1
|
Del Carmen Fernández-Fígares Jiménez M. Plant foods, healthy plant-based diets, and type 2 diabetes: a review of the evidence. Nutr Rev 2024; 82:929-948. [PMID: 37550262 DOI: 10.1093/nutrit/nuad099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
Type 2 diabetes (T2D) is a metabolic chronic disease in which insulin resistance and insufficient insulin production lead to elevated blood glucose levels. The prevalence of T2D is growing worldwide, mainly due to obesity and the adoption of Western diets. Replacing animal foods with healthy plant foods is associated with a lower risk of T2D in prospective studies. In randomized controlled trials, the consumption of healthy plant foods in place of animal foods led to cardiometabolic improvements in patients with T2D or who were at high risk of the disease. Dietary patterns that limit or exclude animal foods and focus on healthy plant foods (eg, fruits, vegetables, whole grains, nuts, legumes), known as healthy, plant-based diets, are consistently associated with a lower risk of T2D in cohort studies. The aim of this review is to examine the differential effects of plant foods and animal foods on T2D risk and to describe the existing literature about the role of healthy, plant-based diets, particularly healthy vegan diets, in T2D prevention and management. The evidence from cohort studies and randomized controlled trials will be reported, in addition to the potential biological mechanisms that seem to be involved.
Collapse
|
2
|
Alqallaf J, Orange ST, Matu J, Griffiths A, Johnson K, Stavropoulos-Kalinoglou A, Holliday A, Wilson O. The Effect of High-Fat Diet on Intramyocellular Lipid Content in Healthy Adults: A Systematic Review, Meta-Analysis, and Meta-Regression. J Nutr 2024; 154:1087-1100. [PMID: 38417551 PMCID: PMC11007750 DOI: 10.1016/j.tjnut.2024.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/01/2024] Open
Abstract
Fatty acids are stored within the muscle as intramyocellular lipids (IMCL). Some, but not all, studies indicate that following a high-fat diet (HFD), IMCL may accumulate and affect insulin sensitivity. This systematic review and meta-analysis aimed to quantify the effects of an HFD on IMCL. It also explored the potential modifying effects of HFD fat content and duration, IMCL measurement technique, physical activity status, and the associations of IMCL with insulin sensitivity. Five databases were systematically searched for studies that examined the effect of ≥3 d of HFD (>35% daily energy intake from fat) on IMCL content in healthy individuals. Meta-regressions were used to investigate associations of the HFD total fat content, duration, physical activity status, IMCL measurement technique, and insulin sensitivity with IMCL responses. Changes in IMCL content and insulin sensitivity (assessed by hyperinsulinemic-euglycemic clamp) are presented as standardized mean difference (SMD) using a random effects model with 95% confidence intervals (95% CIs). Nineteen studies were included in the systematic review and 16 in the meta-analysis. IMCL content increased following HFD (SMD = 0.63; 95% CI: 0.31, 0.94, P = 0.001). IMCL accumulation was not influenced by total fat content (P = 0.832) or duration (P = 0.844) of HFD, physical activity status (P = 0.192), or by the IMCL measurement technique (P > 0.05). Insulin sensitivity decreased following HFD (SMD = -0.34; 95% CI: -0.52, -0.16; P = 0.003), but this was not related to the increase in IMCL content following HFD (P = 0.233). Consumption of an HFD (>35% daily energy intake from fat) for ≥3 d significantly increases IMCL content in healthy individuals regardless of HFD total fat content and duration of physical activity status. All IMCL measurement techniques detected the increased IMCL content following HFD. The dissociation between changes in IMCL and insulin sensitivity suggests that other factors may drive HFD-induced impairments in insulin sensitivity in healthy individuals. This trial was registered at PROSPERO as CRD42021257984.
Collapse
Affiliation(s)
- Jasem Alqallaf
- Carnegie School of Sport, Leeds Beckett University, United Kingdom
| | - Samuel T Orange
- School of Biomedical, Nutritional, and Sport Sciences, Faculty of Medical Sciences, Newcastle University, United Kingdom; Newcastle University Centre for Cancer, Newcastle University, United Kingdom
| | - Jamie Matu
- School of Health, Leeds Beckett University, United Kingdom
| | - Alex Griffiths
- School of Health, Leeds Beckett University, United Kingdom
| | - Kelsie Johnson
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, United Kingdom
| | | | - Adrian Holliday
- School of Biomedical, Nutritional, and Sport Sciences, Faculty of Medical Sciences, Newcastle University, United Kingdom
| | - Oliver Wilson
- Carnegie School of Sport, Leeds Beckett University, United Kingdom.
| |
Collapse
|
3
|
Buitinga M, Veeraiah P, Haans F, Schrauwen-Hinderling VB. Ectopic lipid deposition in muscle and liver, quantified by proton magnetic resonance spectroscopy. Obesity (Silver Spring) 2023; 31:2447-2459. [PMID: 37667838 DOI: 10.1002/oby.23865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 09/06/2023]
Abstract
Advances in the development of noninvasive imaging techniques have spurred investigations into ectopic lipid deposition in the liver and muscle and its implications in the development of metabolic diseases such as type 2 diabetes. Computed tomography and ultrasound have been applied in the past, though magnetic resonance-based methods are currently considered the gold standard as they allow more accurate quantitative detection of ectopic lipid stores. This review focuses on methodological considerations of magnetic resonance-based methods to image hepatic and muscle fat fractions, and it emphasizes anatomical and morphological aspects and how these may influence data acquisition, analysis, and interpretation.
