, WI 54703, USA.
- Mayo Clinic College of Medicine and Sciences, Rochester, MN 55905, USA.
| | - Stefan Kmezic
- Clinic for Abdominal Surgery-First Surgical Clinic, Clinical Center of Serbia, 11 000 Belgrade, Serbia.
| | - Dusan Saponjski
- Center for Radiology and MRI, Clinical Center of Serbia, 11 000 Belgrade, Serbia.
| | - Andrija Antic
- School of Medicine, University of Belgrade, 11 000 Belgrade, Serbia.
- Clinic for Abdominal Surgery-First Surgical Clinic, Clinical Center of Serbia, 11 000 Belgrade, Serbia.
| | - Velimir Markovic
- School of Medicine, University of Belgrade, 11 000 Belgrade, Serbia.
- Clinic for Abdominal Surgery-First Surgical Clinic, Clinical Center of Serbia, 11 000 Belgrade, Serbia.
| | - Dragan Popovic
- School of Medicine, University of Belgrade, 11 000 Belgrade, Serbia.
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, 11 000 Belgrade, Serbia.
| |
Collapse
|
44
|
Caussy C, Ajmera VH, Puri P, Li-Shin Hsu C, Bassirian S, Mgdsyan M, Singh S, Faulkner C, Valasek MA, Rizo E, Richards L, Brenner DA, Sirlin CB, Sanyal AJ, Loomba R. Serum metabolites detect the presence of advanced fibrosis in derivation and validation cohorts of patients with non-alcoholic fatty liver disease. Gut 2019; 68:1884-1892. [PMID: 30567742 PMCID: PMC8328048 DOI: 10.1136/gutjnl-2018-317584] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 10/22/2018] [Accepted: 11/29/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Non-invasive and accurate diagnostic tests for the screening of disease severity in non-alcoholic fatty liver disease (NAFLD) remain a major unmet need. Therefore, we aimed to examine if a combination of serum metabolites can accurately predict the presence of advanced fibrosis. DESIGN This is a cross-sectional analysis of a prospective derivation cohort including 156 well-characterised patients with biopsy-proven NAFLD and two validation cohorts, including (1) 142 patients assessed using MRI elastography (MRE) and(2) 59 patients with biopsy-proven NAFLD with untargeted serum metabolome profiling. RESULTS In the derivation cohort, 23 participants (15%) had advanced fibrosis and 32 of 652 analysed metabolites were significantly associated with advanced fibrosis after false-discovery rate adjustment. Among the top 10 metabolites, 8 lipids (5alpha-androstan-3beta monosulfate, pregnanediol-3-glucuronide, androsterone sulfate, epiandrosterone sulfate, palmitoleate, dehydroisoandrosterone sulfate, 5alpha-androstan-3beta disulfate, glycocholate), one amino acid (taurine) and one carbohydrate (fucose) were identified. The combined area under the receiver operating characteristic curve (AUROC) of the top 10 metabolite panel was higher than FIB--4 and NAFLD Fibrosis Score (NFS) for the detection of advanced fibrosis: 0.94 (95% CI 0.897 to 0.982) versus 0.78 (95% CI0.674 to 0.891), p=0.002 and versus 0.84 (95% CI 0.724 to 0.929), p=0.017, respectively. The AUROC of the top 10 metabolite panel remained excellent in the independent validation cohorts assessed by MRE or liver biopsy: c-statistic of 0.94 and 0.84, respectively. CONCLUSION A combination of 10 serum metabolites demonstrated excellent discriminatory ability for the detection of advanced fibrosis in an derivation and two independent validation cohorts with greater diagnostic accuracy than the FIB-4-index and NFS. This proof-of-concept study demonstrates that a non-invasive blood-based diagnostic test can provide excellent performance characteristics for the detection of advanced fibrosis.
