1
|
Lambert JE, Ramos-Roman MA, Valdez MJ, Browning JD, Rogers T, Parks EJ. Weight loss in MASLD restores the balance of liver fatty acid sources. J Clin Invest 2025; 135:e174233. [PMID: 40309768 PMCID: PMC12043097 DOI: 10.1172/jci174233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/06/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUNDLipogenesis contributes substantially to the pathological accumulation of intrahepatic triacylglycerol (IHTG) in metabolic dysfunction-associated steatotic liver disease (MASLD). Since hepatic lipogenesis is highly sensitive to energy intake, we hypothesized that mechanisms of MASLD regression induced by weight loss would be driven by a marked reduction in the lipogenic pathway.METHODSOverweight adults with high liver fat (HighLF; n = 9; IHTG ≥ 5.6% measured by 1H-magnetic resonance spectroscopy) or low (normal) liver fat (LowLF; n = 6; IHTG < 5.6%) received dietary counseling for 6 months and underwent comprehensive metabolic phenotyping during inpatient studies that captured fasting and fed states. Multiple stable isotopes were used to assess the contribution of lipogenesis, free fatty acids (FFAs), and dietary fat to IHTG.RESULTSBody weight loss (-10% ± 2%) reduced IHTG in individuals with MASLD (19.4% ± 3.6% to 4.5% ± 2.1%, P < 0.001). Insulin sensitivity improved significantly (46%, P < 0.01), while fasting FFA flux from adipose tissue was not different. VLDL-triacylglycerol (VLDL-TG) concentrations fell by 38% (P = 0.02) because of a 67% reduction in contribution from lipogenesis (P = 0.02), whereas the absolute contributions from FFAs and dietary fat to VLDL-TG were not different. Reduced lipogenesis was significantly associated with loss of IHTG.CONCLUSIONThese data underscore the primary role of lipogenesis in MASLD pathology and highlight the importance of controlling this pathway through treatment strategies.TRIAL REGISTRATIONClinicalTrials.gov (NCT01371396).FUNDINGNational Institutes of Health (NIH) grant RL1DK081187; Task Force for Obesity Research at Southwestern (TORS) NIH UL1DE019584; and Clinical and Translational Science Award NIH/National Center for Advancing Translational Sciences UL1-RR024982.
Collapse
Affiliation(s)
| | | | | | | | - Thomas Rogers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | |
Collapse
|
2
|
Bernecker M, Lin A, Feuchtinger A, Molenaar A, Schriever SC, Pfluger PT. Weight cycling exacerbates glucose intolerance and hepatic triglyceride storage in mice with a history of chronic high fat diet exposure. J Transl Med 2025; 23:7. [PMID: 39754229 PMCID: PMC11699648 DOI: 10.1186/s12967-024-06039-0] [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: 07/24/2024] [Accepted: 12/25/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Obese subjects undergoing weight loss often fear the Yoyo dieting effect, which involves regaining or even surpassing their initial weight. To date, our understanding of such long-term obesity and weight cycling effects is still limited and often based on only short-term murine weight gain and loss studies. This study aimed to investigate the long-term impacts of weight cycling on glycemic control and metabolic health, focusing on adipose tissue, liver, and hypothalamus. METHODS Chow-fed mice and mice subjected to prolonged high-fat diet (HFD) consumption for 20 weeks, followed by 24 weeks of dietary interventions to either induce weight gain, weight loss, or weight cycling were monitored for perturbations in feeding efficiency and glucose homeostasis. Post-mortem analyses included qPCR, Western Blotting, biochemical and microscopical assessments for hepatic steatosis and insulin resistance, hypothalamic and adipose tissue inflammation, and circulating lipid, leptin and IL-6 levels. RESULTS Weight cycling led to hyperphagia and rapid weight regain, matching the weights of mice continuously on HFD. Despite weight loss, adipose tissue inflammation persisted with elevated pro-inflammatory markers, macrophage infiltration, and impaired Glut4 expression. HFD-induced dysregulation in hypothalamic expression of orexigenic peptides and synaptic plasticity markers persisted also after weight normalization suggesting long-lasting neural alterations. Weight-cycled mice exhibited higher circulating IL-6 and leptin levels, increased hepatic lipid storage, and dysregulated glucose metabolism compared to those with consistent diets, indicating worsened metabolic effects by Yoyo dieting. CONCLUSION In sum, our study highlights significant metabolic risks associated with weight cycling, particularly following prolonged obesity. Persistent adipose tissue inflammation, perturbed neural peptide and plasticity markers and impaired glucose tolerance emphasize the need for effective and sustainable weight loss strategies to mitigate the adverse outcomes of weight regain and improve long-term metabolic health.
Collapse
Affiliation(s)
- Miriam Bernecker
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of NeuroBiology of Diabetes, TUM School of Medicine & Health, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Anna Lin
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Annette Feuchtinger
- Core Facility Pathology and Tissue Analytics, Helmholtz Munich, Neuherberg, Germany
| | - Anna Molenaar
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- Division of NeuroBiology of Diabetes, TUM School of Medicine & Health, Technical University of Munich, Munich, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Sonja C Schriever
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Paul T Pfluger
- Research Unit NeuroBiology of Diabetes, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- Institute for Diabetes and Obesity, Helmholtz Munich, Neuherberg, Germany.
- Division of NeuroBiology of Diabetes, TUM School of Medicine & Health, Technical University of Munich, Munich, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
| |
Collapse
|
3
|
Laine S, Sjöros T, Garthwaite T, Honka MJ, Löyttyniemi E, Eskola O, Saarenhovi M, Kallio P, Koivumäki M, Vähä-Ypyä H, Sievänen H, Vasankari T, Hirvonen J, Laitinen K, Houttu N, Kalliokoski K, Saunavaara V, Knuuti J, Heinonen IHA. Daily standing time, dietary fiber, and intake of unsaturated fatty acids are beneficially associated with hepatic insulin sensitivity in adults with metabolic syndrome. Front Endocrinol (Lausanne) 2024; 15:1272886. [PMID: 38989003 PMCID: PMC11233550 DOI: 10.3389/fendo.2024.1272886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 06/11/2024] [Indexed: 07/12/2024] Open
Abstract
Background Obesity is associated with impaired glucose metabolism and hepatic insulin resistance. The aim was to investigate the associations of hepatic glucose uptake (HGU) and endogenous glucose production (EGP) to sedentary behavior (SB), physical activity (PA), cardiorespiratory fitness, dietary factors, and metabolic risk markers. Methods Forty-four adults with metabolic syndrome (mean age 58 [SD 7] years, BMI ranging from 25-40kg/; 25 females) were included. HGU was measured by positron emission tomography during the hyperinsulinemic-euglycemic clamp. EGP was calculated by subtracting the glucose infusion rate during clamp from the glucose rate of disappearance. SB and PA were measured with hip-worn accelerometers (26 [SD3] days). Fitness was assessed by maximal bicycle ergometry with respiratory gas measurements and dietary intake of nutrients by 4-day food diaries. Results HGU was not associated with fitness or any of the SB or PA measures. When adjusted for sex, age, and body fat-%, HGU was associated with whole-body insulin sensitivity (β=0.58), water-insoluble dietary fiber (β=0.29), energy percent (E%) of carbohydrates (β=-0.32), saccharose (β=-0.32), mono- and polyunsaturated fatty acids (β=0.35, β=0.41, respectively). EGP was associated with whole-body insulin sensitivity (β=-0.53), and low-density lipoprotein cholesterol [β=-0.31], and when further adjusted for accelerometry wear time, EGP was associated with standing [β=-0.43]. (p-value for all< 0.05). Conclusions Standing more, consuming a diet rich in fiber and unsaturated fatty acids, and a lower intake of carbohydrates, especially sugar, associate beneficially with hepatic insulin sensitivity. Habitual SB, PA, or fitness may not be the primary modulators of HGU and EGP. However, these associations need to be confirmed with intervention studies.
Collapse
Affiliation(s)
- Saara Laine
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
| | - Tanja Sjöros
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
| | - Taru Garthwaite
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
| | - Miikka-Juhani Honka
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
| | | | - Olli Eskola
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
| | - Maria Saarenhovi
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, University of Turku and Turku University Hospital, Turku, Finland
| | - Petri Kallio
- Department of Clinical Physiology and Nuclear Medicine, University of Turku and Turku University Hospital, Turku, Finland
- Paavo Nurmi Center, University of Turku, Turku, Finland
| | - Mikko Koivumäki
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
| | - Henri Vähä-Ypyä
- The UKK Institute for Health Promotion Research, Tampere, Finland
| | - Harri Sievänen
- The UKK Institute for Health Promotion Research, Tampere, Finland
| | - Tommi Vasankari
- The UKK Institute for Health Promotion Research, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jussi Hirvonen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Radiology, University of Turku and Turku University Hospital, Turku, Finland
| | - Kirsi Laitinen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Noora Houttu
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kari Kalliokoski
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
| | - Virva Saunavaara
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
- Department of Medical Physics, Division of Medical Imaging, Turku University Hospital, Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
| | - Ilkka H A Heinonen
- Turku PET Centre, University of Turku, Åbo Akademi University and Turku University Hospital, Turku, Finland
| |
Collapse
|
4
|
Huang X, Zhao Y, Liu T, Wu D, Shu J, Yue W, Zhang W, Liu S. β-Cell Function and Insulin Dynamics in Obese Patients With and Without Diabetes After Sleeve Gastrectomy. Diabetes 2024; 73:572-584. [PMID: 37257028 DOI: 10.2337/db22-1048] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
Improved β-cell function seems to be essential for better glucose homeostasis after Roux-en-Y gastric bypass but is less studied after sleeve gastrectomy (SG). We evaluated the effects of SG on β-cell function in obese patients with diabetes (DM group) and without (control group) in response to both oral and intravenous glucose stimulation. The DM group demonstrated impaired insulin sensitivity and insulin response to glucose before surgery. The insulin sensitivity index of both groups significantly improved after SG. In addition, the insulin response to glucose (early insulinogenic index in oral glucose tolerance test and acute insulin response to glucose in an intravenous glucose tolerance test) increased in the DM group but decreased in the control group. As a result, β-cell function improved significantly in both groups after SG since the disposition index (DI) increased in both. However, the DI of the DM group was not restored to the level of control group up to 1 year after SG. Our results support that obese patients, with and without diabetes, could benefit from SG in β-cell function. For obese patients at risk for or who have been diagnosed with diabetes, interventions should be recommended early to preserve or restore β-cell function, and SG could be an effective choice. Further studies are needed for long-term effects. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Xin Huang
- Division of Bariatric and Metabolic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yian Zhao
- School of Clinical Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Teng Liu
- Division of Bariatric and Metabolic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Dong Wu
- Division of Pancreatic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Jiaxin Shu
- School of Clinical Medicine, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenwen Yue
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wenjing Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shaozhuang Liu
- Division of Bariatric and Metabolic Surgery, Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
5
|
Basset-Sagarminaga J, van de Weijer T, Iozzo P, Schrauwen P, Schrauwen-Hinderling V. Advances and challenges in measuring hepatic glucose uptake with FDG PET: implications for diabetes research. Diabetologia 2024; 67:407-419. [PMID: 38099962 DOI: 10.1007/s00125-023-06055-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/10/2023] [Indexed: 02/06/2024]
Abstract
The liver plays a crucial role in the control of glucose homeostasis and is therefore of great interest in the investigation of the development of type 2 diabetes. Hepatic glucose uptake (HGU) can be measured through positron emission tomography (PET) imaging with the tracer [18F]-2-fluoro-2-deoxy-D-glucose (FDG). HGU is dependent on many variables (e.g. plasma glucose, insulin and glucagon concentrations), and the metabolic state for HGU assessment should be chosen with care and coherence with the study question. In addition, as HGU is influenced by many factors, protocols and measurement conditions need to be standardised for reproducible results. This review provides insights into the protocols that are available for the measurement of HGU by FDG PET and discusses the current state of knowledge of HGU and its impairment in type 2 diabetes. Overall, a scanning modality that allows for the measurement of detailed kinetic information and influx rates (dynamic imaging) may be preferable to static imaging. The combination of FDG PET and insulin stimulation is crucial to measure tissue-specific insulin sensitivity. While the hyperinsulinaemic-euglycaemic clamp allows for standardised measurements under controlled blood glucose levels, some research questions might require a more physiological approach, such as oral glucose loading, with both advantages and complexities relating to fluctuations in blood glucose and insulin levels. The available approaches to address HGU hold great potential but await more systematic exploitation to improve our understanding of the mechanisms underlying metabolic diseases. Current findings from the investigation of HGU by FDG PET highlight the complex interplay between insulin resistance, hepatic glucose metabolism, NEFA levels and intrahepatic lipid accumulation in type 2 diabetes and obesity. Further research is needed to fully understand the underlying mechanisms and potential therapeutic targets for improving HGU in these conditions.
Collapse
Affiliation(s)
- Jeremy Basset-Sagarminaga
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Tineke van de Weijer
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands
| | - Vera Schrauwen-Hinderling
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, the Netherlands.
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.
| |
Collapse
|
6
|
Ali S, Baig S, Wanninayake S, da Silva Xavier G, Dawson C, Paisey R, Geberhiwot T. Glucagon-like peptide-1 analogues in monogenic syndromic obesity: Real-world data from a large cohort of Alström syndrome patients. Diabetes Obes Metab 2024; 26:989-996. [PMID: 38151964 DOI: 10.1111/dom.15398] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/10/2023] [Accepted: 11/20/2023] [Indexed: 12/29/2023]
Abstract
AIM To examine the real-world efficacy of glucagon-like peptide-1 receptor agonists (GLP-1 RAs) in monogenic obesity in patients with Alström syndrome (ALMS). METHODS We screened 72 UK adult patients with ALMS and offered treatment to 34 patients meeting one of the following criteria: body mass index of 25 kg/m2 or higher, insulin resistance, suboptimal glycaemic control on antihyperglycaemic medications or non-alcoholic fatty liver disease. RESULTS In total, 30 patients, with a mean age of 31 ± 11 years and a male to-female ratio of 2:1, completed 6 months of treatment with GLP-1 RAs either in the form of semaglutide or exenatide. On average, treatment with GLP-1 RAs reduced body weight by 5.4 ± 1.7 (95% confidence interval [CI] 3.6-7) kg and HbA1c by 12 ± 3.3 (95% CI 8.7-15.3) mmol/mol, equating to 6% weight loss (P < .01) and 1.1% absolute reduction in HbA1c (P < .01). Significant improvements were also observed in serum total cholesterol, triglycerides, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol and alanine aminotransferase. The improvement of metabolic variables in our cohort of monogenic syndromic obesity was comparable with data for polygenic obesity, irrespective of weight loss. CONCLUSIONS Data from our centre highlight the non-inferiority of GLP-1 RAs in monogenic syndromic obesity to the available GLP-1 RA-use data in polygenic obesity, therefore, these agents can be considered as a treatment option in patients with ALMS, as well as other forms of monogenic obesity.