Collapse
Affiliation(s)
- Mijke Buitinga
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Nutrition and Movement Sciences (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Pandichelvam Veeraiah
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- Scannexus (Ultra-High Field Imaging Center), Maastricht, The Netherlands
- Faculty of Health Medicine and Life Sciences (FHML), Maastricht, The Netherlands
| | - Florian Haans
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Vera B Schrauwen-Hinderling
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Nutrition and Movement Sciences (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Institute for Clinical Diabetology, German Diabetes Center and Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
4
|
Khan I, Kumar R, Prasad M, Srivastav RK, Vishwakarma VK, Akhtar J. Co-Adjuvancy of Solasodine & CoQ10 Against High Fat Diet-Induced Insulin Resistance Rats Via Modulating IRS-I and PPAR-γ Proteins Expression. Drug Res (Stuttg) 2022; 72:327-335. [PMID: 35724671 DOI: 10.1055/a-1806-1366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Insulin resistance (IR) is a condition in which target cells become insensitive to normal insulin concentrations in order to deliver glucose. The goal of this study was to see if solasodine combined with coenzyme Q10 could help rats with insulin resistance caused by a high-fat diet (HFD) by regulating the expression of IRS-I and PPAR-γ proteins.One of the six groups (n=6) got a conventional diet for 16 weeks as a control (normal), the HFD was given to the other five groups for 16 weeks, which further classified as-one group as HFD control while others treated with pioglitazone (10 mg/kg), coenzyme Q10 (50 mg/kg), solasodine (50 mg/kg) and combination of solasodine and coenzyme Q10i.e. SDQ10 (total 50 mg/kg) for the last 4 weeks orally once daily. Blood and tissue samples were collected by the end of study period for the biochemical and histological studies. As a result, HFD fed rats exhibited a significant increase in food and energy intake, body mass index, kidney and pancreas weight, fasting glucose, glycosylated haemoglobin, insulin level, liver enzyme ALT and AST and decrease antioxidant activity of superoxide dismutase and catalase. HFD received animals also produced a lower level of p-IRS1 and PPAR-y protein expression in western blot analysis. SDQ10 in combination successfully restored the above-mentioned complexity of insulin resistance caused by aHFD. Besides, increasesthe antioxidant activity of superoxide dismutase and catalase and normalized the architecture of kidney, pancreas and adipose tissue as well astreatment with SDQ10 raised the level of p-IRS1 and PPAR-y protein in liver tissue. As a result, supplementing with solasodine and coenzyme Q10 reversed the effect of the HFD on p-IRS1 and PPAR-y protein in liver tissue while also alleviating insulin resistance symptoms.
Collapse
Affiliation(s)
- Irfan Khan
- Faculty of Pharmacy, Integral University, Lucknow, U.P, India
| | - Rajesh Kumar
- Faculty of Pharmacy, Kamla Nehru Institute of Management & Technology, Sultanpur, U.P., India
| | - Mahesh Prasad
- Faculty of Pharmacy, Kamla Nehru Institute of Management & Technology, Sultanpur, U.P., India.,Faculty of Pharmacy, Integral University, Lucknow, U.P, India
| | - Ritesh Kumar Srivastav
- Faculty of Pharmacy, Kamla Nehru Institute of Management & Technology, Sultanpur, U.P., India
| | | | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow, U.P, India
| |
Collapse
|
5
|
Ben Salem M, Affes H, Dhouibi R, Charfi S, Turki M, Hammami S, Ayedi F, Sahnoun Z, Zeghal KM, Ksouda K. Effect of Artichoke ( cynara scolymus) on cardiac markers, lipid profile and antioxidants levels in tissue of HFD-induced obesity. Arch Physiol Biochem 2022; 128:184-194. [PMID: 31564131 DOI: 10.1080/13813455.2019.1670213] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Obesity plays a pivotal role in the insulin resistance disease, which is related to hypertension, hyperlipidemia, type 2 diabetes mellitus, and an increased risk of cardiovascular disease. The purpose of the present study was done to evaluate the effect of artichoke leaves extract (ALE) in the high-fat diet (HFD)-induced cellular obesity and cardiac damage in Wistar rats. Body and organ weights, serum lipid profile, cardiac markers, and antioxidants enzymes were measured. Oral administration of ALE at two doses 200 and 400 mg/kg for a period of 60 days showed a significant decrease in body and organ weights, serum total cholesterol, triglycerides, LDH, ALT accompanied by decreasing in oxidative stress biomarker (MDA, and AOPP) and increasing antioxidant enzymes (SOD, CAT, and GPx) levels as compared to HFD groups. The histological findings showed a cardioprotective effect of ALE. These findings suggest that ALE exert anti-oxidant cardiac effects in HFD- induced obese rats.
Collapse
Affiliation(s)
- Maryem Ben Salem
- Laboratory of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Hanen Affes
- Laboratory of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Raouia Dhouibi
- Laboratory of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Slim Charfi
- Laboratory of Anatomopathology, CHU Habib Bourguiba, University of Sfax, Sfax, Tunisia
| | - Mouna Turki
- Biochemistry Laboratory, CHU Habib Bourguiba, University of Sfax, Sfax, Tunisia
| | - Serria Hammami
- Laboratory of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Fatma Ayedi
- Biochemistry Laboratory, CHU Habib Bourguiba, University of Sfax, Sfax, Tunisia
| | - Zouheir Sahnoun
- Laboratory of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Khaled Mounir Zeghal
- Laboratory of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Kamilia Ksouda
- Laboratory of Pharmacology, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| |
Collapse
|
6
|
Krššák M, Lindeboom L, Schrauwen‐Hinderling V, Szczepaniak LS, Derave W, Lundbom J, Befroy D, Schick F, Machann J, Kreis R, Boesch C. Proton magnetic resonance spectroscopy in skeletal muscle: Experts' consensus recommendations. NMR IN BIOMEDICINE 2021; 34:e4266. [PMID: 32022964 PMCID: PMC8244035 DOI: 10.1002/nbm.4266] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 12/21/2019] [Accepted: 01/15/2020] [Indexed: 05/02/2023]
Abstract
1 H-MR spectroscopy of skeletal muscle provides insight into metabolism that is not available noninvasively by other methods. The recommendations given in this article are intended to guide those who have basic experience in general MRS to the special application of 1 H-MRS in skeletal muscle. The highly organized structure of skeletal muscle leads to effects that change spectral features far beyond simple peak heights, depending on the type and orientation of the muscle. Specific recommendations are given for the acquisition of three particular metabolites (intramyocellular lipids, carnosine and acetylcarnitine) and for preconditioning of experiments and instructions to study volunteers.