Collapse
Affiliation(s)
- Cyrielle Caussy
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA,Université Lyon 1, Hospices Civils de Lyon, Lyon, California, France
| | - Veeral H Ajmera
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA
| | - Puneet Puri
- Virginia Commonwealth University, Richmond, Virginia, USA
| | | | - Shirin Bassirian
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA
| | - Mania Mgdsyan
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA
| | - Seema Singh
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA
| | - Claire Faulkner
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA
| | - Mark A Valasek
- Department of Pathology, University of California at San Diego, La Jolla, California, USA
| | - Emily Rizo
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA
| | - Lisa Richards
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA
| | - David A Brenner
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA,Division of Gastroenterology, Department of Medicine, La Jolla, California, USA
| | - Claude B Sirlin
- Liver Imaging Group, Department of Radiology, University of California, San Diego, La Jolla, California, USA
| | - Arun J Sanyal
- Virginia Commonwealth University, Richmond, Virginia, USA
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, La Jolla, California, USA,Division of Gastroenterology, Department of Medicine, La Jolla, California, USA,Division of Epidemiology, Department of Family and Preventive Medicine, University of California at San Diego, La Jolla, California, USA
| |
Collapse
|
45
|
Abstract
OBJECTIVES Pancreatic steatosis or fatty pancreas refers to the fat accumulation in the pancreas, which can lead to inflammation and fibrosis, β-cell dysfunction, fibrosis, and, possibly, pancreatic cancer. This study aimed to study the prevalence of fatty pancreas and its risk factors in patients referred to an endosonography center. METHODS During 18 months, 228 patients who were referred to our endosonography center for various reasons were evaluated for fatty pancreas. Fatty pancreas was defined as hyperechoic pancreas echotexture compared with spleen echotexture. Demographic characteristics, past medical history, and laboratory measurements were compared between groups with and without fatty pancreas to determine the risk factors for fatty pancreas. RESULTS The prevalence of fatty pancreas was 25.9%. Patients with fatty pancreas had a significantly higher mean level of uric acid (P = 0.04), frequency of ischemic heart disease (P = 0.03), hyperlipidemia (P = 0.04), frequency of fatty liver (P < 0.001), and aortic intima thickness (P = 0.01). There was no significant difference in age, sex, body mass index, smoking status, substance abuse, and use of oral contraceptives in the 2 groups. CONCLUSIONS Fatty pancreas is a common disorder. There are meaningful relationships between coronary artery disease, nonalcoholic fatty liver, and atherosclerosis with fatty pancreas.
Collapse
|
46
|
Hepatic and pancreatic fat as imaging biomarkers of metabolic syndrome. RADIOLOGIA 2019; 62:122-130. [PMID: 31447050 DOI: 10.1016/j.rx.2019.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/05/2019] [Accepted: 05/30/2019] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To objectively evaluate hepatic and pancreatic involvement in metabolic syndrome through magnetic resonance imaging (MRI) biomarkers. MATERIAL AND METHODS From an initial retrospective sample of 407 patients diagnosed with metabolic syndrome studied by MRI in a single center during a 2-year period, 154 were excluded because of a lack of clinical and/or laboratory data, pancreatic abnormalities, or inadequate quality of MRI studies. To measure hepatic and pancreatic fat, we used chemical shift imaging (in-phase and out-of-phase), measuring the fat fraction (%) in regions of interest in the pancreas and liver. Associations between the fat fraction and selected clinical and laboratory variables were assessed with beta regression models. RESULTS In the end, 253 patients were included. The hepatic fat fraction was 4.9% and the pancreatic fat fraction was 7.9%. We found no significant associations between the hepatic fat fraction and any of the clinical or laboratory variables. However, the pancreatic fat fraction was positively associated with age (OR=1.025, p<0.001) and baseline glucose (OR=1.005, p<0.001). Patients with diabetes had higher values of pancreatic fat fraction (OR=2.64, p=0.038). Pancreatic fat fraction and hepatic fat fraction were positively associated (OR=69.44, p<0.001). CONCLUSIONS Pancreatic steatosis can be considered a marker of metabolic syndrome and diabetes. Quantitative MRI enables the diagnosis and grading of fatty pancreas through simple chemical shift techniques.