Collapse
Affiliation(s)
- Sadaf Ali
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Shanat Baig
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | | | | | - Charlotte Dawson
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Richard Paisey
- Torbay and South Devon NHS Foundation Trust, Torquay, Torbay, UK
| | - Tarekegn Geberhiwot
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
7
|
Ustsinau U, Ehret V, Fürnsinn C, Scherer T, Helbich TH, Hacker M, Krššák M, Philippe C. Novel approach using [ 18F]FTHA-PET and de novo synthesized VLDL for assessment of FFA metabolism in a rat model of diet induced NAFLD. Clin Nutr 2023; 42:1839-1848. [PMID: 37625314 DOI: 10.1016/j.clnu.2023.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND AND AIMS The worldwide prevalence of Non-alcoholic Fatty Liver Disease (NAFLD) raises concerns about associated risk factors, such as obesity and type 2 Diabetes Mellitus, for leading causes of disability and death. Besides Magnetic Resonance Imaging (MRI) and Spectroscopy (MRS), functional imaging with Positron Emission Tomography (PET) could contribute to a deeper understanding of the pathophysiology of NAFLD. Here we describe a novel approach using the PET tracer [18F]FTHA, which is an analog of long-chain free fatty acids (FFA) and is taken up by tissues to enter mitochondria or to be incorporated into complex lipids for further export as very-low-density lipoprotein (VLDL). METHODS Male Sprague Dawley rats, after 6 weeks on a high-fat diet (HFD), were used as a model of diet induced NAFLD, while a standard diet (SD) served as a control group. Liver fat was estimated by MR spectroscopy at a 9.4 T system for phenotyping. To measure hepatic FFA uptake, rats underwent 60 min dynamic [18F]FTHA-PET scans after unrestricted access to food (HFD: n = 6; SD: n = 6) or overnight (≤16h) fasting (HFD: n = 6; SD: n = 5). FFA removal was assessed from incorporated 18F-residual in de novo synthesized VLDL out of plasma. RESULTS MRS of the liver confirmed the presence of NAFLD (>5.6% fat). Under non-fasting conditions, hepatic [18F]FTHA uptake was significantly increased in NAFLD: SUVmean (p = 0.03) within [0; 60] min interval, SUVmean (p = 0.01) and SUVmax (p = 0.03) within [30; 60] min interval. SUVs for hepatic uptake under fasting conditions were not significantly different between the groups. Analysis of FFA removal demonstrated elevated values of 18F-residue in the VLDL plasma fraction of the healthy group compared to the NAFLD (p = 0.0569). CONCLUSION Our novel approach for assessing FFA metabolism using [18F]FTHA demonstrated differences in the hepatic FFA uptake and FFA incorporation into VLDL between healthy and NAFLD rats. [18F]FTHA-PET could be used to study metabolic disturbances involved in the progression of NAFLD.
Collapse
Affiliation(s)
- Usevalad Ustsinau
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Viktoria Ehret
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Clemens Fürnsinn
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas H Helbich
- Division of Molecular and Structural Preclinical Imaging, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Martin Krššák
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Cecile Philippe
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
8
|
Pang MD, Bastings JJAJ, Op den Kamp-Bruls YMH, Harrold JA, Kjølbaek L, Halford JCG, Adam TCM, Raben A, Schrauwen-Hinderling VB, Goossens GH, Blaak EE. The effect of weight loss on whole-body and tissue-specific insulin sensitivity and hepatic lipid content and composition: SWEET substudy. Obesity (Silver Spring) 2023; 31:1745-1754. [PMID: 37368517 DOI: 10.1002/oby.23773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/22/2023] [Accepted: 03/13/2023] [Indexed: 06/29/2023]
Abstract
OBJECTIVE This study (1) investigated the effect of weight loss on whole-body and tissue-specific insulin sensitivity and on intrahepatic lipid (IHL) content and composition and (2) investigated the association between weight-loss-induced changes in insulin sensitivity and IHL content in individuals with overweight or obesity. METHODS In this secondary analysis of the European SWEET project, 50 adults (age 18-65 years) with overweight or obesity (BMI ≥ 25 kg/m2 ) followed a low-energy diet (LED) for 2 months. At baseline and after the LED, body composition (dual-energy x-ray absorptiometry), IHL content and composition (proton magnetic resonance spectroscopy), whole-body insulin sensitivity (Matsuda index), muscle insulin sensitivity index (MISI), and hepatic insulin resistance index (HIRI) were determined (7-point oral glucose tolerance test). RESULTS The LED reduced body weight (p < 0.001). This was accompanied by increased Matsuda index and reduced HIRI (both p < 0.001) but no change in MISI (p = 0.260). Weight loss decreased IHL content (mean [SEM], 3.9% [0.7%] vs. 1.6% [0.5%], p < 0.001) and the hepatic saturated fatty acid fraction (41.0% [1.5%] vs. 36.6% [1.9%], p = 0.039). The reduced IHL content was associated with an improvement in HIRI (r = 0.402, p = 0.025). CONCLUSIONS Weight loss decreased IHL content and the hepatic saturated fatty acid fraction. The decrease in IHL content was associated with weight-loss-induced improvement in hepatic insulin sensitivity in individuals with overweight or obesity.
Collapse
Affiliation(s)
- Michelle D Pang
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Jacco J A J Bastings
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Yvonne M H Op den Kamp-Bruls
- Department of Radiology and Nuclear Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Joanne A Harrold
- Department of Psychology, Institute of Population Health, University of Liverpool, Liverpool, UK
| | - Louise Kjølbaek
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Jason C G Halford
- Department of Psychology, Institute of Population Health, University of Liverpool, Liverpool, UK
- School of Psychology, University of Leeds, Leeds, UK
| | - Tanja C M Adam
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Anne Raben
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
- Clinical Research, Copenhagen University Hospital-Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Vera B Schrauwen-Hinderling
- Department of Radiology and Nuclear Medicine, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Institute for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Gijs H Goossens
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ellen E Blaak
- Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
9
|
Bril F, Sanyal A, Cusi K. Metabolic Syndrome and Its Association with Nonalcoholic Steatohepatitis. Clin Liver Dis 2023; 27:187-210. [PMID: 37024202 DOI: 10.1016/j.cld.2023.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
The relationship between insulin resistance, metabolic syndrome (MetS), and nonalcoholic fatty liver disease (NAFLD) is complicated. Although insulin resistance is almost universal in people with NAFLD and MetS, NAFLD may be present without features of MetS and vice versa. While NAFLD has a strong correlation with cardiometabolic risk factors, these are not intrinsic components of this condition. Taken together, our knowledge gaps call for caution regarding the common assertion that NAFLD is the hepatic manifestation of the MetS, and for defining NAFLD in broad terms as a "metabolic dysfunction" based on a diverse and poorly understood constellation of cardiometabolic features.
Collapse
Affiliation(s)
- Fernando Bril
- Division of Endocrinology, Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Arun Sanyal
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine Internal Medicine, Virginia Commonwealth University
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, Gainesville, FL, USA
| |
Collapse
|
10
|
Hepatic Positron Emission Tomography: Applications in Metabolism, Haemodynamics and Cancer. Metabolites 2022; 12:metabo12040321. [PMID: 35448508 PMCID: PMC9026326 DOI: 10.3390/metabo12040321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 11/28/2022] Open
Abstract
Evaluating in vivo the metabolic rates of the human liver has been a challenge due to its unique perfusion system. Positron emission tomography (PET) represents the current gold standard for assessing non-invasively tissue metabolic rates in vivo. Here, we review the existing literature on the assessment of hepatic metabolism, haemodynamics and cancer with PET. The tracer mainly used in metabolic studies has been [18F]2-fluoro-2-deoxy-D-glucose (18F-FDG). Its application not only enables the evaluation of hepatic glucose uptake in a variety of metabolic conditions and interventions, but based on the kinetics of 18F-FDG, endogenous glucose production can also be assessed. 14(R,S)-[18F]fluoro-6-thia-Heptadecanoic acid (18F-FTHA), 11C-Palmitate and 11C-Acetate have also been applied for the assessment of hepatic fatty acid uptake rates (18F-FTHA and 11C-Palmitate) and blood flow and oxidation (11C-Acetate). Oxygen-15 labelled water (15O-H2O) has been used for the quantification of hepatic perfusion. 18F-FDG is also the most common tracer used for hepatic cancer diagnostics, whereas 11C-Acetate has also shown some promising applications in imaging liver malignancies. The modelling approaches used to analyse PET data and also the challenges in utilizing PET in the assessment of hepatic metabolism are presented.
Collapse
|
11
|
Bavaro DF, Laghetti P, Poliseno M, De Gennaro N, Di Gennaro F, Saracino A. A Step Closer to the "Fourth 90": A Practical Narrative Review of Diagnosis and Management of Nutritional Issues of People Living with HIV. Diagnostics (Basel) 2021; 11:2047. [PMID: 34829394 PMCID: PMC8618448 DOI: 10.3390/diagnostics11112047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/02/2022] Open
Abstract
The quality of life of people living with HIV (PLWH) has remarkably increased thanks to the introduction of combined antiretroviral therapy. Still, PLWH are exposed to an increased risk of cardiovascular diseases, diabetes, chronic kidney disease, and liver disease. Hence, the purpose of this review is to summarize the current knowledge about diagnosis and nutritional management with specific indication of macro and micronutrients intake for the main comorbidities of PLWH. In fact, a prompt diagnosis and management of lifestyle behaviors are fundamental steps to reach the "fourth 90". To achieve an early diagnosis of these comorbidities, clinicians have at their disposal algorithms such as the Framingham Score to assess cardiovascular risk; transient elastography and liver biopsy to detect NAFLD and NASH; and markers such as the oral glucose tolerance test and GFR to identify glucose impairment and renal failure, respectively. Furthermore, maintenance of ideal body weight is the goal for reducing cardiovascular risk and to improve diabetes, steatosis and fibrosis; while Mediterranean and low-carbohydrate diets are the dietetic approaches proposed for cardioprotective effects and for glycemic control, respectively. Conversely, diet management of chronic kidney disease requires different nutritional assessment, especially regarding protein intake, according to disease stage and eventually concomitant diabetes.
Collapse
Affiliation(s)
- Davide Fiore Bavaro
- Clinic of Infectious Diseases, University Hospital Policlinico, University of Bari, 70124 Bari, Italy; (P.L.); (N.D.G.); (F.D.G.); (A.S.)
| | - Paola Laghetti
- Clinic of Infectious Diseases, University Hospital Policlinico, University of Bari, 70124 Bari, Italy; (P.L.); (N.D.G.); (F.D.G.); (A.S.)
| | | | - Nicolò De Gennaro
- Clinic of Infectious Diseases, University Hospital Policlinico, University of Bari, 70124 Bari, Italy; (P.L.); (N.D.G.); (F.D.G.); (A.S.)
| | - Francesco Di Gennaro
- Clinic of Infectious Diseases, University Hospital Policlinico, University of Bari, 70124 Bari, Italy; (P.L.); (N.D.G.); (F.D.G.); (A.S.)
| | - Annalisa Saracino
- Clinic of Infectious Diseases, University Hospital Policlinico, University of Bari, 70124 Bari, Italy; (P.L.); (N.D.G.); (F.D.G.); (A.S.)
| |
Collapse
|
12
|
Song Y, Jensen MD. Red blood cell triglycerides - a unique pool that incorporates plasma free fatty acids and relates to metabolic health. J Lipid Res 2021; 62:100131. [PMID: 34619142 PMCID: PMC8566996 DOI: 10.1016/j.jlr.2021.100131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/14/2021] [Accepted: 09/28/2021] [Indexed: 01/12/2023] Open
Abstract
Most research into red blood cell (RBC) lipids focuses on membrane phospholipids and their relationships to metabolic conditions and diet. Triglycerides (TGs) exist in most cells; the TG-fatty acids serve as readily available fuel for oxidative phosphorylation. Because RBCs lack mitochondria, they would not be expected to store fatty acids in TG. We followed up on a previous in vitro study that found FFA can be incorporated into RBC-TG by testing whether intravenously infused [U-13C]palmitate could be detected in RBC-TG. We also quantified RBC-TG fatty acid concentrations and profiles as they relate to plasma FFA and lipid concentrations. We found that 1) RBC-TG concentrations measured by glycerol and LC/MS were correlated (r = 0.77; P < 0.001) and averaged <50 nmol/ml RBC; 2) RBC-TG concentrations were stable over 18 h; 3) [U-13C]palmitate was detectable in RBC-TG from half the participants; 4) RBC-TGs were enriched in saturated fatty acids and depleted in unsaturated fatty acid compared with plasma FFA and previously reported RBC membrane phospholipids; 5) RBC-TG fatty acid profiles differed significantly between obese and nonobese adults; 6) weight loss altered the RBC-TG fatty acid profile in the obese group; and 7) the RBC-TG fatty acid composition correlated with plasma lipid concentrations. This is the first report showing that plasma FFA contributes to RBC-TG in vivo, in humans, and that the RBC-TG fatty acid profile is related to metabolic health. The storage of saturated fatty acids in RBC-TG stands in stark contrast to the highly unsaturated profile reported in RBC membrane phospholipids.
Collapse
Affiliation(s)
- Yilin Song
- Division of Endocrinology, Diabetes and Metabolism, Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota 55905, US
| | - Michael D Jensen
- Division of Endocrinology, Diabetes and Metabolism, Endocrine Research Unit, Mayo Clinic, Rochester, Minnesota 55905, US.
| |
Collapse
|
13
|
Hallsworth K, Dombrowski SU, McPherson S, Anstee QM, Avery L. Using the theoretical domains framework to identify barriers and enabling factors to implementation of guidance for the diagnosis and management of nonalcoholic fatty liver disease: a qualitative study. Transl Behav Med 2021; 10:1016-1030. [PMID: 31120519 PMCID: PMC7543077 DOI: 10.1093/tbm/ibz080] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver condition worldwide and is steadily on the increase. In response, national and international guidance have been developed to standardize diagnosis and guide management of the condition. However, research has highlighted a discordance between published guidance and clinical practice. The purpose of this study is to identify barriers and enabling factors to implementation of guidance to inform the development of an intervention. We interviewed 21 health care professionals and 12 patients with NAFLD. Topic guides were developed with reference to national and international guidance. Data were content analyzed using the Theoretical Domains Framework. Beliefs about consequences and professional role and identity were the most prominent domains identified from health care professionals in the context of diagnosis and management of NAFLD. Environmental context and resources, memory, attention and decision processes, goals, behavioral regulation, knowledge, and skills emerged as important barriers/facilitators to implementation of guidance targeting management of NAFLD. Knowledge and beliefs about consequences were the most prominent domains from the perspective of patients. Social influences, environmental context and resources and behavioral regulation were most prominent in the context of NAFLD management. Guideline implementation can be improved by use of interventions that target standardized use of diagnostic criteria by health care professionals. Training of health care professionals was identified as important to improve care delivered to patients in order to effectively manage NAFLD. Interventions that target knowledge of patients, in particular, raising awareness that NAFLD can be progressive when not actively managed would facilitate implementation of guidance.