Collapse
Affiliation(s)
- Martin Krššák
- Division of Endocrinology and Metabolism, Department of Internal Medicine III & High Field MR Centre, Department of Biomedical Imaging and Image guided TherapyMedical University of ViennaViennaAustria
| | - Lucas Lindeboom
- Department of Radiology and Nuclear Medicine and Department of Nutrition and Movement ScienceMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Vera Schrauwen‐Hinderling
- Department of Radiology and Nuclear Medicine and Department of Nutrition and Movement ScienceMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Lidia S. Szczepaniak
- Biomedical Research Consulting in Magnetic Resonance SpectroscopyAlbuquerqueNew Mexico
| | - Wim Derave
- Department of Movement and Sports SciencesGhent UniversityGhentBelgium
| | - Jesper Lundbom
- Department of Diagnostics and TherapeuticsUniversity of HelsinkiHelsinkiFinland
| | | | - Fritz Schick
- Section on Experimental Radiology, Department of Diagnostic and Interventional RadiologyUniversity Hospital TübingenTübingenGermany
| | - Jürgen Machann
- Section on Experimental Radiology, Department of Diagnostic and Interventional RadiologyUniversity Hospital TübingenTübingenGermany
- Institute for Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of TübingenTübingenGermany
- German Center for Diabetes Research (DZD)TübingenGermany
| | - Roland Kreis
- Departments of Radiology and Biomedical ResearchUniversity and InselspitalBernSwitzerland
| | - Chris Boesch
- Departments of Radiology and Biomedical ResearchUniversity and InselspitalBernSwitzerland
| |
Collapse
|
7
|
Molecular adaptation in adipose tissue in response to overfeeding with a high-fat diet under sedentary conditions in South Asian and Caucasian men. Br J Nutr 2019; 122:241-251. [PMID: 31475655 DOI: 10.1017/s0007114519001260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
For the same BMI, South Asians have a higher body fat percentage than Caucasians. There might be differences in the fatty acid (FA) handling in adipose tissue when both ethnicities are exposed to high-fat overfeeding. The objective of the present study was to investigate the molecular adaptation in relation to FA metabolism in response to overfeeding with a high-fat diet (OHFD) in South Asian and Caucasian men. Ten South Asian men (BMI 18-29 kg/m2) and ten Caucasian men (BMI 22-33 kg/m2), matched for body fat percentage, aged 20-40 years were included. A weight-maintenance diet (30 % fat, 55 % carbohydrate and 15 % protein) was given for 3 d followed by 3 d of overfeeding (150 % energy requirement) with a high-fat diet (60 % fat, 25 % carbohydrate and 15 % protein) while staying in a respiration chamber. Before and after overfeeding, abdominal subcutaneous fat biopsies were taken. Proteins were isolated, analysed and quantified for short-chain 3-hydroxyacyl-CoA dehydrogenase (HADH), carnitine palmitoyl-transferase 1α (CPT1a), adipose TAG lipase, perilipin A (PLINA), perilipin B, lipoprotein lipase and fatty acid binding protein 4 using Western blotting. OHFD decreased the HADH level (P < 0·05) in Caucasians more than in Asians (P < 0·05), but the baseline and after intervention HADH level was relatively higher in Caucasians. The level of CPT1a decreased in South Asians and increased in Caucasians (P < 0·05). PLINA did not change with diet but the level was higher in South Asians (P < 0·05). The observed differences in HADH and PLINA levels as well as in CPT1a response may be important for differences in the long-term regulation of energy (fat) metabolism in these populations.
Collapse
|
8
|
Substrate utilization and metabolic profile in response to overfeeding with a high-fat diet in South Asian and white men: a sedentary lifestyle study. Int J Obes (Lond) 2019; 44:136-146. [PMID: 31040398 DOI: 10.1038/s41366-019-0368-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/02/2019] [Accepted: 03/10/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND For the same BMI, South Asians have a higher body fat percentage, a higher liver fat content and a more adverse metabolic profile than whites. South Asians may have a lower fat oxidation than whites, which could result in an unfavorable metabolic profile when exposed to increased high-fat foods consumption and decreased physical activity as in current modern lifestyle. OBJECTIVE To determine substrate partitioning, liver fat accumulation and metabolic profile in South Asian and white men in response to overfeeding with high-fat diet under sedentary conditions in a respiration chamber. DESIGN Ten South Asian men (BMI, 18-29 kg/m2) and 10 white men (BMI, 22-33 kg/m2), matched for body fat percentage, aged 20-40 year were included. A weight maintenance diet (30% fat, 55% carbohydrate, and 15% protein) was given for 3 days. Thereafter, a baseline measurement of liver fat content (1H-MRS) and blood parameters was performed. Subsequently, subjects were overfed (150% energy requirement) with a high-fat diet (60% fat, 25% carbohydrate, and 15% protein) over 3 consecutive days while staying in a respiration chamber mimicking a sedentary lifestyle. Energy expenditure and substrate use were measured for 3 × 24-h. Liver fat and blood parameters were measured again after the subjects left the chamber. RESULTS The 24-h fat oxidation as a percentage of total energy expenditure did not differ between ethnicities (P = 0.30). Overfeeding increased liver fat content (P = 0.02), but the increase did not differ between ethnicities (P = 0.64). In South Asians, overfeeding tended to increase LDL-cholesterol (P = 0.08), tended to decrease glucose clearance (P = 0.06) and tended to elevate insulin response (P = 0.07) slightly more than whites. CONCLUSIONS Despite a similar substrate partitioning and similar accretion of liver fat, overfeeding with high-fat under sedentary conditions tended to have more adverse effects on the lipid profile and insulin sensitivity in South Asians.
Collapse
|
9
|
Abstract
Although metabolic abnormalities commonly occur in non-obese Asians, their pathogenesis is not fully understood. Proton magnetic resonance spectroscopy has been used to analyze intracellular lipids in humans, and results suggest that ectopic fat accumulation in muscle and liver may induce insulin resistance in each tissue independently of obesity. Thus, measurement of ectopic fat currently plays an important role in the study of insulin resistance in non-obese Asians. In addition, studies using 2-step hyperinsulinemic euglycemic clamp with a glucose tracer may clarify how tissue-specific insulin resistance in muscle, liver, and adipose tissue contributes to the development of metabolic disease in non-obese Japanese. Although numerous studies have elucidated the pathophysiology of insulin resistance in obese subjects, research on "metabolic gradation," defined as the gradual transition from an insulin-sensitive to an insulin-resistant state, is less common, especially in terms of early metabolic changes. This review addresses a simple question: when and how is insulin resistance induced in non-obese East Asians? Several studies revealed that impaired insulin clearance and hyperinsulinemia not only compensated for insulin resistance, but also secondarily facilitated insulin resistance and weight gain. In this regard, we recently found that impaired insulin clearance and hyperinsulinemia could occur in apparently healthy subjects without significant insulin resistance, suggesting that this change may be an initial trigger that drives subsequent insulin resistance and weight gain. Further research is required to clarify the pathogenesis of metabolic gradation in non-obese Asians.