Collapse
|
47
|
Preoperative Evaluation of Pancreatic Fibrosis and Lipomatosis: Correlation of Magnetic Resonance Findings With Histology Using Magnetization Transfer Imaging and Multigradient Echo Magnetic Resonance Imaging. Invest Radiol 2019; 53:720-727. [PMID: 30247172 DOI: 10.1097/rli.0000000000000496] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES The purpose of this study was to evaluate the diagnostic performance of magnetization transfer (MT) imaging and multigradient echo magnetic resonance imaging (MRI) to quantify pancreatic fibrosis and lipomatosis in patients before pancreatoduodenectomy for postoperative risk stratification with histopathology as the reference standard. MATERIALS AND METHODS Twenty-four patients (age, 68 ± 8 years, 16 males) prospectively underwent quantitative MT imaging using a 2-dimensional gradient echo sequence with and without MT prepulse and multigradient echo imaging on a 3 T MRI 1 day before pancreatoduodenectomy due to adenocarcinoma of the pancreatic head region (n = 20), neuroendocrine tumor (n = 3), or intraductal papillary mucinous neoplasm (n = 1). Magnetization transfer ratio (MTR) and proton density fat fraction (PDFF) were measured in pancreatic tail (PT) and at the resection margin (RM). Histopathologically, pancreatic fibrosis was graded as mild, moderate, or severe (F1-F3), lipomatosis was graded as 0% to 10%, 11% to 30%, and greater than 30% fat deposition (L1-L3). In addition, MTR and histopathologic fibrosis was assessed in pancreatic adenocarcinoma. Mann-Whitney U test and Spearman correlation were used. RESULTS Patients with advanced pancreatic fibrosis (F3) showed a significantly higher MTR compared with the F1 group at the RM and PT (38 ± 4 vs 32.3 ± 1.6, P = 0.018 and 39.7 ± 5.5 vs 31.2 ± 1.7, P = 0.001). Spearman correlation coefficient of MTR and fibrosis grade was r = 0.532 (P = 0.011) and 0.554 (P = 0.008), respectively. Pancreatic parenchyma with advanced fat deposition (L2-L3) showed significantly higher PDFF compared with lipomatosis grade L1 (RM: P = 0.002 and PT: P = 0.001). Proton density fat fraction of pancreatic parenchyma exhibited a moderate and significant correlation with histopathologic lipomatosis grade (RM: r = 0.668 and PT: r = 0.707, P < 0.001). Magnetization transfer ratio was significantly higher in pancreatic adenocarcinoma compared with pancreatic parenchyma (44 ± 5.5 vs 36.0 ± 4.4 and 37.4 ± 5.4, P = 0.004). CONCLUSIONS Multiparametric MRI of the pancreas including MTR and PDFF maps may provide quantitative and noninvasive information on pancreatic fibrosis and lipomatosis before surgery.
Collapse
|
48
|
Caussy C, Bhargava M, Villesen IF, Gudmann NS, Leeming DJ, Karsdal MA, Faulkner C, Bao D, Liu A, Lo MT, Bettencourt R, Bassirian S, Richards L, Brenner DA, Chen CH, Sirlin CB, Loomba R. Collagen Formation Assessed by N-Terminal Propeptide of Type 3 Procollagen Is a Heritable Trait and Is Associated With Liver Fibrosis Assessed by Magnetic Resonance Elastography. Hepatology 2019; 70:127-141. [PMID: 30859582 PMCID: PMC6984974 DOI: 10.1002/hep.30610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 03/03/2019] [Indexed: 12/18/2022]
Abstract
N-terminal propeptide of type 3 procollagen (PRO-C3) is a biomarker of liver fibrosis in nonalcoholic fatty liver disease (NAFLD). This study examines the association between PRO-C3 concentration and liver fibrosis assessed by magnetic resonance elastography (MRE)-measured stiffness (MRE-stiffness) and the heritability of PRO-C3 concentration in a cohort of twins and families with and without NAFLD. We performed a cross-sectional analysis of a well-characterized prospective cohort of 306 participants, including 44 probands with NAFLD-cirrhosis and their 72 first-degree relatives, 24 probands with NAFLD without advanced fibrosis and their 24 first-degree relatives, and 72 controls without NAFLD and their 72 first-degree relatives. Liver steatosis was assessed by magnetic resonance imaging proton density fat fraction, and liver fibrosis was assessed by MRE-stiffness. Serum PRO-C3 was assessed by competitive, enzyme-linked immunosorbent assay. We assessed the familial correlation of PRO-C3 concentration, the