Collapse
Affiliation(s)
- Kate Hallsworth
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Liver Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | | | - Stuart McPherson
- Liver Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Quentin M Anstee
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Liver Unit, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Leah Avery
- Centre for Rehabilitation, Exercise and Sports Science, Teesside University, Tees Valley, UK
| |
Collapse
|
14
|
Rebelos E, Iozzo P, Guzzardi MA, Brunetto MR, Bonino F. Brain-gut-liver interactions across the spectrum of insulin resistance in metabolic fatty liver disease. World J Gastroenterol 2021; 27:4999-5018. [PMID: 34497431 PMCID: PMC8384743 DOI: 10.3748/wjg.v27.i30.4999] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/29/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Metabolic associated fatty liver disease (MAFLD), formerly named "nonalcoholic fatty liver disease" occurs in about one-third of the general population of developed countries worldwide and behaves as a major morbidity and mortality risk factor for major causes of death, such as cardiovascular, digestive, metabolic, neoplastic and neuro-degenerative diseases. However, progression of MAFLD and its associated systemic complications occur almost invariably in patients who experience the additional burden of intrahepatic and/or systemic inflammation, which acts as disease accelerator. Our review is focused on the new knowledge about the brain-gut-liver axis in the context of metabolic dysregulations associated with fatty liver, where insulin resistance has been assumed to play an important role. Special emphasis has been given to digital imaging studies and in particular to positron emission tomography, as it represents a unique opportunity for the noninvasive in vivo study of tissue metabolism. An exhaustive revision of targeted animal models is also provided in order to clarify what the available preclinical evidence suggests for the causal interactions between fatty liver, dysregulated endogenous glucose production and insulin resistance.
Collapse
Affiliation(s)
- Eleni Rebelos
- Turku PET Centre, University of Turku, Turku 20500, Finland
| | - Patricia Iozzo
- Institute of Clinical Physiology, National Research Council, Pisa 56124, Italy
| | | | - Maurizia Rossana Brunetto
- Hepatology Unit and Laboratory of Molecular Genetics and Pathology of Hepatitis, Pisa University Hospital, Pisa 56121, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56121, Italy
- Institute of Biostructure and Bioimaging, National Research Council, Napoli 80145, Italy
| | - Ferruccio Bonino
- Institute of Biostructure and Bioimaging, National Research Council, Napoli 80145, Italy
| |
Collapse
|
15
|
Hajifathalian K, Mehta A, Ang B, Skaf D, Shah SL, Saumoy M, Dawod Q, Dawod E, Shukla A, Aronne L, Brown RS, Cohen DE, Dannenberg AJ, Fortune B, Kumar S, Sharaiha RZ. Improvement in insulin resistance and estimated hepatic steatosis and fibrosis after endoscopic sleeve gastroplasty. Gastrointest Endosc 2021; 93:1110-1118. [PMID: 32861753 DOI: 10.1016/j.gie.2020.08.023] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/23/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease in the United States and is closely associated with obesity and insulin resistance (IR). Weight loss is the best treatment for NAFLD. Endoscopic sleeve gastroplasty (ESG) is a promising endoscopic procedure for treatment of obesity. Our aim is to evaluate the change in IR and estimated hepatic steatosis and fibrosis after ESG. METHODS One hundred eighteen patients with obesity and NAFLD underwent ESG and were followed for 2 years. Weight loss was evaluated as % total body weight loss. IR was evaluated using the homeostasis model assessment of insulin resistance (HOMA-IR). The previously validated hepatic steatosis index and NAFLD fibrosis score were used to estimate hepatic steatosis and risk of fibrosis. RESULTS Patients' mean body mass index was 40 ± 7 kg/m2 at baseline. Eighty-four percent of patients completed 2 years of follow-up. At 2 years, the mean total body weight loss was 15.5% (95% confidence interval, 13.3%-17.8%). Patients' HOMA-IR improved significantly from 6.7 ± 11 to 3.0 ± 1.6 after only 1 week from ESG (P = .019) with continued improvement up to 2 years (P = .03). Patients' hepatic steatosis index score improved significantly, decreasing by 4 points per year (P for trend, <.001). Patients' NAFLD fibrosis score improved significantly, decreasing by 0.3 point per year (P for trend, .034). Twenty-four patients (20%) improved their risk of hepatic fibrosis from F3-F4 or indeterminate to F0-F2, whereas only 1 patient (1%) experienced an increase in the estimated risk of fibrosis (P = .02). CONCLUSIONS Our results suggest a significant and sustained improvement in estimated hepatic steatosis and fibrosis after ESG in patients with NAFLD. Importantly, we showed an early and weight-independent improvement in insulin resistance, which lasted for 2 years after the procedure.
Collapse
Affiliation(s)
- Kaveh Hajifathalian
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - Amit Mehta
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - Bryan Ang
- Joan & Sanford I. Weill Medical College of Cornell University, New York, New York
| | - Daniel Skaf
- Joan & Sanford I. Weill Medical College of Cornell University, New York, New York
| | - Shawn L Shah
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - Monica Saumoy
- Gastroenterology and Hepatology Department, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Qais Dawod
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - Enad Dawod
- Joan & Sanford I. Weill Medical College of Cornell University, New York, New York
| | - Alpana Shukla
- Division of Endocrinology Diabetes and Metabolism, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - Louis Aronne
- Division of Endocrinology Diabetes and Metabolism, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - Robert S Brown
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - David E Cohen
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - Andrew J Dannenberg
- Department of Medicine, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York, USA
| | - Brett Fortune
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - Sonal Kumar
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| | - Reem Z Sharaiha
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York-Presbyterian Hospital, New York, New York
| |
Collapse
|
16
|
Holzhütter HG, Berndt N. Computational Hypothesis: How Intra-Hepatic Functional Heterogeneity May Influence the Cascading Progression of Free Fatty Acid-Induced Non-Alcoholic Fatty Liver Disease (NAFLD). Cells 2021; 10:cells10030578. [PMID: 33808045 PMCID: PMC7999144 DOI: 10.3390/cells10030578] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is the most common type of chronic liver disease in developed nations, affecting around 25% of the population. Elucidating the factors causing NAFLD in individual patients to progress in different rates and to different degrees of severity, is a matter of active medical research. Here, we aim to provide evidence that the intra-hepatic heterogeneity of rheological, metabolic and tissue-regenerating capacities plays a central role in disease progression. We developed a generic mathematical model that constitutes the liver as ensemble of small liver units differing in their capacities to metabolize potentially cytotoxic free fatty acids (FFAs) and to repair FFA-induced cell damage. Transition from simple steatosis to more severe forms of NAFLD is described as self-amplifying process of cascading liver failure, which, to stop, depends essentially on the distribution of functional capacities across the liver. Model simulations provided the following insights: (1) A persistently high plasma level of FFAs is sufficient to drive the liver through different stages of NAFLD; (2) Presence of NAFLD amplifies the deleterious impact of additional tissue-damaging hits; and (3) Coexistence of non-steatotic and highly steatotic regions is indicative for the later occurrence of severe NAFLD stages.
Collapse
Affiliation(s)
- Hermann-Georg Holzhütter
- Institute of Biochemistry, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Correspondence:
| | - Nikolaus Berndt
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany;
| |
Collapse
|
17
|
Behari J, Graham L, Wang R, Schirda C, Borhani AA, Methé BA, Li K, Morris A, Luu HN, Palmieri S, Yuan JM. Dynamics of hepatic steatosis resolution and changes in gut microbiome with weight loss in nonalcoholic fatty liver disease. Obes Sci Pract 2021; 7:217-225. [PMID: 33841891 PMCID: PMC8019274 DOI: 10.1002/osp4.476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/09/2020] [Accepted: 11/28/2020] [Indexed: 01/22/2023] Open
Abstract
Background Weight loss is recommended as the primary treatment for nonalcoholic fatty liver disease (NAFLD). However, the magnitude and velocity of hepatic steatosis resolution with weight loss is unclear, making it difficult to counsel patients seeking weight loss for treatment of NAFLD. The aim of this study was to determine the rate of hepatic steatosis improvement and stool microbiome changes associated with rapid diet‐induced weight loss in NAFLD. Methods Fourteen NAFLD patients (mean ± standard deviation, body mass index [BMI] 36.4 ± 4 kg/m2) enrolled in a 12‐week meal replacement program underwent frequent measurement of Fibroscan‐controlled attenuation parameter (CAP). Magnetic resonance imaging (MRI‐Dixon method) for hepatic fat quantitation and stool microbiome analysis (16S rRNA gene sequencing) were completed in 11 subjects at baseline and Week 12. Results At Week 12, mean (95% confidence interval) weight loss was −13.4 (−15.2, −11.5)% and CAP score −26.6 (−35.6, −17.6)% (both Ps < 0.001). CAP scores changed at a rate of −4.9 dB/m/kg (−30.1 dB/m per unit BMI) in Weeks 1–4 and −0.6 dB/m/kg (−2.4 dB/m per unit BMI) in Weeks 8–12. MRI‐determined hepatic fat fraction decreased by −74.1% (p < 0.001) at a rate of −0.51%/kg (−3.19% per unit BMI), with complete steatosis resolution in 90% patients. BMI change was associated with decreased stool microbial diversity (coefficient = 0.17; Shannon Index), increased abundance of Prevotella_9 (Bacteroidetes; coefficient = 0.96) and decreased abundance of Phascolarctobacterium (Firmicutes; coefficient = −0.42) (both Ps < 0.05). Conclusions Diet‐induced intensive weight loss is associated with rapid improvement and complete resolution of hepatic steatosis and decreased stool microbial diversity. These findings highlight the dynamic nature of hepatic fat and may help clinicians to develop evidence‐based treatment goals for patients with NAFLD and obesity who undertake weight loss interventions. Further research is warranted to understand the effects of intensive weight loss and gut microbiome changes on long‐term NAFLD resolution.
Collapse
Affiliation(s)
- Jaideep Behari
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition University of Pittsburgh School of Medicine Pittsburgh Pennsylvania USA.,Pittsburgh Liver Research Center University of Pittsburgh Pittsburgh Pennsylvania USA.,Cancer Epidemiology and Prevention Program University of Pittsburgh Medical Center Hillman Cancer Center Pittsburgh Pennsylvania USA
| | - Lisa Graham
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition University of Pittsburgh School of Medicine Pittsburgh Pennsylvania USA
| | - Renwei Wang
- Cancer Epidemiology and Prevention Program University of Pittsburgh Medical Center Hillman Cancer Center Pittsburgh Pennsylvania USA
| | - Claudiu Schirda
- Department of Radiology University of Pittsburgh School of Medicine Pittsburgh Pennsylvania USA
| | - Amir A Borhani
- Department of Radiology University of Pittsburgh School of Medicine Pittsburgh Pennsylvania USA
| | - Barbara A Methé
- Center for Medicine and the Microbiome University of Pittsburgh Pittsburgh Pennsylvania USA.,Division of Pulmonary, Allergy and Critical Care Medicine University of Pittsburgh School of Medicine Pittsburgh Pennsylvania USA
| | - Kelvin Li
- Center for Medicine and the Microbiome University of Pittsburgh Pittsburgh Pennsylvania USA
| | - Alison Morris
- Center for Medicine and the Microbiome University of Pittsburgh Pittsburgh Pennsylvania USA.,Division of Pulmonary, Allergy and Critical Care Medicine University of Pittsburgh School of Medicine Pittsburgh Pennsylvania USA
| | - Hung N Luu
- Cancer Epidemiology and Prevention Program University of Pittsburgh Medical Center Hillman Cancer Center Pittsburgh Pennsylvania USA.,Department of Epidemiology, Graduate School of Public Health University of Pittsburgh Pittsburgh Pennsylvania USA
| | - Sabrina Palmieri
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition University of Pittsburgh School of Medicine Pittsburgh Pennsylvania USA
| | - Jian-Min Yuan
- Cancer Epidemiology and Prevention Program University of Pittsburgh Medical Center Hillman Cancer Center Pittsburgh Pennsylvania USA.,Department of Epidemiology, Graduate School of Public Health University of Pittsburgh Pittsburgh Pennsylvania USA
| |
Collapse
|
18
|
Koffert J, Lahti L, Nylund L, Salminen S, Hannukainen JC, Salminen P, de Vos WM, Nuutila P. Partial restoration of normal intestinal microbiota in morbidly obese women six months after bariatric surgery. PeerJ 2020; 8:e10442. [PMID: 33304658 PMCID: PMC7700738 DOI: 10.7717/peerj.10442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/06/2020] [Indexed: 12/18/2022] Open
Abstract
We studied the impact of bariatric surgery on the intestinal microbiota of morbidly obese study subjects. A total of 13 morbidly obese women (five of which had type 2 diabetes) and 14 healthy age- and gender-matched controls were recruited and the microbiota composition of fecal samples were determined by using a phylogenetic microarray. Sampling of the patients took place just one month before and 6 months after the operation. Within six months after bariatric surgery, the obese subjects had lost on average a quarter of their weight whereas four of the five of the diabetic subjects were in remission. Bariatric surgery was associated with an increased microbial community richness and Bacteroidetes/Firmicutes ratio. In addition, we observed an increased relative abundance of facultative anaerobes, such as Streptococcus spp., and a reduction in specific butyrate-producing Firmicutes. The observed postoperative alterations in intestinal microbiota reflect adaptation to the changing conditions in the gastrointestinal tract, such as energy restriction and the inability to process fiber-rich foods after bariatric surgery.