Collapse
Affiliation(s)
- Yoshifumi Tamura
- Department of Metabolism & Endocrinology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Faculty of International Liberal Arts, Juntendo University, Tokyo, Japan
| |
Collapse
|
10
|
Lundsgaard AM, Holm JB, Sjøberg KA, Bojsen-Møller KN, Myrmel LS, Fjære E, Jensen BAH, Nicolaisen TS, Hingst JR, Hansen SL, Doll S, Geyer PE, Deshmukh AS, Holst JJ, Madsen L, Kristiansen K, Wojtaszewski JFP, Richter EA, Kiens B. Mechanisms Preserving Insulin Action during High Dietary Fat Intake. Cell Metab 2019; 29:50-63.e4. [PMID: 30269983 DOI: 10.1016/j.cmet.2018.08.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/21/2018] [Accepted: 08/30/2018] [Indexed: 01/03/2023]
Abstract
Prolonged intervention studies investigating molecular metabolism are necessary for a deeper understanding of dietary effects on health. Here we provide mechanistic information about metabolic adaptation to fat-rich diets. Healthy, slightly overweight men ingested saturated or polyunsaturated fat-rich diets for 6 weeks during weight maintenance. Hyperinsulinemic clamps combined with leg balance technique revealed unchanged peripheral insulin sensitivity, independent of fatty acid type. Both diets increased fat oxidation potential in muscle. Hepatic insulin clearance increased, while glucose production, de novo lipogenesis, and plasma triacylglycerol decreased. High fat intake changed the plasma proteome in the immune-supporting direction and the gut microbiome displayed changes at taxonomical and functional level with polyunsaturated fatty acid (PUFA). In mice, eucaloric feeding of human PUFA and saturated fatty acid diets lowered hepatic triacylglycerol content compared with low-fat-fed control mice, and induced adaptations in the liver supportive of decreased gluconeogenesis and lipogenesis. Intake of fat-rich diets thus induces extensive metabolic adaptations enabling disposition of dietary fat without metabolic complications.
Collapse
Affiliation(s)
- Anne-Marie Lundsgaard
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Jacob B Holm
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Clinical Microbiomics, Copenhagen, Denmark
| | - Kim A Sjøberg
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | | | | | - Even Fjære
- Institute of Marine Research, Bergen, Norway
| | - Benjamin A H Jensen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Department of Medicine, Laval University, Quebec, QC, Canada
| | - Trine S Nicolaisen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Janne R Hingst
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Sine L Hansen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Sophia Doll
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Munich, Germany
| | - Philip E Geyer
- Department of Proteomics and Signal Transduction, Max-Planck-Institute of Biochemistry, Munich, Germany
| | - Atul S Deshmukh
- The Novo Nordisk Foundation Center for Protein Research, Clinical Proteomics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Madsen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Institute of Marine Research, Bergen, Norway
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Institute of Metagenomics, BGI-Shenzhen, Shenzhen, China
| | - Jørgen F P Wojtaszewski
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Erik A Richter
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark
| | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Universitetsparken 13, Copenhagen 2100, Denmark.
| |
Collapse
|
11
|
Gancheva S, Jelenik T, Álvarez-Hernández E, Roden M. Interorgan Metabolic Crosstalk in Human Insulin Resistance. Physiol Rev 2018; 98:1371-1415. [PMID: 29767564 DOI: 10.1152/physrev.00015.2017] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Excessive energy intake and reduced energy expenditure drive the development of insulin resistance and metabolic diseases such as obesity and type 2 diabetes mellitus. Metabolic signals derived from dietary intake or secreted from adipose tissue, gut, and liver contribute to energy homeostasis. Recent metabolomic studies identified novel metabolites and enlarged our knowledge on classic metabolites. This review summarizes the evidence of their roles as mediators of interorgan crosstalk and regulators of insulin sensitivity and energy metabolism. Circulating lipids such as free fatty acids, acetate, and palmitoleate from adipose tissue and short-chain fatty acids from the gut effectively act on liver and skeletal muscle. Intracellular lipids such as diacylglycerols and sphingolipids can serve as lipotoxins by directly inhibiting insulin action in muscle and liver. In contrast, fatty acid esters of hydroxy fatty acids have been recently shown to exert a series of beneficial effects. Also, ketoacids are gaining interest as potent modulators of insulin action and mitochondrial function. Finally, branched-chain amino acids not only predict metabolic diseases, but also inhibit insulin signaling. Here, we focus on the metabolic crosstalk in humans, which regulates insulin sensitivity and energy homeostasis in the main insulin-sensitive tissues, skeletal muscle, liver, and adipose tissue.