shared gene effects between PRO-C3 concentration and liver steatosis and fibrosis, and the association between PRO-C3 concentration and genetic variants in the patatin-like phospholipase domain-containing 3 (PNPLA3), transmembrane 6 superfamily member 2 (TM6SF2), membrane-bound O-acyltransferase domain-containing (MBOAT), and glucokinase regulator (CGKR) genes. In multivariable-adjusted models including age, sex, body mass index, and ethnicity, serum PRO-C3 correlated strongly with liver fibrosis (r2 = 0.50, P < 0.001) and demonstrated robust heritability (h2 , 0.36; 95% confidence interval [CI], 0.07, 0.59; P = 0.016). PRO-C3 concentration and steatosis had a strong genetic correlation (shared genetic determination: 0.62; 95% CI, 0.236, 1.001; P = 0.002), whereas PRO-C3 concentration and fibrosis had a strong environmental correlation (shared environmental determination: 0.55; 95% CI, 0.317, 0.717; P < 0.001). PRO-C3 concentrations were higher in carriers of the TM6SF2 rs58542926-T allele compared with noncarriers: 15.7 (± 10.5) versus 10.8 (± 5.7) ng/L (P = 0.047). Conclusion: Serum PRO-C3 correlates with MRE-assessed fibrosis, is heritable, shares genetic correlation with liver steatosis and shares environmental correlation with liver fibrosis. PRO-C3 concentration appears to be linked to both fibrosis and steatosis and increased in carriers of the TM6SF2 rs58542926 risk allele.
Collapse
Affiliation(s)
- Cyrielle Caussy
- NAFLD Research Center, Department of Medicine, La Jolla, California,Université Lyon 1, Hospices Civils de Lyon, Lyon, France
| | - Meera Bhargava
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | | | | | | | | | - Claire Faulkner
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Denny Bao
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Amy Liu
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Min-Tzu Lo
- Department of Radiology, University of California at San Diego, La Jolla, California
| | - Ricki Bettencourt
- NAFLD Research Center, Department of Medicine, La Jolla, California,Division of Epidemiology, Department of Family and Preventive Medicine, University of California at San Diego, La Jolla, California
| | - Shirin Bassirian
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - Lisa Richards
- NAFLD Research Center, Department of Medicine, La Jolla, California
| | - David A. Brenner
- NAFLD Research Center, Department of Medicine, La Jolla, California,Division of Gastroenterology, Department of Medicine, La Jolla, California
| | - Chi-Hua Chen
- Department of Radiology, University of California at San Diego, La Jolla, California
| | - Claude B. Sirlin
- Liver Imaging Group, Department of Radiology, University of California at San Diego, La Jolla, California
| | - Rohit Loomba
- NAFLD Research Center, Department of Medicine, La Jolla, California,Division of Epidemiology, Department of Family and Preventive Medicine, University of California at San Diego, La Jolla, California,Division of Gastroenterology, Department of Medicine, La Jolla, California
| |
Collapse
|
49
|
Fukui H, Hori M, Fukuda Y, Onishi H, Nakamoto A, Ota T, Ogawa K, Ninomiya K, Tatsumi M, Osuga K, Yamada D, Eguchi H, Miyoshi E, Tomiyama N. Evaluation of fatty pancreas by proton density fat fraction using 3-T magnetic resonance imaging and its association with pancreatic cancer. Eur J Radiol 2019; 118:25-31. [PMID: 31439250 DOI: 10.1016/j.ejrad.2019.06.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 05/20/2019] [Accepted: 06/26/2019] [Indexed: 01/03/2023]
Abstract
PURPOSE To evaluate whether pancreatic magnetic resonance imaging-proton density fat fraction (MRI-PDFF) correlates with histological pancreatic fat fraction and its possible usefulness as a biomarker of pancreatic cancer compared with pancreatic index (PI) using computed tomography (CT number of the pancreas divided by that of the spleen). METHOD We included 55 consecutive patients (24 with pancreatic cancer and 31 controls; median age, 72 years) who preoperatively underwent MRI-PDFF using IDEAL-IQ and unenhanced CT and did not receive preoperative therapy. Histologic pancreatic fat fraction was measured in non-tumorous pancreatic tissues at the