Collapse
Affiliation(s)
- Jukka Koffert
- Department of Gastroenterology, Turku University Hospital, Turku, Finland.,Turku PET Centre, University of Turku, Turku, Finland
| | - Leo Lahti
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
| | - Lotta Nylund
- Functional Foods Forum, University of Turku, Turku, Finland.,Food Chemistry and Food Development, Department of Biochemistry, University of Turku, Turku, Finland
| | - Seppo Salminen
- Functional Foods Forum, University of Turku, Turku, Finland
| | | | - Paulina Salminen
- Division of Digestive Surgery and Urology, Turku University Hospital, Turku, Finland
| | - Willem M de Vos
- RPU Immunobiology and Human Microbiome, University of Helsinki, Helsinki, Finland.,Laboratory of Microbiology, Wageningen University and Research, Wageningen, The Netherlands
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland.,Department of Endocrinology, Turku University hospital, Turku, Finland
| |
Collapse
|
19
|
Kruse M, Kemper M, Gancheva S, Osterhoff M, Dannenberger D, Markgraf D, Machann J, Hierholzer J, Roden M, Pfeiffer AFH. Dietary Rapeseed Oil Supplementation Reduces Hepatic Steatosis in Obese Men-A Randomized Controlled Trial. Mol Nutr Food Res 2020; 64:e2000419. [PMID: 32920973 DOI: 10.1002/mnfr.202000419] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/13/2020] [Indexed: 01/01/2023]
Abstract
SCOPE Effective treatment for obesity associated non-alcoholic fatty liver disease (NAFLD) is limited. Dietary supplementation of n-3 polyunsaturated fatty acids, specifically alpha linolenic acid (ALA), can resolve intrahepatic lipid content (IHL). This study investigates the effect of daily supplementation of either refined rapeseed (RA), containing high amounts of ALA, or refined olive (OL) oil on IHL and glucose metabolism in NAFLD patients. METHODS AND RESULTS 27 obese men consumed an isocaloric diet including either 50 g of RA or OL daily for 8 weeks. Hepatic proton magnetic resonance spectroscopy, hyperinsulinemic-euglycemic clamp studies and blood tests are performed before and at the end of the study. At 8 weeks a significant reduction in IHL is observed for RA (13.1 ± 1.6 before versus 11.1 ± 1.6% after intervention) versus OL (13.3 ± 2.5 before versus 15.7 ± 2.7% after intervention). For RA, a 21% reduction (P < 0.02) in serum free fatty acids (FFA) and a 1.68-fold increase (P = 0.03) of serum interleukin-6 (IL-6) is observed after 8 weeks. CONCLUSION RA has a beneficial effect on hepatic lipid metabolism as shown by reduced IHL and serum FFA. RA induced IL-6 production seems to be liver protective confirming previous results.
Collapse
Affiliation(s)
- Michael Kruse
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Clinical Nutrition, 14558, Nuthetal, Germany.,Charité University Medicine Berlin, Department of Endocrinology, 10117, Berlin, Germany
| | - Margrit Kemper
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Clinical Nutrition, 14558, Nuthetal, Germany.,Charité University Medicine Berlin, Department of Endocrinology, 10117, Berlin, Germany.,German Center for Diabetes Research (DZD), München, 85764, Neuherberg, Germany
| | - Sofiya Gancheva
- German Center for Diabetes Research (DZD), München, 85764, Neuherberg, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, 40225, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center Düsseldorf, Leibniz Center for Diabetes Research at Heinrich-Heine University, 40225, Düsseldorf, Germany
| | - Martin Osterhoff
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Clinical Nutrition, 14558, Nuthetal, Germany.,Charité University Medicine Berlin, Department of Endocrinology, 10117, Berlin, Germany
| | - Dirk Dannenberger
- Leibniz Institute for Farm Animal Biology, Institute of Muscle Biology and Growth, 18196, Dummerstorf, Germany
| | - Daniel Markgraf
- German Center for Diabetes Research (DZD), München, 85764, Neuherberg, Germany.,Institute for Clinical Diabetology, German Diabetes Center Düsseldorf, Leibniz Center for Diabetes Research at Heinrich-Heine University, 40225, Düsseldorf, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD), München, 85764, Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, 72076, Tübingen, Germany.,Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Johannes Hierholzer
- Department of Diagnostic and Interventional Radiology, Ernst von Bergmann Hospital, 14467, Potsdam, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD), München, 85764, Neuherberg, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich-Heine University, 40225, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center Düsseldorf, Leibniz Center for Diabetes Research at Heinrich-Heine University, 40225, Düsseldorf, Germany
| | - Andreas F H Pfeiffer
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Clinical Nutrition, 14558, Nuthetal, Germany.,Charité University Medicine Berlin, Department of Endocrinology, 10117, Berlin, Germany.,German Center for Diabetes Research (DZD), München, 85764, Neuherberg, Germany
| |
Collapse
|
20
|
Gullberg GT, Shrestha UM, Seo Y. PET imaging of glucose and fatty acid metabolism for NAFLD patients. J Nucl Cardiol 2020; 27:1689-1697. [PMID: 30547298 PMCID: PMC8356561 DOI: 10.1007/s12350-018-01532-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Grant T Gullberg
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA.
| | - Uttam M Shrestha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| |
Collapse
|
21
|
Ren G, Bowers RL, Kim T, Araya-Ramirez F, Mahurin AJ, Dean DM, Grandjean PW, Mathews ST. Alterations of Serum Ser312-Phosphorylated Fetuin-A from Exercise-Induced Moderate Body Weight Loss in Individuals with Obesity. Obesity (Silver Spring) 2020; 28:544-551. [PMID: 32012464 DOI: 10.1002/oby.22730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/14/2019] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Phosphorylated fetuin-A (pFet-A) inhibits insulin action and has been shown to be associated with obesity and insulin resistance. The objective of this cohort study was to assess the effect of incremental body weight loss on alterations in serum pFet-A and indexes of insulin sensitivity. METHODS A total of 16 men with obesity attained a targeted weight loss of 8% to 10% of their initial body weight by achieving an energy expenditure/deficit of 2,000 to 2,500 kcal/wk. Anthropometric assessments and blood samples were obtained every 4 weeks. Weight loss was calculated and partitioned as 2% to 4%, 4% to 6%, 6% to 8%, and 8% to 10% compared with initial body weight. RESULTS Targeted body weight loss of 8% to 10% decreased serum pFet-A, pFet-A:Fet-A ratio, fasting insulin, log(homeostasis model assessment of insulin resistance), quantitative insulin sensitivity check index, adipose insulin resistance, and insulin resistance index significantly. Percent changes in serum pFet-A were associated with percent changes in indexes of insulin sensitivity. Unlike insulin sensitivity indexes, which were altered starting with 6% to 8% weight loss, serum pFet-A levels were significantly decreased by 19.6% starting with 2% to 4% weight loss and decreased by 25.6%, 36.8%, and 42.3% with 4% to 6%, 6% to 8%, and 8% to 10% weight loss, respectively. CONCLUSIONS This study reports for the first time that the insulin-sensitizing effects of moderate weight loss are associated with a reduction in serum pFet-A levels.
Collapse
Affiliation(s)
- Guang Ren
- Department of Nutrition and Dietetics, Auburn University, Auburn, Alabama, USA
| | - Robert L Bowers
- School of Kinesiology, Auburn University, Auburn, Alabama, USA
| | - Teayoun Kim
- Department of Nutrition and Dietetics, Auburn University, Auburn, Alabama, USA
| | | | - A Jack Mahurin
- School of Kinesiology, Auburn University, Auburn, Alabama, USA
| | - David M Dean
- School of Kinesiology, Auburn University, Auburn, Alabama, USA
| | | | - Suresh T Mathews
- Department of Nutrition and Dietetics, Auburn University, Auburn, Alabama, USA
| |
Collapse
|
22
|
Warner SO, Yao MV, Cason RL, Winnick JJ. Exercise-Induced Improvements to Whole Body Glucose Metabolism in Type 2 Diabetes: The Essential Role of the Liver. Front Endocrinol (Lausanne) 2020; 11:567. [PMID: 32982968 PMCID: PMC7484211 DOI: 10.3389/fendo.2020.00567] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/13/2020] [Indexed: 01/22/2023] Open
Abstract
Type 2 diabetes (T2D) is a metabolic disease characterized by obesity, insulin resistance, and the dysfunction of several key glucoregulatory organs. Among these organs, impaired liver function is recognized as one of the earliest contributors to impaired whole-body glucose homeostasis, with well-characterized hepatic insulin resistance resulting in elevated rates of hepatic glucose production (HGP) and fasting hyperglycemia. One portion of this review will provide an overview of how HGP is regulated during the fasted state in healthy humans and how this process becomes dysregulated in patients with T2D. Less well-appreciated is the liver's role in post-prandial glucose metabolism, where it takes up and metabolizes one-third of orally ingested glucose. An abundance of literature has shown that the process of hepatic glucose uptake is impaired in patients with T2D, thereby contributing to glucose intolerance. A second portion of this review will outline how hepatic glucose uptake is regulated during the post-prandial state, and how it becomes dysfunctional in patients with T2D. Finally, it is well-known that exercise training has an insulin-sensitizing effect on the liver, which contributes to improved whole-body glucose metabolism in patients with T2D, thereby making it a cornerstone in the management of the disease. To this end, the impact of exercise on hepatic glucose metabolism will be thoroughly discussed, referencing key findings in the literature. At the same time, sources of heterogeneity that contribute to inconsistent findings in the field will be pointed out, as will important topics for future investigation.
Collapse
Affiliation(s)
- Shana O. Warner
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Michael V. Yao
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Rebecca L. Cason
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Jason J. Winnick
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- *Correspondence: Jason J. Winnick
| |
Collapse
|
23
|
Stokes CS, Lammert F, Krawczyk M. Short-term Dietary Interventions for the Management of Nonalcoholic Fatty Liver. Curr Med Chem 2019; 26:3483-3496. [PMID: 28482789 DOI: 10.2174/0929867324666170508144409] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 04/16/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) affects millions of individuals on a global scale and currently no gold standard treatment exists. The risk of developing NAFLD is considerably higher with increasing body mass index. Consequently, weight loss should be recommended to all overweight patients with fatty liver. However, lifestyle interventions, irrespective of weight status, may also influence the condition. The aim herein is to present examples of short-term interventions which assess direct effects of dietary-related components on hepatic steatosis. METHODS This review includes studies with short-term dietary-related interventions of up to 16 weeks that evaluate their efficacy in reducing intrahepatic lipid contents (hepatic steatosis). This review primarily focuses on the three main macronutrients: dietary carbohydrates, fats and proteins. RESULTS High saturated fat intake and high consumption of carbohydrates, particularly from simple sugars such as fructose are reported as risk factors for hepatic steatosis. Overall, shortterm hypocaloric diets have shown beneficial effects in reducing intrahepatic lipid contents. Macronutrient manipulations such as carbohydrate restriction as well as the consumption of unsaturated fatty acids are also reported to have efficacious effects. CONCLUSION This review highlights the different dietary interventions that can influence hepatic steatosis in the short term, illustrating both pro and anti-steatotic effects.
Collapse
Affiliation(s)
- Caroline S Stokes
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.,Laboratory of Metabolic Liver Diseases, Center for Preclinical Research, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
24
|
Oh TJ, Yu JM, Min KW, Son HS, Lee MK, Yoon KH, Song YD, Park JY, Jeong IK, Cha BS, Kim YS, Baik SH, Kim IJ, Kim DM, Kim SR, Lee KW, Park JH, Lee IK, Park TS, Choi SH, Park SW. Efficacy and Safety of Voglibose Plus Metformin in Patients with Type 2 Diabetes Mellitus: A Randomized Controlled Trial. Diabetes Metab J 2019; 43:276-286. [PMID: 30604594 PMCID: PMC6581551 DOI: 10.4093/dmj.2018.0051] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/22/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Combination of metformin to reduce the fasting plasma glucose level and an α-glucosidase inhibitor to decrease the postprandial glucose level is expected to generate a complementary effect. We compared the efficacy and safety of a fixed-dose combination of voglibose plus metformin (vogmet) with metformin monotherapy in drug-naïve newly-diagnosed type 2 diabetes mellitus. METHODS A total of 187 eligible patients aged 20 to 70 years, with a glycosylated hemoglobin (HbA1c) level of 7.0% to 11.0%, were randomized into either vogmet or metformin treatments for 24 weeks. A change in the HbA1c level from baseline was measured at week 24. RESULTS The reduction in the levels of HbA1c was -1.62%±0.07% in the vogmet group and -1.31%±0.07% in the metformin group (P=0.003), and significantly more vogmet-treated patients achieved the target HbA1c levels of <6.5% (P=0.002) or <7% (P=0.039). Glycemic variability was also significantly improved with vogmet treatment, estimated by M-values (P=0.004). Gastrointestinal adverse events and hypoglycemia (%) were numerically lower in the vogmet-treated group. Moreover, a significant weight loss was observed with vogmet treatment compared with metformin (-1.63 kg vs. -0.86 kg, P=0.039). CONCLUSION Vogmet is a safe antihyperglycemic agent that controls blood glucose level effectively, yields weight loss, and is superior to metformin in terms of various key glycemic parameters without increasing the risk of hypoglycemia.