Collapse
Affiliation(s)
- Sofiya Gancheva
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| | - Tomas Jelenik
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| | - Elisa Álvarez-Hernández
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| |
Collapse
|
12
|
Sa’ad-Aldin K, Altamimi M. Effect of whole-grain plant-based diet on the diabetes mellitus type 2 features in newly diagnosed patients: a pilot study. Int J Diabetes Dev Ctries 2018. [DOI: 10.1007/s13410-018-0689-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
13
|
Sions JM, Teyhen DS, Hicks GE. Criterion Validity of Ultrasound Imaging: Assessment of Multifidi Cross-Sectional Area in Older Adults With and Without Chronic Low Back Pain. J Geriatr Phys Ther 2018; 40:74-79. [PMID: 26703525 DOI: 10.1519/jpt.0000000000000073] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND AND PURPOSE Ultrasound (US) imaging may be a cost-conscious alternative to magnetic resonance imaging (MRI), which is the criterion standard for muscle cross-sectional area (CSA) assessment. Within the trunk, when compared with MRI, US has been shown to be valid for assessing lumbar multifidi CSA in younger, asymptomatic individuals. To date, there are no studies validating US for multifidi CSA assessment in older adults or individuals with low back pain. Given age- and pain-related muscle changes, validation of US is needed in these populations. If valid for multifidi CSA assessment, US may be used to evaluate short-term changes in muscle size in response to exercise-based interventions among older adults. The primary objective of this study was to evaluate the validity of US for multifidi CSA assessment as compared with MRI in older adults with and without chronic low back pain (CLBP). The secondary objective was to determine whether a single US image was valid for assessment of multifidi CSA or whether the average of 3 US images should be recommended. METHODS Twenty community-dwelling older adults (ie, 10 with and 10 without CLBP), ages 60 to 85 years, were recruited. US images and MRI slices of multifidi muscle were obtained and L4 multifidi CSAs were measured. Intraclass correlation coefficients (ICCs) were calculated to assess agreement between MRI measures and a single US image and MRI measures and the average of 3 US images. RESULTS AND DISCUSSION ICC point estimates were excellent for older adults with CLBP for a single US image (ICCs = 0.90-0.97), but ICC point estimates for participants without CLBP ranged from fair to excellent (ICCs = 0.48-0.86). ICC point estimates for the average of 3 US images for both groups were better than for a single image (ICCs = 0.95-0.99). CONCLUSIONS For assessment of L4 multifidi CSA, US is a valid alternative to MRI for older adults with and without CLBP. However, limitations of US, such as the inability to quantify intramuscular fat, which may be increased with aging and CLBP, should be considered. CSA measurement of 3 US images, rather than a single image, is recommended.
Collapse
Affiliation(s)
- Jaclyn Megan Sions
- 1Department of Physical Therapy, University of Delaware, Newark, Delaware. 2Office of the Surgeon General, U.S. Army Medical Command, Falls Church, Virginia
| | | | | |
Collapse
|
14
|
Effects of a High-Protein Diet Including Whole Eggs on Muscle Composition and Indices of Cardiometabolic Health and Systemic Inflammation in Older Adults with Overweight or Obesity: A Randomized Controlled Trial. Nutrients 2018; 10:nu10070946. [PMID: 30041437 PMCID: PMC6073477 DOI: 10.3390/nu10070946] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/25/2022] Open
Abstract
Age-related increases in intermuscular adipose tissue (IMAT) impair muscle quality, decrease functional capacity, and promote several cardiometabolic and inflammatory disorders. Whether these age-related alterations in muscle composition improve by consuming a high-protein (HP) diet with whole eggs are unclear. This parallel-design, randomized-controlled trial assessed the effects of a 12-week eucaloric HP diet with three whole eggs per day (1.4 g protein kg−1 day−1) versus a normal-protein diet void of eggs (NP, 0.8 g protein kg−1 day−1) on muscle composition (IMAT), cardiometabolic health, and systemic inflammation in older adults with overweight or obesity (12 men and 10 women; age 70 ± 5 years, BMI 31.3 ± 3.2 kg/m2, mean ± SD). No changes in muscle composition were observed over time, independent of protein intake. Total body weight was reduced in both groups (−3.3 ± 1.2%) and lean mass was preserved only with the HP diet. LDL concentration and hip circumference decreased only with the NP diet, while MCP-1 and HsCRP concentrations increased over time in both groups. A HP diet with whole eggs promotes lean mass retention with modest weight loss, but does not positively influence muscle composition, cardiometabolic health or systemic inflammation, compared to a NP diet void of eggs.
Collapse
|
15
|
Daemen S, van Polanen N, Hesselink MKC. The effect of diet and exercise on lipid droplet dynamics in human muscle tissue. ACTA ACUST UNITED AC 2018. [PMID: 29514886 DOI: 10.1242/jeb.167015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The majority of fat in the human body is stored as triacylglycerols in white adipose tissue. In the obese state, adipose tissue mass expands and excess lipids are stored in non-adipose tissues, such as skeletal muscle. Lipids are stored in skeletal muscle in the form of small lipid droplets. Although originally viewed as dull organelles that simply store lipids as a consequence of lipid overflow from adipose tissue, lipid droplets are now recognized as key components in the cell that exert a variety of relevant functions in multiple tissues (including muscle). Here, we review the effect of diet and exercise interventions on myocellular lipid droplets and their putative role in insulin sensitivity from a human perspective. We also provide an overview of lipid droplet biology and identify gaps for future research.
Collapse
Affiliation(s)
- Sabine Daemen
- Department of Human Biology and Human Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200MD Maastricht, The Netherlands
| | - Nynke van Polanen
- Department of Human Biology and Human Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200MD Maastricht, The Netherlands
| | - Matthijs K C Hesselink
- Department of Human Biology and Human Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, 6200MD Maastricht, The Netherlands
| |
Collapse
|
16
|
Baek KW, Cha HJ, Ock MS, Kim HS, Gim JA, Park JJ. Effects of regular-moderate exercise on high-fat diet-induced intramyocellular lipid accumulation in the soleus muscle of Sprague-Dawley rats. J Exerc Rehabil 2018; 14:32-38. [PMID: 29511650 PMCID: PMC5833965 DOI: 10.12965/jer.1835166.583] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/16/2018] [Indexed: 11/22/2022] Open
Abstract
Previously, we monitored the expression level of the pro-apoptotic proteins caspase-3 and cleaved poly-ADP-ribose polymerase in the skeletal muscle of high-fat diet-induced obese rats in order to assess muscle damage. In this study, we analyzed whether exercise or dietary adjustment was more effective at preventing high-fat diet-induced muscle damage. High-fat diet-induced obese rats were divided into three groups: the high-fat diet (HFD), the combined high-fat diet and exercise (HFD+EXE), and the dietary adjustment (DA) groups. For 6 weeks, the HFD+EXE group was subjected to exercise on an animal treadmill. Capsase-3 protein was quantified, and histopathology of the soleus muscle was performed. Both the HFD+EXE and DA interventions resulted in a reduction of lipid accumulation in the soleus muscle, and nucleus infiltration was significantly lower in the DA group. The inflammatory response, caspase-3 level, and relative muscle weight were significantly higher in the HFD+EXE group compared to the HFD group. An increase in intramyocellular lipids in the soleus muscle by obesity and exercise stimulated apoptosis. When the rats exercised, muscle growth was normal and unrelated to the effects of lipid accumulation. These data indicate that exercise was more effective than dietary adjustment in reducing lipid accumulation and increasing muscle metabolism.