resection stump. A board-certified radiologist evaluated MRI-PDFF and PI. Correlations were evaluated among MRI-PDFF, PI, and histologic pancreatic fat fraction; the usefulness of MRI-PDFF as a predictor of pancreatic cancer was assessed. RESULTS Histologic pancreatic fat fraction significantly correlated with MRI-PDFF and PI (r = 0.802 and -0.534, respectively; P < 0.01). The absolute correlation coefficient was significantly higher for MRI-PDFF than for PI (P < 0.01). Compared with the control group, the pancreatic cancer group had higher MRI-PDFF and histologic pancreatic fat fraction (P < 0.01) but lower PI (P < 0.01). In multivariate analysis, MRI-PDFF was found to be the sole independent risk factor for pancreatic cancer (odds ratio: 1.19; P < 0.01). CONCLUSIONS Pancreatic fat, which was associated with pancreatic cancer, could be quantified by MRI-PDFF measurement; therefore, MRI-PDFF should be considered as a promising and superior imaging biomarker for estimating the probability of pancreatic cancer than PI.
Collapse
Affiliation(s)
- Hideyuki Fukui
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan.
| | - Masatoshi Hori
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Yasunari Fukuda
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Japan
| | - Hiromitsu Onishi
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Atsushi Nakamoto
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Takashi Ota
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Kazuya Ogawa
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Keisuke Ninomiya
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Mitsuaki Tatsumi
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Keigo Osuga
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry & Clinical Investigation, Osaka University Graduate School of Medicine, Japan
| | - Noriyuki Tomiyama
- Department of Diagnostic and Interventional Radiology, Osaka University Graduate School of Medicine, Japan
| |
Collapse
|
50
|
Chouhan MD, Firmin L, Read S, Amin Z, Taylor SA. Quantitative pancreatic MRI: a pathology-based review. Br J Radiol 2019; 92:20180941. [PMID: 30982337 DOI: 10.1259/bjr.20180941] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
MRI plays an important role in the clinical management of pancreatic disorders and interpretation is reliant on qualitative assessment of anatomy. Conventional sequences capturing pancreatic structure can however be adapted to yield quantitative measures which provide more diagnostic information, with a view to increasing diagnostic accuracy, improving patient stratification, providing robust non-invasive outcome measures for therapeutic trials and ultimately personalizing patient care. In this review, we evaluate the use of established techniques such as secretin-enhanced MR cholangiopancreatography, diffusion-weighted imaging, T 1, T 2* and fat fraction mapping, but also more experimental methods such as MR elastography and arterial spin labelling, and their application to the assessment of diffuse pancreatic disease (including chronic, acute and autoimmune pancreatitis/IgG4 disease, metabolic disease and iron deposition disorders) and cystic/solid focal pancreatic masses. Finally, we explore some of the broader challenges to their implementation and future directions in this promising area.
Collapse
Affiliation(s)
- Manil D Chouhan
- 1 University College London (UCL) Centre for Medical Imaging, Division of Medicine, UCL , London , UK.,2 Department of Imaging, University College London Hospitals (UCLH) NHS Foundation Trust , London , UK
| | - Louisa Firmin
- 2 Department of Imaging, University College London Hospitals (UCLH) NHS Foundation Trust , London , UK
| | - Samantha Read
- 2 Department of Imaging, University College London Hospitals (UCLH) NHS Foundation Trust , London , UK
| | - Zahir Amin
- 2 Department of Imaging, University College London Hospitals (UCLH) NHS Foundation Trust , London , UK
| | - Stuart A Taylor
- 1 University College London (UCL) Centre for Medical Imaging, Division of Medicine, UCL , London , UK.,2 Department of Imaging, University College London Hospitals (UCLH) NHS Foundation Trust , London , UK
| |
Collapse
|