Collapse
Affiliation(s)
- Tae Jung Oh
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jae Myung Yu
- Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Kyung Wan Min
- Department of Internal Medicine, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, Korea
| | - Hyun Shik Son
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Moon Kyu Lee
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kun Ho Yoon
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Young Duk Song
- Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, Goyang, Korea
| | - Joong Yeol Park
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - In Kyung Jeong
- Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul, Korea
| | - Bong Soo Cha
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yong Seong Kim
- Department of Internal Medicine, Inha University School of Medicine, Incheon, Korea
| | - Sei Hyun Baik
- Diabetes Center, Endocrinology and Metabolism, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - In Joo Kim
- Department of Internal Medicine, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Korea
| | - Doo Man Kim
- Department of Internal Medicine, Hallym University Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Sung Rae Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kwan Woo Lee
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Korea
| | - Jeong Hyung Park
- Department of Internal Medicine, Inje University Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - In Kyu Lee
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Tae Sun Park
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University Medical School, Jeonju, Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea.
| | - Sung Woo Park
- Department of Endocrinology and Metabolism, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
25
|
Uranga RM, Keller JN. The Complex Interactions Between Obesity, Metabolism and the Brain. Front Neurosci 2019; 13:513. [PMID: 31178685 PMCID: PMC6542999 DOI: 10.3389/fnins.2019.00513] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 05/06/2019] [Indexed: 12/22/2022] Open
Abstract
Obesity is increasing at unprecedented levels globally, and the overall impact of obesity on the various organ systems of the body is only beginning to be fully appreciated. Because of the myriad of direct and indirect effects of obesity causing dysfunction of multiple tissues and organs, it is likely that there will be heterogeneity in the presentation of obesity effects in any given population. Taken together, these realities make it increasingly difficult to understand the complex interplay between obesity effects on different organs, including the brain. The focus of this review is to provide a comprehensive view of metabolic disturbances present in obesity, their direct and indirect effects on the different organ systems of the body, and to discuss the interaction of these effects in the context of brain aging and the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Romina María Uranga
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas, Bahía Blanca, Argentina
- Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Jeffrey Neil Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| |
Collapse
|
26
|
Motiani KK, Savolainen AM, Toivanen J, Eskelinen JJ, Yli-Karjanmaa M, Virtanen KA, Saunavaara V, Heiskanen MA, Parkkola R, Haaparanta-Solin M, Solin O, Savisto N, Löyttyniemi E, Knuuti J, Nuutila P, Kalliokoski KK, Hannukainen JC. Effects of short-term sprint interval and moderate-intensity continuous training on liver fat content, lipoprotein profile, and substrate uptake: a randomized trial. J Appl Physiol (1985) 2019; 126:1756-1768. [PMID: 30998125 DOI: 10.1152/japplphysiol.00900.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Type 2 diabetes (T2D) and increased liver fat content (LFC) alter lipoprotein profile and composition and impair liver substrate uptake. Exercise training mitigates T2D and reduces LFC, but the benefits of different training intensities in terms of lipoprotein classes and liver substrate uptake are unclear. The aim of this study was to evaluate the effects of moderate-intensity continuous training (MICT) or sprint interval training (SIT) on LFC, liver substrate uptake, and lipoprotein profile in subjects with normoglycemia or prediabetes/T2D. We randomized 54 subjects (normoglycemic group, n = 28; group with prediabetes/T2D, n = 26; age = 40-55 yr) to perform either MICT or SIT for 2 wk and measured LFC with magnetic resonance spectroscopy, lipoprotein composition with NMR, and liver glucose uptake (GU) and fatty acid uptake (FAU) using PET. At baseline, the group with prediabetes/T2D had higher LFC, impaired lipoprotein profile, and lower whole body insulin sensitivity and aerobic capacity compared with the normoglycemic group. Both training modes improved aerobic capacity (P < 0.001) and lipoprotein profile (reduced LDL and increased large HDL subclasses; all P < 0.05) with no training regimen (SIT vs. MICT) or group effect (normoglycemia vs. prediabetes/T2D). LFC tended to be reduced in the group with prediabetes/T2D compared with the normoglycemic group posttraining (P = 0.051). When subjects were divided according to LFC (high LFC, >5.6%; low LFC, <5.6%), training reduced LFC in subjects with high LFC (P = 0.009), and only MICT increased insulin-stimulated liver GU (P = 0.03). Short-term SIT and MICT are effective in reducing LFC in subjects with fatty liver and in improving lipoprotein profile regardless of baseline glucose tolerance. Short-term MICT is more efficient in improving liver insulin sensitivity compared with SIT. NEW & NOTEWORTHY In the short term, both sprint interval training and moderate-intensity continuous training (MICT) reduce liver fat content and improve lipoprotein profile; however, MICT seems to be preferable in improving liver insulin sensitivity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Virva Saunavaara
- Turku PET Centre, University of Turku , Turku , Finland.,Department of Medical Physics, Turku University Hospital , Turku , Finland
| | | | - Riitta Parkkola
- Department of Radiology, Turku University Hospital , Turku , Finland
| | - Merja Haaparanta-Solin
- Turku PET Centre, University of Turku , Turku , Finland.,MediCity Research Laboratory Turku, University of Turku , Turku , Finland
| | - Olof Solin
- Turku PET Centre, University of Turku , Turku , Finland.,Department of Chemistry, University of Turku , Turku , Finland.,Turku PET Centre, Åbo Akademi University , Turku , Finland
| | - Nina Savisto
- Turku PET Centre, University of Turku , Turku , Finland
| | | | - Juhani Knuuti
- Turku PET Centre, University of Turku , Turku , Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku , Turku , Finland.,Department of Endocrinology, Turku University Hospital , Turku , Finland
| | | | | |
Collapse
|
27
|
Xu W, Zhou H, Xuan H, Saha P, Wang G, Chen W. Novel metabolic disorders in skeletal muscle of Lipodystrophic Bscl2/Seipin deficient mice. Mol Cell Endocrinol 2019; 482:1-10. [PMID: 30521848 PMCID: PMC6340772 DOI: 10.1016/j.mce.2018.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 01/23/2023]
Abstract
Bscl2-/- mice recapitulate many of the major metabolic manifestations in Berardinelli-Seip congenital lipodystrophy type 2 (BSCL2) individuals, including lipodystrophy, hepatosteatosis, muscular hypertrophy, and insulin resistance. Metabolic defects in Bscl2-/- mice with regard to glucose and lipid metabolism in skeletal muscle have never been investigated. Here, we identified Bscl2-/- mice displayed reduced intramyocellular triglyceride (IMTG) content but increased glycogen storage predominantly in oxidative type I soleus muscle (SM). These changes were associated with increased incomplete fatty acid oxidation and glycogen synthesis. Interestingly, SM in Bscl2-/- mice demonstrated a fasting duration induced insulin sensitivity which was further confirmed by hyperinsulinemic-euglycemic clamp in SM of overnight fasted Bscl2-/- mice but reversed by raising circulating NEFA levels through intralipid infusion. Furthermore, mice with skeletal muscle-specific inactivation of BSCL2 manifested no changes in muscle deposition of lipids and glycogen, suggesting BSCL2 does not play a cell-autonomous role in muscle lipid and glucose homeostasis. Our study uncovers a novel link between muscle metabolic defects and insulin resistance, and underscores an important role of circulating NEFA in regulating oxidative muscle insulin signaling in BSCL2 lipodystrophy.
Collapse
Affiliation(s)
- Wenqiong Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, PR China; Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hongyi Zhou
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hongzhuan Xuan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA; School of Life Science, Liaocheng University, Liaocheng, Shandong Province, 252059, PR China
| | - Pradip Saha
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gongxian Wang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, PR China.
| | - Weiqin Chen
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
28
|
González-Granillo M, Savva C, Li X, Fitch M, Pedrelli M, Hellerstein M, Parini P, Korach-André M, Gustafsson JÅ. ERβ activation in obesity improves whole body metabolism via adipose tissue function and enhanced mitochondria biogenesis. Mol Cell Endocrinol 2019; 479:147-158. [PMID: 30342056 DOI: 10.1016/j.mce.2018.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/13/2018] [Accepted: 10/07/2018] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Estrogens play a key role in the distribution of adipose tissue and have their action by binding to both estrogen receptors (ER), α and β. Although ERβ has a role in the energy metabolism, limited data of the physiological mechanism and metabolic response involved in the pharmacological activation of ERβ is available. METHODS For clinical relevance, non-ovariectomized female mice were subjected to high fat diet together with pharmacological (DIP - 4-(2-(3,5-dimethylisoxazol-4-yl)-1H-indol-3-yl)phenol) interventions to ERβ selective activation. The physiological mechanism was assessed in vivo by magnetic resonance imaging and spectroscopy, and oral glucose and intraperitoneal insulin tolerance test before and after DIP treatment. Liver and adipose tissue metabolic response was measured in HFD + vehicle and HFD + DIP by stable isotope, RNA sequencing and protein content. RESULTS HFD-fed females treated with DIP had a tissue-specific response towards ERβ selective activation. The metabolic profile showed an improved fasting glucose level, insulin sensitivity and reduced liver steatosis. CONCLUSIONS Our data demonstrate that selective activation of ERβ exerts a tissue-specific activity which promotes a beneficial effect on whole body metabolic response to obesity.
Collapse
Affiliation(s)
- Marcela González-Granillo
- Department of Medicine, Metabolism Unit and KI/AZ Integrated CardioMetabolic Center (ICMC), Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden; Department of Biosciences and Nutrition Huddinge, Karolinska Institutet, Sweden.
| | - Christina Savva
- Department of Medicine, Metabolism Unit and KI/AZ Integrated CardioMetabolic Center (ICMC), Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden; Department of Biosciences and Nutrition Huddinge, Karolinska Institutet, Sweden
| | - Xidan Li
- Department of Medicine, Metabolism Unit and KI/AZ Integrated CardioMetabolic Center (ICMC), Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mark Fitch
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, USA
| | - Matteo Pedrelli
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Sweden
| | - Marc Hellerstein
- Department of Nutritional Sciences & Toxicology, University of California, Berkeley, USA
| | - Paolo Parini
- Department of Medicine and Department of Laboratory Medicine, Karolinska Institutet, Sweden
| | - Marion Korach-André
- Department of Medicine, Metabolism Unit and KI/AZ Integrated CardioMetabolic Center (ICMC), Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden; Department of Biosciences and Nutrition Huddinge, Karolinska Institutet, Sweden.
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition Huddinge, Karolinska Institutet, Sweden; Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signalling, University of Houston, Houston, TX, USA
| |
Collapse
|
29
|
Shang W, Li H, Strappe P, Zhou Z, Blanchard C. Konjac glucomannans attenuate diet-induced fat accumulation on livers and its regulation pathway. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
30
|
Anholm C, Kumarathurai P, Jürs A, Pedersen LR, Nielsen OW, Kristiansen OP, Fenger M, Holst JJ, Madsbad S, Sajadieh A, Haugaard SB. Liraglutide improves the beta-cell function without increasing insulin secretion during a mixed meal in patients, who exhibit well-controlled type 2 diabetes and coronary artery disease. Diabetol Metab Syndr 2019; 11:42. [PMID: 31164926 PMCID: PMC6543623 DOI: 10.1186/s13098-019-0438-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/17/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Hyperinsulinemia aggravates insulin resistance and cardio-vascular disease. How the insulinotropic glucagon-like peptide-1 receptor agonist liraglutide in a physiologic post-prandial setting may act on pancreatic alpha and beta-cell function in patients with coronary artery disease (CAD) and type 2 diabetes (T2DM) is less clear. METHODS Insulin resistant patients with established CAD and newly diagnosed well-controlled T2DM were recruited to a placebo-controlled, cross-over trial with two treatment periods of 12 weeks and a 2 weeks wash-out period before and in-between. Treatment was liraglutide or placebo titrated from 0.6 mg q.d. to 1.8 mg q.d. within 4 weeks and metformin titrated from 500 mg b.i.d to 1000 mg b.i.d. within 4 weeks. Before and after intervention in both 12 weeks periods insulin, C-peptide, glucose, and glucagon were measured during a meal test. Beta-cell function derived from the oral glucose tolerance setting was calculated as changes in insulin secretion per unit changes in glucose concentration (Btotal) and whole-body insulin resistance using ISIcomposite. RESULTS Liraglutide increased the disposition index [Btotal × ISIcomposite, by 40% (n = 24, p < 0.001)] compared to placebo. Post-prandial insulin and glucose was reduced by metformin in combination with liraglutide and differed, but not significantly different from placebo, moreover, glucagon concentration was unaffected. Additionally, insulin clearance tended to increase during liraglutide therapy (n = 26, p = 0.06). CONCLUSIONS The insulinotropic drug liraglutide may without increasing the insulin concentration reduce postprandial glucose but not glucagon excursions and improve beta-cell function in newly diagnosed and well-controlled T2DM.Trial registration Clinicaltrials.gov ID: NCT01595789.
Collapse
Affiliation(s)
- Christian Anholm
- Department of Internal Medicine, Copenhagen University Hospital Glostrup, Nordre Ringvej 57, 2600 Glostrup, Denmark
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Preman Kumarathurai
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Anders Jürs
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Lene Rørholm Pedersen
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Olav Wendelboe Nielsen
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Ole Peter Kristiansen
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Mogens Fenger
- Department of Clinical Biochemistry, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
| | - Jens Juul Holst
- NovoNordisk Foundation Center for Metabolic Research and Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Copenhagen, Denmark
| | - Ahmad Sajadieh
- Department of Cardiology, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| | - Steen Bendix Haugaard
- Department of Internal Medicine, Copenhagen University Hospital Glostrup, Nordre Ringvej 57, 2600 Glostrup, Denmark
- Department of Endocrinology I, Copenhagen University Hospital Bispebjerg, Copenhagen, Denmark
| |
Collapse
|
31
|
Rayner JJ, Abdesselam I, Peterzan MA, Akoumianakis I, Akawi N, Antoniades C, Tomlinson JW, Neubauer S, Rider OJ. Very low calorie diets are associated with transient ventricular impairment before reversal of diastolic dysfunction in obesity. Int J Obes (Lond) 2018; 43:2536-2544. [PMID: 30464235 PMCID: PMC6892735 DOI: 10.1038/s41366-018-0263-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/15/2018] [Accepted: 10/26/2018] [Indexed: 01/06/2023]
Abstract
Objectives Very low calorie diets (VLCDs) are effective at clearing hepatic steatosis and improving insulin sensitivity. Whilst long-term weight loss is beneficial to the cardiovascular system, the acute elevation in fatty acids during caloric restriction is potentially detrimental to cardiac metabolism and function. We sought to investigate any cardiovascular changes occurring over the course of a modern VLCD regime, alongside the expected peripheral metabolic improvements. Methods 25 obese volunteers (BMI 36.8 ± 5.8 kg/m2) underwent magnetic resonance imaging, echocardiography, metabolic profiling, and bio-impedance analysis before 1 and 8 weeks following a VLCD (800 kcal/day). Results were compared to 15 age- and sex-matched controls. Results After 1 week of VLCD, despite only modest weight loss, significant drops occurred in liver fat and insulin resistance (HOMA-IR; by 14–50%, all p < 0.01). In contrast, myocardial triglyceride content (MTGC) increased (by 48%, p = 0.030), and was associated with deterioration in both systolic (LVEF by 4%, p = 0.041) and diastolic function (e/e′ 8.6 ± 1.4 to 9.4 ± 1.7, p = 0.019). Aortic stiffness also increased by 35% (p = 0.015). At 8 weeks, liver steatosis and visceral fat were lower than baseline (by 20–55%, p < 0.001), and peripheral metabolic improvements continued. MTGC also fell to below baseline (1.5 ± 0.6 vs 2.1 ± 1%, p = 0.05) with improved myocardial function (e/e′ 8.6 ± 1.4 to 7.5 ± 1.5, p = 0.003). Conclusions Whilst VLCDs result in dramatic improvements in insulin resistance, they are associated with transient but significant cardiovascular functional decline, which may have an impact on those with the coexisting cardiac disease. However, after 8 weeks, the diet was associated with normalisation of cardiac function, suggesting they may form a potential therapeutic intervention for diastolic dysfunction in obesity and diabetes.