Collapse
Affiliation(s)
- Kyung-Wan Baek
- Division of Sport Science, Pusan National University, Busan, Korea.,Department of Parasitology and Genetics, Kosin University College of Medicine, Busan, Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan, Korea
| | - Mee Sun Ock
- Department of Parasitology and Genetics, Kosin University College of Medicine, Busan, Korea
| | - Hong Soo Kim
- Department of Gastroenterology/Hepatology, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Jeong-An Gim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Korea
| | - Jung-Jun Park
- Division of Sport Science, Pusan National University, Busan, Korea
| |
Collapse
|
17
|
Ahmed S, Singh D, Khattab S, Babineau J, Kumbhare D. The Effects of Diet on the Proportion of Intramuscular Fat in Human Muscle: A Systematic Review and Meta-analysis. Front Nutr 2018. [PMID: 29516003 PMCID: PMC5826234 DOI: 10.3389/fnut.2018.00007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background There is an increasing trend in the consumption of poor-quality diets worldwide, contributing to the increase of non-communicable diseases. Diet directly influences physiological composition and subsequently physical health. Studies have shown that dietary macronutrient and energy content can influence the proportion of intramuscular fat (IMF), which mediates various metabolic and endocrine dysfunction. The purpose of this systematic review was to identify evidence in the literature assessing the association between different dietary interventions on the proportion of IMF in humans. Methods Three medical databases were investigated (Medline, EMBASE, and Cochrane) to identify studies assessing changes in IMF after dietary interventions. The primary outcome measure was the change in IMF proportions after a dietary intervention. The effects of high-fat, high-carbohydrate, low-calorie, and starvation diets were assessed qualitatively. A meta-analysis assessing the effect of high-fat diets was conducted. Follow-up sensitivity and subgroup analyses were also conducted. Results One thousand eight hundred and sixty-six articles were identified for review. Of these articles, 13 were eligible for inclusion after a full screening. High-fat diets increased IMF proportions, standardized mean difference = 1.24 (95% confidence interval, 0.43–2.05) and a significant overall effect size (P = 0.003). Diets with an increased proportion of carbohydrates decreased IMF proportions; however, increasing caloric intake with carbohydrates increased IMF. Starvation diets increased IMF stores, and hypocaloric diets did not result in any IMF proportion changes. Conclusion This systematic review suggests that high-fat diets and diets with caloric intake increased above the amount required to maintain BMI with carbohydrates, and short-term starvation diets are associated with increases in IMF content. Further studies are needed to assess the effects of macronutrient combinations on IMF and the influence of diet-induced IMF alterations on health outcomes. In addition, IMF poses a possibly effective clinical marker of health.
Collapse
Affiliation(s)
- Sara Ahmed
- McMaster University, Hamilton, ON, Canada
| | - Dhanveer Singh
- Royal College of Physicians and Surgeons in Ireland, Dublin, Ireland
| | | | - Jessica Babineau
- Library and Information Services, Toronto Rehabilitation Institute, University Health Network, Toronto, ON, Canada
| | - Dinesh Kumbhare
- Division of Physical Medicine and Rehabilitation, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
18
|
Covington JD, Johannsen DL, Coen PM, Burk DH, Obanda DN, Ebenezer PJ, Tam CS, Goodpaster BH, Ravussin E, Bajpeyi S. Intramyocellular Lipid Droplet Size Rather Than Total Lipid Content is Related to Insulin Sensitivity After 8 Weeks of Overfeeding. Obesity (Silver Spring) 2017; 25:2079-2087. [PMID: 29071793 PMCID: PMC5705570 DOI: 10.1002/oby.21980] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/05/2017] [Accepted: 07/16/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Intramyocellular lipid (IMCL) is inversely related to insulin sensitivity in sedentary populations, yet no prospective studies in humans have examined IMCL accumulation with overfeeding. METHODS Twenty-nine males were overfed a high-fat diet (140% caloric intake, 44% from fat) for 8 weeks. Measures of IMCL, whole-body fat oxidation from a 24-hour metabolic chamber, muscle protein extracts, and muscle ceramide measures were obtained before and after the intervention. RESULTS Eight weeks of overfeeding did not increase overall IMCL. The content of smaller lipid droplets peripherally located in the myofiber decreased, while increases in larger droplets correlated inversely with glucose disposal rate. Overfeeding resulted in inhibition of Akt activity, which correlated with the reductions in smaller, peripherally located lipid droplets and drastic increases in ceramide content. Additionally, peripherally located lipid droplets were associated with more efficient lipid oxidation. Finally, participants who maintained a greater number of smaller, peripherally located lipid droplets displayed a better resistance to weight gain with overfeeding. CONCLUSIONS These results show that lipid droplet size and location rather than mere IMCL content are important to understanding insulin sensitivity.