Collapse
Affiliation(s)
- Jennifer J Rayner
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| | - Ines Abdesselam
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Mark A Peterzan
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ioannis Akoumianakis
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Nadia Akawi
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Oliver J Rider
- Oxford Centre for Clinical Magnetic Resonance Research, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
32
|
Woo M, Song YO, Kang KH, Noh JS. Anti-Obesity Effects of Collagen Peptide Derived from Skate ( Raja kenojei) Skin Through Regulation of Lipid Metabolism. Mar Drugs 2018; 16:md16090306. [PMID: 30200239 PMCID: PMC6164805 DOI: 10.3390/md16090306] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 11/16/2022] Open
Abstract
This study investigated the anti-obesity effects of collagen peptide derived from skate skin on lipid metabolism in high-fat diet (HFD)-fed mice. All C57BL6/J male mice were fed a HFD with 60% kcal fat except for mice in the normal group which were fed a chow diet. The collagen-fed groups received collagen peptide (1050 Da) orally (100, 200, or 300 mg/kg body weight per day) by gavage, whereas the normal and control groups were given water (n = 9 per group). The body weight gain and visceral adipose tissue weight were lower in the collagen-fed groups than in the control group (p < 0.05). Plasma and hepatic lipid levels were significantly reduced by downregulating the hepatic protein expression levels for fatty acid synthesis (sterol regulatory element binding protein-1 (SREBP-1), fatty acid synthase (FAS), and acetyl-CoA carboxylase (ACC)) and cholesterol synthesis (SREBP-2 and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR)) and upregulating those for β-oxidation (peroxisome proliferator-activated receptor alpha (PPAR-α) and carnitine palmitoyltransferase 1 (CPT1)) and synthesis of bile acid (cytochrome P450 family 7 subfamily A member 1 (CYP7A1)) (p < 0.05). In the collagen-fed groups, the hepatic protein expression level of phosphorylated 5' adenosine monophosphate-activated protein kinase (p-AMPK) and plasma adiponectin levels were higher, and the leptin level was lower (p < 0.05). Histological analysis revealed that collagen treatment suppressed hepatic lipid accumulation and reduced the lipid droplet size in the adipose tissue. These effects were increased in a dose-dependent manner. The findings indicated that skate collagen peptide has anti-obesity effects through suppression of fat accumulation and regulation of lipid metabolism.
Collapse
Affiliation(s)
- Minji Woo
- Department of Food Science and Nutrition and Kimchi Research Institute, Pusan National University, Busan 46241, Korea.
| | - Yeong Ok Song
- Department of Food Science and Nutrition and Kimchi Research Institute, Pusan National University, Busan 46241, Korea.
| | | | - Jeong Sook Noh
- Department of Food Science & Nutrition, Tongmyong University, Busan 48520, Korea.
| |
Collapse
|
33
|
Green CJ, Parry SA, Gunn PJ, Ceresa CDL, Rosqvist F, Piché ME, Hodson L. Studying non-alcoholic fatty liver disease: the ins and outs of in vivo, ex vivo and in vitro human models. Horm Mol Biol Clin Investig 2018; 41:/j/hmbci.ahead-of-print/hmbci-2018-0038/hmbci-2018-0038.xml. [PMID: 30098284 DOI: 10.1515/hmbci-2018-0038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/22/2018] [Indexed: 02/07/2023]
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing. Determining the pathogenesis and pathophysiology of human NAFLD will allow for evidence-based prevention strategies, and more targeted mechanistic investigations. Various in vivo, ex situ and in vitro models may be utilised to study NAFLD; but all come with their own specific caveats. Here, we review the human-based models and discuss their advantages and limitations in regards to studying the development and progression of NAFLD. Overall, in vivo whole-body human studies are advantageous in that they allow for investigation within the physiological setting, however, limited accessibility to the liver makes direct investigations challenging. Non-invasive imaging techniques are able to somewhat overcome this challenge, whilst the use of stable-isotope tracers enables mechanistic insight to be obtained. Recent technological advances (i.e. normothermic machine perfusion) have opened new opportunities to investigate whole-organ metabolism, thus ex situ livers can be investigated directly. Therefore, investigations that cannot be performed in vivo in humans have the potential to be undertaken. In vitro models offer the ability to perform investigations at a cellular level, aiding in elucidating the molecular mechanisms of NAFLD. However, a number of current models do not closely resemble the human condition and work is ongoing to optimise culturing parameters in order to recapitulate this. In summary, no single model currently provides insight into the development, pathophysiology and progression across the NAFLD spectrum, each experimental model has limitations, which need to be taken into consideration to ensure appropriate conclusions and extrapolation of findings are made.
Collapse
Affiliation(s)
- Charlotte J Green
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Siôn A Parry
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Pippa J Gunn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
| | - Carlo D L Ceresa
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fredrik Rosqvist
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Marie-Eve Piché
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Quebec Heart and Lung Institute, Laval University, Quebec, Canada
| | - Leanne Hodson
- University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, Churchill Hospital,Old Road Headington, Oxford OX3 7LE, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
34
|
Francois ME, Gilbertson NM, Eichner NZM, Heiston EM, Fabris C, Breton M, Mehaffey JH, Hassinger T, Hallowell PT, Malin SK. Combining Short-Term Interval Training with Caloric Restriction Improves ß-Cell Function in Obese Adults. Nutrients 2018; 10:nu10060717. [PMID: 29865281 PMCID: PMC6024769 DOI: 10.3390/nu10060717] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 05/24/2018] [Accepted: 05/30/2018] [Indexed: 02/07/2023] Open
Abstract
Although low-calorie diets (LCD) improve glucose regulation, it is unclear if interval exercise (INT) is additive. We examined the impact of an LCD versus LCD + INT training on ß-cell function in relation to glucose tolerance in obese adults. Twenty-six adults (Age: 46 ± 12 year; BMI 38 ± 6 kg/m²) were randomized to 2-week of LCD (~1200 kcal/day) or energy-matched LCD + INT (60 min/day alternating 3 min at 90 and 50% HRpeak). A 2 h 75 g oral glucose tolerance test (OGTT) was performed. Insulin secretion rates (ISR) were determined by deconvolution modeling to assess glucose-stimulated insulin secretion ([GSIS: ISR/glucose total area under the curve (tAUC)]) and ß-cell function (Disposition Index [DI: GSIS/IR]) relative to skeletal muscle (Matsuda Index), hepatic (HOMA-IR) and adipose (Adipose-IRfasting) insulin resistance (IR). LCD + INT, but not LCD alone, reduced glucose and total-phase ISR tAUC (Interactions: p = 0.04 and p = 0.05, respectively). Both interventions improved skeletal muscle IR by 16% (p = 0.04) and skeletal muscle and hepatic DI (Time: p < 0.05). Improved skeletal muscle DI was associated with lower glucose tAUC (r = -0.57, p < 0.01). Thus, LCD + INT improved glucose tolerance more than LCD in obese adults, and these findings relate to ß-cell function. These data support LCD + INT for preserving pancreatic function for type 2 diabetes prevention.
Collapse
Affiliation(s)
- Monique E Francois
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA.
| | - Nicole M Gilbertson
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA.
| | - Natalie Z M Eichner
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA.
| | - Emily M Heiston
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA.
| | - Chiara Fabris
- Center for Diabetes Technology, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA.
| | - Marc Breton
- Center for Diabetes Technology, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA.
| | - J Hunter Mehaffey
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA.
| | - Taryn Hassinger
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA.
| | - Peter T Hallowell
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA 22903, USA.
| | - Steven K Malin
- Department of Kinesiology, University of Virginia, Charlottesville, VA 22903, USA.
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA 22903, USA.
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|
35
|
Franch-Nadal J, Caballeria L, Mata-Cases M, Mauricio D, Giraldez-García C, Mancera J, Goday A, Mundet-Tudurí X, Regidor E. Fatty liver index is a predictor of incident diabetes in patients with prediabetes: The PREDAPS study. PLoS One 2018; 13:e0198327. [PMID: 29856820 PMCID: PMC5983533 DOI: 10.1371/journal.pone.0198327] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/17/2018] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES We evaluated the ability of the Fatty Liver Index (FLI), a surrogate marker of hepatic steatosis, to predict the development of type 2 diabetes (T2D) at 3 years follow-up in a Spanish cohort with prediabetes from a prospective observational study in primary care (PREDAPS). METHODS FLI was calculated at baseline for 1,142 adult subjects with prediabetes attending primary care centers, and classified into three categories: FLI <30 (no steatosis), FLI 30-60 (intermediate) and FLI ≥60 (hepatic steatosis). We estimated the incidence rate of T2D in each FLI category at 3 years of follow-up. The association between FLI and incident T2D was calculated using Cox regression models adjusted for age, sex, educational level, family history of diabetes, lifestyles, hypertension, lipid profile and transaminases. RESULTS The proportion of subjects with prediabetes and hepatic steatosis (FLI ≥60) at baseline was 55.7%. The incidence rate of T2D at 3 years follow-up was 1.3, 2.9 and 6.0 per 100 person-years for FLI<30, FLI 30->60 and FLI ≥60, respectively. The most significant variables increasing the risk of developing T2D were metabolic syndrome (hazard ratio [HR] = 3.02; 95% confidence interval [CI] = 2.14-4.26) and FLI ≥60 (HR = 4.52; 95%CI = 2.10-9.72). Moreover, FLI ≥60 was independently associated with T2D incidence: the HR was 4.97 (95% CI: 2.28-10.80) in the base regression model adjusted by sex, age and educational level, and 3.21 (95%CI: 1.45-7.09) in the fully adjusted model. CONCLUSIONS FLI may be considered an easy and valuable early indicator of high risk of incident T2D in patients with prediabetes attended in primary care, which could allow the adoption of effective measures needed to prevent and reduce the progression of the disease.
Collapse
Affiliation(s)
- Josep Franch-Nadal
- redGDPS Foundation, Madrid, Spain
- Unitat de Suport a la Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain
- Diabetes and Associated Metabolic Diseases Networking Biomedical Research Centre (CIBERDEM), Madrid, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Llorenç Caballeria
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), Madrid, Spain
- Unitat de Suport a la Recerca Barcelonès Nord, Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain
| | - Manel Mata-Cases
- redGDPS Foundation, Madrid, Spain
- Unitat de Suport a la Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain
- Diabetes and Associated Metabolic Diseases Networking Biomedical Research Centre (CIBERDEM), Madrid, Spain
| | - Didac Mauricio
- redGDPS Foundation, Madrid, Spain
- Unitat de Suport a la Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain
- Diabetes and Associated Metabolic Diseases Networking Biomedical Research Centre (CIBERDEM), Madrid, Spain
- Department of Endocrinology and Nutrition, Health Sciences Research Institute & University Hospital Germans Trias i Pujol, Badalona, Spain
| | - Carolina Giraldez-García
- redGDPS Foundation, Madrid, Spain
- Preventive Medicine Service, University Hospital Infanta Elena, Madrid, Spain
- Preventive Medicine, Public Health and History of Science Department, Complutense University of Madrid, Madrid, Spain
| | - José Mancera
- redGDPS Foundation, Madrid, Spain
- Health Center Ciudad Jardín, Málaga, Spain
| | - Albert Goday
- redGDPS Foundation, Madrid, Spain
- Endocrinology Service, Hospital del Mar, Barcelona, Spain
| | - Xavier Mundet-Tudurí
- redGDPS Foundation, Madrid, Spain
- Unitat de Suport a la Recerca Barcelona Ciutat, Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Barcelona, Spain
- Autonomous University of Barcelona, Bellaterra, Spain
| | - Enrique Regidor
- redGDPS Foundation, Madrid, Spain
- Preventive Medicine, Public Health and History of Science Department, Complutense University of Madrid, Madrid, Spain
- Epidemiology and Public Health Networking Biomedical Research Centre (CIBERESP), Madrid, Spain
- Health Research Institute, Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| |
Collapse
|
36
|
Chooi YC, Ding C, Chan Z, Choo J, Sadananthan SA, Michael N, Lee Y, Velan SS, Magkos F. Moderate Weight Loss Improves Body Composition and Metabolic Function in Metabolically Unhealthy Lean Subjects. Obesity (Silver Spring) 2018; 26:1000-1007. [PMID: 29676049 DOI: 10.1002/oby.22185] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 02/26/2018] [Accepted: 03/12/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Individuals who have "metabolically obese normal weight" (MONW) have an increased risk for cardiometabolic disease. Moderate weight loss has multiple benefits in people with obesity, but its effects in lean people are unknown. Thus, the effects of diet-induced 5% weight loss on body composition and metabolic function in MONW subjects were evaluated. METHODS Total body fat, visceral adipose tissue (VAT) and subcutaneous abdominal adipose tissue (SAT) volumes, intrahepatic and intramyocellular lipid contents, insulin sensitivity (hyperinsulinemic-euglycemic clamp), glucose tolerance, and postprandial insulin secretion and clearance rates (mixed meal with minimal modeling) were measured before and after 4.8% ± 0.5% weight loss in 11 MONW Asians (48 ± 3 years old, six men and five women, BMI 22.7 ± 0.4 kg/m2 ). RESULTS Weight loss decreased total fat mass by ∼9%, VAT and SAT volumes by ∼11% and ∼17%, respectively, and intrahepatic fat by ∼50% (all P < 0.05). Fasting plasma insulin, triglyceride, and total low- and high-density lipoprotein cholesterol concentrations were also reduced (P < 0.05). Insulin sensitivity indexes (M-value and M/I ratio) increased by 21% to 26% (both P < 0.05); β-cell responsivity and postprandial insulin secretion rate did not change, but insulin clearance rate increased by 16% (P < 0.05). CONCLUSIONS Diet-induced moderate weight loss improves body composition, lipid profile, and insulin sensitivity and thereby reduces cardiometabolic risk in MONW Asians.
Collapse
Affiliation(s)
- Yu Chung Chooi
- Clinical Nutrition Research Centre, Agency for Science, Technology and Research, National University Health System, Singapore
| | - Cherlyn Ding
- Clinical Nutrition Research Centre, Agency for Science, Technology and Research, National University Health System, Singapore
| | - Zhiling Chan
- Clinical Nutrition Research Centre, Agency for Science, Technology and Research, National University Health System, Singapore
| | - John Choo
- Clinical Nutrition Research Centre, Agency for Science, Technology and Research, National University Health System, Singapore
| | - Suresh Anand Sadananthan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Navin Michael
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
| | - Yijun Lee
- Clinical Nutrition Research Centre, Agency for Science, Technology and Research, National University Health System, Singapore
| | - S Sendhil Velan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore
- Laboratory of Molecular Imaging, Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore
| | - Faidon Magkos
- Clinical Nutrition Research Centre, Agency for Science, Technology and Research, National University Health System, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
37
|
Oh S, Han G, Kim B, Shoda J. Regular Exercise as a Secondary Practical Treatment for Nonalcoholic Fatty Liver Disease. EXERCISE MEDICINE 2018. [DOI: 10.26644/em.2018.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
38
|
Immonen H, Hannukainen JC, Kudomi N, Pihlajamäki J, Saunavaara V, Laine J, Salminen P, Lehtimäki T, Pham T, Iozzo P, Nuutila P. Increased Liver Fatty Acid Uptake Is Partly Reversed and Liver Fat Content Normalized After Bariatric Surgery. Diabetes Care 2018; 41:368-371. [PMID: 29158250 DOI: 10.2337/dc17-0738] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 11/01/2017] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Changes in liver fatty acid metabolism are important in understanding the mechanisms of diabetes remission and metabolic changes after bariatric surgery. RESEARCH DESIGN AND METHODS Liver fatty acid uptake (LFU), blood flow, and fat content (LFC) were measured in 25 obese subjects before bariatric surgery and 6 months after using positron emission tomography/computed tomography and MRS; 14 lean individuals served as the control subjects. RESULTS The increased LFU in obese subjects was associated with body adiposity. LFU was reduced postoperatively but was still high compared with the control subjects. LFC was normalized. Liver blood flow (per unit volume) was higher in obese subjects than in the control subjects at baseline and was further increased postoperatively; however, the total organ blood flow was unchanged as the liver volume decreased. CONCLUSIONS The findings suggest that in a postoperative state, intrahepatic fatty acids are not stored in the liver but are used for oxidation to provide energy. Changes in perfusion may contribute to improved liver metabolism postoperatively.
Collapse
Affiliation(s)
- Heidi Immonen
- Department of Endocrinology, Turku University Hospital, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland
| | | | | | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Virva Saunavaara
- Turku PET Centre, Turku University Hospital, Turku, Finland.,Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Jukka Laine
- Department of Pathology, Turku University Hospital, Turku, Finland
| | - Paulina Salminen
- Division of Digestive and Urology, Turku University Hospital, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, Tampere, Finland
| | - Tam Pham
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Patricia Iozzo
- Turku PET Centre, Turku University Hospital, Turku, Finland.,Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Pirjo Nuutila
- Department of Endocrinology, Turku University Hospital, Turku, Finland .,Turku PET Centre, Turku University Hospital, Turku, Finland
| |
Collapse
|
39
|
Soares AF, Duarte JMN, Gruetter R. Increased hepatic fatty acid polyunsaturation precedes ectopic lipid deposition in the liver in adaptation to high-fat diets in mice. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2017; 31:341-354. [PMID: 29027041 DOI: 10.1007/s10334-017-0654-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/26/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE We monitored hepatic lipid content (HLC) and fatty acid (FA) composition in the context of enhanced lipid handling induced by a metabolic high-fat diet (HFD) challenge and fasting. MATERIALS AND METHODS Mice received a control diet (10% of kilocalories from fat, N = 14) or an HFD (45% or 60% of kilocalories from fat, N = 10 and N = 16, respectively) for 26 weeks. A subset of five mice receiving an HFD (60% of kilocalories from fat) were switched to the control diet for the final 7 weeks. At nine time points, magnetic resonance spectroscopy was performed in vivo at 14.1 T, interleaved with glucose tolerance tests. RESULTS Glucose intolerance promptly developed with the HFD, followed by a progressive increase of fasting insulin level, simultaneously with that of HLC. These metabolic defects were normalized by dietary reversal. HFD feeding immediately increased polyunsaturation of hepatic FA, before lipid accumulation. Fasting-induced changes in hepatic lipids (increased HLC and FA polyunsaturation, decreased FA monounsaturation) in control-diet-fed mice were not completely reproduced in HFD-fed mice, not even after dietary reversal. CONCLUSION A similar adaptation of hepatic lipids to both fasting and an HFD suggests common mechanisms of lipid trafficking from adipose tissue to the liver. Altered hepatic lipid handling with fasting indicates imperfect metabolic recovery from HFD exposure.
Collapse
Affiliation(s)
- Ana Francisca Soares
- Laboratory for Functional and Metabolic Imaging, Swiss Federal Institute of Technology, Bâtiment CH, Station 6, 1015, Lausanne, Switzerland.
| | - João M N Duarte
- Laboratory for Functional and Metabolic Imaging, Swiss Federal Institute of Technology, Bâtiment CH, Station 6, 1015, Lausanne, Switzerland
| | - Rolf Gruetter
- Laboratory for Functional and Metabolic Imaging, Swiss Federal Institute of Technology, Bâtiment CH, Station 6, 1015, Lausanne, Switzerland.,Department of Radiology, University of Geneva, Geneva, Switzerland.,Department of Radiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
40
|
Soares AF, Paz-Montoya J, Lei H, Moniatte M, Gruetter R. Sexual dimorphism in hepatic lipids is associated with the evolution of metabolic status in mice. NMR IN BIOMEDICINE 2017; 30:e3761. [PMID: 28661066 DOI: 10.1002/nbm.3761] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/16/2017] [Accepted: 05/16/2017] [Indexed: 05/28/2023]
Abstract
Ectopic lipid accumulation in the liver is implicated in metabolic disease in an age- and sex-dependent manner. The role of hepatic lipids has been well established within the scope of metabolic insults in mice, but has been insufficiently characterized under standard housing conditions, where age-related metabolic alterations are known to occur. We studied a total of 10 male and 10 female mice longitudinally. At 3, 7 and 11 months of age, non-invasive 1 H-magnetic resonance spectroscopy (1 H-MRS) was used to monitor hepatic lipid content (HLC) and fatty acid composition in vivo, and glucose homeostasis was assessed with glucose and insulin challenges. At the end of the study, hepatic lipids were comprehensively characterized by nuclear magnetic resonance (NMR) and liquid chromatography-mass spectrometric analyses of liver tissue samples. In males, HLC increased from 1.4 ± 0.1% at 3 months to 2.9 ± 0.3% at 7 months (p < 0.01) and 2.7 ± 0.3% at 11 months (p < 0.05), in correlation with fasting insulin levels (p < 0.01, r = 0.51) and parameters from the insulin tolerance test (ITT; p < 0.001, r = -0.69 versus area under the curve; p < 0.01, r = -0.57 versus blood glucose drop at 1 h post-ITT; p < 0.01, r = 0.55 versus blood glucose at 3 h post-ITT). The metabolic performance of females remained the same throughout the study, and HLC was higher than that of males at 3 months (2.7 ± 0.2%, p < 0.01), but comparable at 7 months (2.2 ± 0.2%) and 11 months (2.2 ± 0.1%). Strong sexual dimorphism in bioactive lipid species, including diacylglycerols (higher in males, p < 0.0001), phosphatidylinositols (higher in females, p < 0.001) and omega-3 polyunsaturated fatty acids (higher in females, p < 0.01), was found to be in good correlation with metabolic scores at 11 months. Therefore, in mice housed under standard conditions, sex-specific composition of bioactive lipids is associated with metabolic protection in females, whose metabolic performance was independent of hepatic cytosolic lipid content.
Collapse
Affiliation(s)
- Ana Francisca Soares
- École Polytechnique Fédérale de Lausanne, Laboratory for Functional and Metabolic Imaging (LIFMET), Lausanne, VD, Switzerland
| | - Jonathan Paz-Montoya
- École Polytechnique Fédérale de Lausanne, Proteomics Core Facility (PCF), Lausanne, VD, Switzerland
| | - Hongxia Lei
- Center for Biomedical Imaging (CIBM), Lausanne VD, Switzerland and University of Geneva, Department of Radiology, Geneva, Switzerland
| | - Marc Moniatte
- École Polytechnique Fédérale de Lausanne, Proteomics Core Facility (PCF), Lausanne, VD, Switzerland
| | - Rolf Gruetter
- École Polytechnique Fédérale de Lausanne, Laboratory for Functional and Metabolic Imaging (LIFMET), Lausanne, VD, Switzerland
- Center for Biomedical Imaging (CIBM), Lausanne VD, Switzerland and University of Geneva, Department of Radiology, Geneva, Switzerland
| |
Collapse
|
41
|
Romero-Gómez M, Zelber-Sagi S, Trenell M. Treatment of NAFLD with diet, physical activity and exercise. J Hepatol 2017; 67:829-846. [PMID: 28545937 DOI: 10.1016/j.jhep.2017.05.016] [Citation(s) in RCA: 909] [Impact Index Per Article: 113.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/09/2017] [Accepted: 05/15/2017] [Indexed: 02/06/2023]
Abstract
Lifestyle intervention can be effective when treating non-alcoholic fatty liver diseases (NAFLD) patients. Weight loss decreases cardiovascular and diabetes risk and can also regress liver disease. Weight reductions of ⩾10% can induce a near universal non-alcoholic steatohepatitis resolution and fibrosis improvement by at least one stage. However, modest weight loss (>5%) can also produce important benefits on the components of the NAFLD activity score (NAS). Additionally, we need to explore the role of total calories and type of weight loss diet, micro- and macronutrients, evidence-based benefits of physical activity and exercise and finally support these modifications through established behavioural change models and techniques for long-term maintenance of lifestyle modifications. Following a Mediterranean diet can reduce liver fat even without weight loss and is the most recommended dietary pattern for NAFLD. The Mediterranean diet is characterised by reduced carbohydrate intake, especially sugars and refined carbohydrates (40% of the calories vs. 50-60% in a typical low fat diet), and increased monounsaturated and omega-3 fatty acid intake (40% of the calories as fat vs. up-to 30% in a typical low fat diet). Both TV sitting (a reliable marker of overall sedentary behaviour) and physical activity are associated with cardio-metabolic health, NAFLD and overall mortality. A 'triple hit behavioural phenotype' of: i) sedentary behaviour, ii) low physical activity, and iii) poor diet have been defined. Clinical evidence strongly supports the role of lifestyle modification as a primary therapy for the management of NAFLD and NASH. This should be accompanied by the implementation of strategies to avoid relapse and weight regain.
Collapse
Affiliation(s)
- Manuel Romero-Gómez
- Mac.Ro UCM IC Digestive Diseases and Ciberehd, University Hospital Virgen del Rocio, Institute of Biomedicine of Seville, University of Seville, Sevilla, Spain.
| | - Shira Zelber-Sagi
- Department Gastroenterology, Tel-Aviv Medical Center, Tel-Aviv, Israel; School of Public Health, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Michael Trenell
- NIHR Innovation Observatory, Newcastle University, Newcastle, UK
| |
Collapse
|
42
|
Peters HPF, Schrauwen P, Verhoef P, Byrne CD, Mela DJ, Pfeiffer AFH, Risérus U, Rosendaal FR, Schrauwen-Hinderling V. Liver fat: a relevant target for dietary intervention? Summary of a Unilever workshop. J Nutr Sci 2017; 6:e15. [PMID: 28630692 PMCID: PMC5468740 DOI: 10.1017/jns.2017.13] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
Currently it is estimated that about 1 billion people globally have non-alcoholic fatty liver disease (NAFLD), a condition in which liver fat exceeds 5 % of liver weight in the absence of significant alcohol intake. Due to the central role of the liver in metabolism, the prevalence of NAFLD is increasing in parallel with the prevalence of obesity, insulin resistance and other risk factors of metabolic diseases. However, the contribution of liver fat to the risk of type 2 diabetes mellitus and CVD, relative to other ectopic fat depots and to other risk markers, is unclear. Various studies have suggested that the accumulation of liver fat can be reduced or prevented via dietary changes. However, the amount of liver fat reduction that would be physiologically relevant, and the timeframes and dose-effect relationships for achieving this through different diet-based approaches, are unclear. Also, it is still uncertain whether the changes in liver fat per se or the associated metabolic changes are relevant. Furthermore, the methods available to measure liver fat, or even individual fatty acids, differ in sensitivity and reliability. The present report summarises key messages of presentations from different experts and related discussions from a workshop intended to capture current views and research gaps relating to the points above.
Collapse
Affiliation(s)
- Harry P. F. Peters
- Unilever R&D Vlaardingen, Olivier van Noortlaan 120, Vlaardingen, The Netherlands
| | - Patrick Schrauwen
- Department of Human Biology, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Petra Verhoef
- Unilever R&D Vlaardingen, Olivier van Noortlaan 120, Vlaardingen, The Netherlands
| | - Christopher D. Byrne
- Nutrition and Metabolism, Faculty of Medicine, University of Southampton & Southampton National Institute for Health Research Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - David J. Mela
- Unilever R&D Vlaardingen, Olivier van Noortlaan 120, Vlaardingen, The Netherlands
| | - Andreas F. H. Pfeiffer
- Department of Endocrinology, Diabetes and Nutrition, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin
- Department of Clinical Nutrition, German Institute of Human Nutrition, Potsdam and German Center for Diabetes Research, DZD, Neuherberg, Germany
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism Unit, Uppsala University, Sweden
| | - Frits R. Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vera Schrauwen-Hinderling
- Department of Human Biology, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Radiology, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
43
|
Bunt JC, Blackstone R, Thearle MS, Vinales KL, Votruba S, Krakoff J. Changes in glycemia, insulin and gut hormone responses to a slowly ingested solid low-carbohydrate mixed meal after laparoscopic gastric bypass or band surgery. Int J Obes (Lond) 2017; 41:706-713. [PMID: 28119531 DOI: 10.1038/ijo.2017.22] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/22/2016] [Accepted: 01/11/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate early changes in glycemia, insulin physiology and gut hormone responses to an easily tolerated and slowly ingested solid, low-carbohydrate mixed meal test (MMT) following laparoscopic adjustable gastric banding (LAGB) or Roux-en-Y gastric bypass (RYGB) surgery. SUBJECTS/METHODS This was a prospective non-randomized study. Plasma glucose, insulin and c-peptide (to estimate hepatic insulin extraction; %HIE), incretins (GIP, aGLP-1) and pancreatic polypeptide (PP) responses to the MMT were measured at 4-8 weeks before and after surgery in obese, metabolically healthy patients (RYGB=10F or LAGB =7F/1M). Supplementary clamp data on basal endogenous glucose production (EGP) and peripheral insulin action (Rd=rate of glucose disposal) and metabolic clearance rates of insulin (MCR-INS) were available in five of the RYGB patients. Repeated measures were appropriately accounted for in the analyses. RESULTS Following LAGB surgery, C-peptide and insulin MMT profiles (P=0.004 and P=0.0005, respectively) were lower with no change in %HIE (P=0.98). In contrast, in RYGB subjects, both fasting glucose and insulin (Δ=-0.66 mmol l-1, P⩽0.05 and Δ=-44.4 pmol l-1, P⩽0.05, respectively) decreased, and MMT glucose (P<0.0001) and insulin (P=0.001) but not c-peptide (P= 0.69) decreased. Estimated %HIE increased at fasting (Δ=8.4%, P⩽0.05) and during MMT (P=0.0005). Early (0-20 min) prandial glucose (0.27±0.26 versus 0.006±0.21 mmol l-1, P⩽0.05) and insulin (63(48, 66) versus 18(12, 24) pmol l-1, P⩽0.05) responses increased after RYGB. RYGB altered the trajectory of prandial aGLP-1 responses (treatment × trajectory P=0.02), and PP was lower (P<0.0001). Clamp data in a subset of RYGB patients showed early improvement in basal EGP (P=0.001), and MCR-INS (P=0.015). CONCLUSION RYGB results in distinctly different changes in plasma glucose, insulin and gut hormone response patterns to a solid, slowly ingested low-carbohydrate MMT versus LAGB. Altered nutrient delivery, along with indirect evidence for changes in hepatic and peripheral insulin physiology, are consistent with the greater early improvement in glycemia observed after RYGB versus LAGB surgery.