Collapse
Affiliation(s)
- Jeffrey D. Covington
- Pennington Biomedical Research Center, Laboratory of Skeletal Muscle Physiology, 6400 Perkins Road, Baton Rouge, LA 70808
- Louisiana State University Health Sciences Center, School of Medicine, 433 Bolivar St, New Orleans, LA 70112
| | - Darcy L. Johannsen
- Pennington Biomedical Research Center, Laboratory of Skeletal Muscle Physiology, 6400 Perkins Road, Baton Rouge, LA 70808
| | - Paul M. Coen
- Translational Research Institute for Metabolism and Diabetes Florida Hospital • Sanford-Burnham Medical Research Institute, 301 East Princeton Street, Orlando, FL 32804
| | - David H. Burk
- Pennington Biomedical Research Center, Laboratory of Skeletal Muscle Physiology, 6400 Perkins Road, Baton Rouge, LA 70808
| | - Diana N. Obanda
- Pennington Biomedical Research Center, Laboratory of Skeletal Muscle Physiology, 6400 Perkins Road, Baton Rouge, LA 70808
| | - Philip J. Ebenezer
- Pennington Biomedical Research Center, Laboratory of Skeletal Muscle Physiology, 6400 Perkins Road, Baton Rouge, LA 70808
| | - Charmaine S. Tam
- The Charles Perkins Centre and The School of Biological Sciences, University of Sydney, NSW, Australia
| | - Bret H. Goodpaster
- Translational Research Institute for Metabolism and Diabetes Florida Hospital • Sanford-Burnham Medical Research Institute, 301 East Princeton Street, Orlando, FL 32804
| | - Eric Ravussin
- Pennington Biomedical Research Center, Laboratory of Skeletal Muscle Physiology, 6400 Perkins Road, Baton Rouge, LA 70808
| | - Sudip Bajpeyi
- Pennington Biomedical Research Center, Laboratory of Skeletal Muscle Physiology, 6400 Perkins Road, Baton Rouge, LA 70808
- Universtiy of Texas at El Paso, Department of Kinesiology, 500 University Ave, El Paso, TX, 79968
| |
Collapse
|
19
|
Gepner Y, Shelef I, Schwarzfuchs D, Cohen N, Bril N, Rein M, Tsaban G, Zelicha H, Yaskolka Meir A, Tene L, Sarusy B, Rosen P, Hoffman JR, Stout JR, Thiery J, Ceglarek U, Stumvoll M, Blüher M, Stampfer MJ, Shai I. Intramyocellular triacylglycerol accumulation across weight loss strategies; Sub-study of the CENTRAL trial. PLoS One 2017; 12:e0188431. [PMID: 29190720 PMCID: PMC5708655 DOI: 10.1371/journal.pone.0188431] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Intramyocellular triacylglycerol (IMTG) is utilized as metabolic fuel during exercise and is linked to insulin resistance, but the long-term effect of weight loss strategies on IMTG among participants with abdominal fat, remain unclear. METHODS In an 18-month trial, sedentary participants with abdominal fat/dyslipidemia were randomized to either a low-fat (LF) or Mediterranean/low-carbohydrate (MED/LC) diet (including 28g·day-1 of walnuts). After 6-months, the participants were re-randomized to moderate intense physical activity (PA+) or non-physical activity (PA-). Magnetic resonance imaging (MRI) was used to quantify changes of IMTG, abdominal sub-depots, hepatic and intermuscular fats. RESULTS Across the 277 participants [86% men, age = 48 years, body-mass-index (BMI) = 31kg/m2, visceral fat = 33%] 86% completed the 18-m trial. At baseline, women had higher IMTG than men (3.4% vs. 2.3%, p<0.001) and increased IMTG was associated with aging and higher BMI, visceral and intermuscular fats, HbA1c%, HDL-c and leptin(p<0.05), but not with intra-hepatic fat. After 18 month of intervention and a -3 kg mean weight loss, participants significantly increased IMTG by 25%, with a distinct effect in the MED/LCPA+ group as compared to the other intervention groups (57% vs. 9.5-18.5%, p<0.05). Changes in IMTG were associated with visceral and intermuscular fat, metabolic syndrome, insulin and leptin (p<0.05 for all), however, these associations did not remain after adjustment for visceral fat changes. CONCLUSIONS Lifestyle strategies differentially affect IMTG accumulation; combination of exercise with decreased carbohydrate/increased unsaturated fat proportion intake greatly increase IMTG. Our findings suggest that increased IMTG during diet-induced moderate weight loss may not be directly related to cardiometabolic risk. TRIAL REGISTRATION ClinicalTrials.gov NCT01530724.
Collapse
Affiliation(s)
- Yftach Gepner
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- Institute of Exercise Physiology and Wellness, Sport and Exercise Science; University of Central Florida, Orlando, FL, United States of America
| | - Ilan Shelef
- Soroka University Medical Center, Beer-Sheva, Israel
| | | | - Noa Cohen
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nitzan Bril
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Michal Rein
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Gal Tsaban
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hila Zelicha
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anat Yaskolka Meir
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Lilac Tene
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Philip Rosen
- Soroka University Medical Center, Beer-Sheva, Israel
| | - Jay R. Hoffman
- Institute of Exercise Physiology and Wellness, Sport and Exercise Science; University of Central Florida, Orlando, FL, United States of America
| | - Jeffrey R. Stout
- Institute of Exercise Physiology and Wellness, Sport and Exercise Science; University of Central Florida, Orlando, FL, United States of America
| | - Joachim Thiery
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | | | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Meir J. Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard School of Public Health, Boston, MA, United States of America
| | - Iris Shai
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
20
|
Blaak EE. Characterisation of fatty acid metabolism in different insulin-resistant phenotypes by means of stable isotopes. Proc Nutr Soc 2017; 76:1-7. [PMID: 28100287 DOI: 10.1017/s0029665116003013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The obese insulin resistant and/or prediabetic state is characterised by systemic lipid overflow, mainly driven by an impaired lipid buffering capacity of adipose tissue, and an impaired capacity of skeletal muscle to increase fat oxidation upon increased supply. This leads to the accumulation of bioactive lipid metabolites in skeletal muscle interfering with insulin sensitivity via various mechanisms. In this review, the contribution of dietary v. endogenous fatty acids to lipid overflow, their extraction or uptake by skeletal muscle as well as the fractional synthetic rate, content and composition of the muscle lipid pools is discussed in relation to the development or presence of insulin resistance and/or an impaired glucose metabolism. These parameters are studied in vivo in man by combining a dual stable isotope methodology with [2H2]- and [U-13C]-palmitate tracers with the arterio-venous balance technique across forearm muscle and biochemical analyses in muscle biopsies. The insulin-resistant state is characterised by an elevated muscle TAG extraction, despite similar supply, and a reduced skeletal muscle lipid turnover, in particular after intake of a high fat, SFA fat meal, but not after a high fat, PUFA meal. Data are placed in the context of current literature, and underlying mechanisms and implications for long-term nutritional interventions are discussed.