Collapse
Affiliation(s)
- J C Bunt
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, NIDDK, NIH, DHHS, Phoenix, AZ, USA
| | - R Blackstone
- Scottsdale Healthcare Bariatric Center, Scottsdale, AZ, USA.,Department of Surgery, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - M S Thearle
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, NIDDK, NIH, DHHS, Phoenix, AZ, USA
| | - K L Vinales
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, NIDDK, NIH, DHHS, Phoenix, AZ, USA
| | - S Votruba
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, NIDDK, NIH, DHHS, Phoenix, AZ, USA
| | - J Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, NIDDK, NIH, DHHS, Phoenix, AZ, USA
| |
Collapse
|
44
|
Rubinow KB. Estrogens and Body Weight Regulation in Men. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:285-313. [PMID: 29224100 DOI: 10.1007/978-3-319-70178-3_14] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Our understanding of the metabolic roles of sex steroids in men has evolved substantially over recent decades. Whereas testosterone once was believed to contribute to metabolic risk in men, the importance of adequate androgen exposure for the maintenance of metabolic health has been demonstrated unequivocally. A growing body of evidence now also supports a critical role for estrogens in metabolic regulation in men. Recent data from clinical intervention studies indicate that estradiol may be a stronger determinant of adiposity than testosterone in men, and even short-term estradiol deprivation contributes to fat mass accrual. The following chapter will outline findings to date regarding the mechanisms, whereby estrogens contribute to the regulation of body weight and adiposity in men. It will present emergent clinical data as well as preclinical findings that reveal mechanistic insights into estrogen-mediated regulation of body composition. Findings in both males and females will be reviewed, to draw comparisons and to highlight knowledge gaps regarding estrogen action specifically in males. Finally, the clinical relevance of estrogen exposure in men will be discussed, particularly in the context of a rising global prevalence of obesity and expanding clinical use of sex steroid-based therapies in men.
Collapse
Affiliation(s)
- Katya B Rubinow
- Division of Metabolism, Endocrinology, and Nutrition Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
45
|
Ekholm E, Hansen L, Johnsson E, Iqbal N, Carlsson B, Chen H, Hirshberg B. COMBINED TREATMENT WITH SAXAGLIPTIN PLUS DAPAGLIFLOZIN REDUCES INSULIN LEVELS BY INCREASED INSULIN CLEARANCE AND IMPROVES β-CELL FUNCTION. Endocr Pract 2016; 23:258-265. [PMID: 27849380 DOI: 10.4158/ep161323.or] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To determine if reduction in serum insulin with dapagliflozin plus saxagliptin or dapagliflozin add-on to metformin contributed to increased insulin clearance and to assess the effects of these treatments on β-cell function. METHODS Patients (glycated hemoglobin, 8 to 12%; 64 to 108 mmol/mol) were randomized to 24-week, double-blind treatment with saxagliptin 5 mg/day plus dapagliflozin 10 mg/day (n = 179), saxagliptin 5 mg/day plus placebo (n = 176), or dapagliflozin 10 mg/day plus placebo (n = 179) added to metformin. C-peptide to insulin ratio was used as an index of insulin clearance during a meal tolerance test, and β-cell function was evaluated by Homeostasis Model Assessment 2. RESULTS At 24 weeks, compared with baseline, saxagliptin + dapagliflozin and saxagliptin + placebo increased mean (95% confidence interval [CI]) C-peptide area under the curve (AUC0-180 min) (40.2 [9.2 to 71.3] ng/mL and 95.4 [63.4 to 127.4] ng/mL, respectively); no change was noted with dapagliflozin + placebo (14.5 [-17.6 to 46.8] ng/mL). Insulin AUC was reduced from baseline with saxagliptin + dapagliflozin (-1,120.4 [-1,633.9 to -606.9] μU/mL) and dapagliflozin + placebo (-1,018.6 [-1550.5 to -486.8] μU/mL) but increased with saxagliptin + placebo (661.2 [131.1 to 1,191.3] μU/mL). C-peptide to insulin ratio did not change versus baseline with saxagliptin + placebo but increased after saxagliptin + dapagliflozin and dapagliflozin + placebo, largely due to decreased insulin AUC with dapagliflozin. All treatments improved β-cell function (mean change [95% CI] from baseline, saxagliptin+dapagliflozin: 20.6% [16.5% to 24.8%]; dapagliflozin + placebo: 17.0% [12.7% to 21.4%]; saxagliptin + placebo: 11.0% [6.6% to 15.5%]). CONCLUSION Increased C-peptide to insulin ratio with saxagliptin + dapagliflozin and dapagliflozin + placebo add-on to metformin compared with saxagliptin + placebo add-on to metformin suggests that dapagliflozin increases insulin clearance and may contribute to lower circulating insulin. All treatments improved β-cell function, with the greatest improvements with saxagliptin + dapagliflozin and dapagliflozin + placebo. ABBREVIATIONS A1c = glycated hemoglobin AUC0-180 min = area under the curve from 0 to 180 minutes HOMA-2β = homeostasis model assessment-2 β-cell function SGLT-2 = sodium-glucose cotransporter-2 T2D = type 2 diabetes.
Collapse
|
46
|
Changes of insulin sensitivity and secretion after bariatric/metabolic surgery. Surg Obes Relat Dis 2016; 12:1199-205. [PMID: 27568471 DOI: 10.1016/j.soard.2016.05.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 05/11/2016] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes (T2D) is classically characterized by failure of pancreatic β-cell function and insulin secretion to compensate for a prevailing level of insulin resistance, typically associated with visceral obesity. Although this is usually a chronic, progressive disease in which delay of end-organ complications is the primary therapeutic goal for medical and behavioral approaches, several types of bariatric surgery, especially those that include intestinal bypass components, exert powerful antidiabetes effects to yield remission of T2D in most cases. It has become increasingly clear that in addition to the known benefits of acute caloric restriction and chronic weight loss to ameliorate T2D, bariatric/metabolic operations also engage a variety of weight-independent mechanisms to improve glucose homeostasis, enhancing insulin sensitivity and secretion to varying degrees depending on the specific operation. In this paper, we review the effects of Roux-en-Y gastric bypass, biliopancreatic diversion, and vertical sleeve gastrectomy on the primary determinants of glucose homeostasis: insulin sensitivity, insulin secretion, and, to the lesser extent that it is known, insulin-independent glucose disposal. A full understanding of these effects should help optimize surgical and device-based designs to provide maximal antidiabetes impact, and it holds the promise to identify targets for possible novel diabetes pharmacotherapeutics. These insights also contribute to the conceptual rationale for use of bariatric operations as "metabolic surgery," employed primarily to treat T2D, including among patients not obese enough to qualify for surgery based on traditional criteria related to high body mass index.
Collapse
|
47
|
Vogt LJ, Steveling A, Meffert PJ, Kromrey ML, Kessler R, Hosten N, Krüger J, Gärtner S, Aghdassi AA, Mayerle J, Lerch MM, Kühn JP. Magnetic Resonance Imaging of Changes in Abdominal Compartments in Obese Diabetics during a Low-Calorie Weight-Loss Program. PLoS One 2016; 11:e0153595. [PMID: 27110719 PMCID: PMC4844151 DOI: 10.1371/journal.pone.0153595] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 03/31/2016] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES To investigate changes in the fat content of abdominal compartments and muscle area during weight loss using confounder-adjusted chemical-shift-encoded magnetic resonance imaging (MRI) in overweight diabetics. METHODS Twenty-nine obese diabetics (10/19 men/women, median age: 59.0 years, median body mass index (BMI): 34.0 kg/m2) prospectively joined a standardized 15-week weight-loss program (six weeks of formula diet exclusively, followed by reintroduction of regular food with gradually increasing energy content over nine weeks) over 15 weeks. All subjects underwent a standardized MRI protocol including a confounder-adjusted chemical-shift-encoded MR sequence with water/fat separation before the program as well at the end of the six weeks of formula diet and at the end of the program at 15 weeks. Fat fractions of abdominal organs and vertebral bone marrow as well as volumes of visceral and subcutaneous fat were determined. Furthermore, muscle area was evaluated using the L4/L5 method. Data were compared using the Wilcoxon signed-rank test for paired samples. RESULTS Median BMI decreased significantly from 34.0 kg/m2 to 29.9 kg/m2 (p < 0.001) at 15 weeks. Liver fat content was normalized (14.2% to 4.1%, p < 0.001) and vertebral bone marrow fat (57.5% to 53.6%, p = 0.018) decreased significantly throughout the program, while fat content of pancreas (9.0%), spleen (0.0%), and psoas muscle (0.0%) did not (p > 0.15). Visceral fat volume (3.2 L to 1.6 L, p < 0.001) and subcutaneous fat diameter (3.0 cm to 2.2 cm, p < 0.001) also decreased significantly. Muscle area declined by 6.8% from 243.9 cm2 to 226.8 cm2. CONCLUSION MRI allows noninvasive monitoring of changes in abdominal compartments during weight loss. In overweight diabetics, weight loss leads to fat reduction in abdominal compartments, such as visceral fat, as well as liver fat and vertebral bone marrow fat while pancreas fat remains unchanged.
Collapse
Affiliation(s)
- Lena J. Vogt
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Antje Steveling
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Peter J. Meffert
- Institute of Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Marie-Luise Kromrey
- Department of Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Rebecca Kessler
- Department of Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Norbert Hosten
- Department of Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Janine Krüger
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Simone Gärtner
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Ali A. Aghdassi
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Julia Mayerle
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Markus M. Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Jens-Peter Kühn
- Department of Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
- * E-mail:
| |
Collapse
|
48
|
Gut Microbiota and Lifestyle Interventions in NAFLD. Int J Mol Sci 2016; 17:447. [PMID: 27023533 PMCID: PMC4848903 DOI: 10.3390/ijms17040447] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023] Open
Abstract
The human digestive system harbors a diverse and complex community of microorganisms that work in a symbiotic fashion with the host, contributing to metabolism, immune response and intestinal architecture. However, disruption of a stable and diverse community, termed "dysbiosis", has been shown to have a profound impact upon health and disease. Emerging data demonstrate dysbiosis of the gut microbiota to be linked with non-alcoholic fatty liver disease (NAFLD). Although the exact mechanism(s) remain unknown, inflammation, damage to the intestinal membrane, and translocation of bacteria have all been suggested. Lifestyle intervention is undoubtedly effective at improving NAFLD, however, not all patients respond to these in the same manner. Furthermore, studies investigating the effects of lifestyle interventions on the gut microbiota in NAFLD patients are lacking. A deeper understanding of how different aspects of lifestyle (diet/nutrition/exercise) affect the host-microbiome interaction may allow for a more tailored approach to lifestyle intervention. With gut microbiota representing a key element of personalized medicine and nutrition, we review the effects of lifestyle interventions (diet and physical activity/exercise) on gut microbiota and how this impacts upon NAFLD prognosis.
Collapse
|
49
|
Hallsworth K, Avery L, Trenell MI. Targeting Lifestyle Behavior Change in Adults with NAFLD During a 20-min Consultation: Summary of the Dietary and Exercise Literature. Curr Gastroenterol Rep 2016; 18:11. [PMID: 26908279 PMCID: PMC4764638 DOI: 10.1007/s11894-016-0485-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is largely linked to poor diet, lack of physical activity/exercise, and being overweight. In the absence of approved pharmaceutical agents, lifestyle modification, encompassing dietary change and increased physical activity/exercise to initiate weight loss, is the recommended therapy for NAFLD. Despite this, the use of lifestyle therapy within clinical settings is lacking with limited guidance available about what it should involve, how it should be delivered, and whether it can be feasibly delivered as part of standard care. This paper highlights the evidence for the use of lifestyle modification in NAFLD. While there is evidence to support use of behavioral strategies to support lifestyle behavior change in other clinical populations, these are yet to be assessed in people with NAFLD. However, there is sufficient evidence to suggest that behavioral intervention targeting diet and physical activity to promote weight loss in general is effective and a number of practical strategies are presented on how this could be achieved.
Collapse
Affiliation(s)
- Kate Hallsworth
- Institute of Cellular Medicine, Newcastle University, MoveLab, 4th Floor William Leech Building, Newcastle upon Tyne, NE2 4HH, UK.
| | - Leah Avery
- Institute of Cellular Medicine, Newcastle University, MoveLab, 4th Floor William Leech Building, Newcastle upon Tyne, NE2 4HH, UK
| | - Michael I Trenell
- Institute of Cellular Medicine, Newcastle University, MoveLab, 4th Floor William Leech Building, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
50
|
Iozzo P. Metabolic imaging in obesity: underlying mechanisms and consequences in the whole body. Ann N Y Acad Sci 2015; 1353:21-40. [PMID: 26335600 DOI: 10.1111/nyas.12880] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Obesity is a phenotype resulting from a series of causative factors with a variable risk of complications. Etiologic diversity requires personalized prevention and treatment. Imaging procedures offer the potential to investigate the interplay between organs and pathways underlying energy intake and consumption in an integrated manner, and may open the perspective to classify and treat obesity according to causative mechanisms. This review illustrates the contribution provided by imaging studies to the understanding of human obesity, starting with the regulation of food intake and intestinal metabolism, followed by the role of adipose tissue in storing, releasing, and utilizing substrates, including the interconversion of white and brown fat, and concluding with the examination of imaging risk indicators related to complications, including type 2 diabetes, liver pathologies, cardiac and kidney diseases, and sleep disorders. The imaging modalities include (1) positron emission tomography to quantify organ-specific perfusion and substrate metabolism; (2) computed tomography to assess tissue density as an indicator of fat content and browning/ whitening; (3) ultrasounds to examine liver steatosis, stiffness, and inflammation; and (4) magnetic resonance techniques to assess blood oxygenation levels in the brain, liver stiffness, and metabolite contents (triglycerides, fatty acids, glucose, phosphocreatine, ATP, and acetylcarnitine) in a variety of organs.
Collapse
Affiliation(s)
- Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy.,The Turku PET Centre, University of Turku, Turku, Finland
| |
Collapse
|