Collapse
Affiliation(s)
- Ellen E Blaak
- Department of Human Biology,Maastricht University,Maastricht,The Netherlands
| |
Collapse
|
21
|
The Flexibility of Ectopic Lipids. Int J Mol Sci 2016; 17:ijms17091554. [PMID: 27649157 PMCID: PMC5037826 DOI: 10.3390/ijms17091554] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 02/07/2023] Open
Abstract
In addition to the subcutaneous and the visceral fat tissue, lipids can also be stored in non-adipose tissue such as in hepatocytes (intrahepatocellular lipids; IHCL), skeletal (intramyocellular lipids; IMCL) or cardiac muscle cells (intracardiomyocellular lipids; ICCL). Ectopic lipids are flexible fuel stores that can be depleted by physical exercise and repleted by diet. They are related to obesity and insulin resistance. Quantification of IMCL was initially performed invasively, using muscle biopsies with biochemical and/or histological analysis. 1H-magnetic resonance spectroscopy (1H-MRS) is now a validated method that allows for not only quantifying IMCL non-invasively and repeatedly, but also assessing IHCL and ICCL. This review summarizes the current available knowledge on the flexibility of ectopic lipids. The available evidence suggests a complex interplay between quantitative and qualitative diet, fat availability (fat mass), insulin action, and physical exercise, all important factors that influence the flexibility of ectopic lipids. Furthermore, the time frame of the intervention on these parameters (short-term vs. long-term) appears to be critical. Consequently, standardization of physical activity and diet are critical when assessing ectopic lipids in predefined clinical situations.
Collapse
|
22
|
Sundaresan A, Radhiga T, Pugalendi KV. Ursolic acid and rosiglitazone combination improves insulin sensitivity by increasing the skeletal muscle insulin-stimulated IRS-1 tyrosine phosphorylation in high-fat diet-fed C57BL/6J mice. J Physiol Biochem 2016; 72:345-52. [PMID: 27090933 DOI: 10.1007/s13105-016-0484-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 04/06/2016] [Indexed: 12/21/2022]
Abstract
The aim of this present study was to investigate the effect of ursolic acid (UA) and rosiglitazone (RSG) on insulin sensitivity and proximal insulin signaling pathways in high-fat diet (HFD)-fed C57/BL/6J mice. Male C57BL/6J mice were fed either normal diet or HFD for 10 weeks, after which animals in each dietary group were divided into the following six groups (normal diet, normal diet plus UA and RSG, HFD alone, HFD plus UA, HFD plus RSG, and HFD plus UA and RSG) for the next 5 weeks. UA (5 mg/kg BW) and RSG (4 mg/kg BW) were administered as suspensions directly into the stomach using a gastric tube. The HFD diet elevated fasting plasma glucose, insulin, and homeostasis model assessment index. The expression of insulin receptor substrate (IRS)-1, phosphoinositide 3-kinase (PI3-kinase), Akt, and glucose transporter (GLUT) 4 were determined by Western blot analyses. The results demonstrated that combination treatment (UA/RSG) ameliorated HFD-induced glucose intolerance and insulin resistance by improving the homeostatic model assessment (HOMA) index. Further, combination treatment (UA/RSG) stimulated the IRS-1, PI3-kinase, Akt, and GLUT 4 translocation. These results strongly suggest that combination treatment (UA/RSG) activates IRS-PI3-kinase-Akt-dependent signaling pathways to induce GLUT 4 translocation and increases the expression of insulin receptor to improve glucose intolerance.
Collapse
MESH Headings
- Animals
- Anti-Obesity Agents/adverse effects
- Anti-Obesity Agents/therapeutic use
- Antioxidants/therapeutic use
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat/adverse effects
- Drug Therapy, Combination/adverse effects
- Glucose Transporter Type 4/metabolism
- Hypoglycemic Agents/adverse effects
- Hypoglycemic Agents/therapeutic use
- Insulin Receptor Substrate Proteins/agonists
- Insulin Receptor Substrate Proteins/metabolism
- Insulin Resistance
- Male
- Mice, Inbred C57BL
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Obesity/chemically induced
- Obesity/complications
- Phosphatidylinositol 3-Kinase/metabolism
- Phosphorylation/drug effects
- Protein Processing, Post-Translational/drug effects
- Protein Transport/drug effects
- Rosiglitazone
- Second Messenger Systems/drug effects
- Thiazolidinediones/adverse effects
- Thiazolidinediones/therapeutic use
- Triterpenes/adverse effects
- Triterpenes/therapeutic use
- Weight Gain/drug effects
- Ursolic Acid
Collapse
Affiliation(s)
- Arjunan Sundaresan
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608 002, Tamil Nadu, India
| | - Thangaiyan Radhiga
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608 002, Tamil Nadu, India
| | - Kodukkur Viswanathan Pugalendi
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, 608 002, Tamil Nadu, India.
| |
Collapse
|
23
|
Morton TL, Galior K, McGrath C, Wu X, Uzer G, Uzer GB, Sen B, Xie Z, Tyson D, Rubin J, Styner M. Exercise Increases and Browns Muscle Lipid in High-Fat Diet-Fed Mice. Front Endocrinol (Lausanne) 2016; 7:80. [PMID: 27445983 PMCID: PMC4928595 DOI: 10.3389/fendo.2016.00080] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022] Open
Abstract
Muscle lipid increases with high-fat feeding and diabetes. In trained athletes, increased muscle lipid is not associated with insulin resistance, a phenomenon known as the athlete's paradox. To understand if exercise altered the phenotype of muscle lipid, female C57BL/6 mice fed CTL or high-fat diet (HFD for 6 or 18 weeks) were further divided into sedentary or exercising groups (CTL-E or HFD-E) with voluntary access to running wheels for the last 6 weeks of experiments, running 6 h/night. Diet did not affect running time or distance. HFD mice weighed more than CTL after 18 weeks (p < 0.01). Quadriceps muscle TG was increased in running animals and in sedentary mice fed HFD for 18 weeks (p < 0.05). In exercised animals, markers of fat, Plin1, aP2, FSP27, and Fasn, were increased significantly in HFD groups. Ucp1 and Pgc1a, markers for brown fat, increased with exercise in the setting of high fat feeding. Fndc5, which encodes irisin, and CytC were sensitive to exercise regardless of diet. Plin5 was increased with HFD and unaffected by exercise; the respiratory exchange ratio was 15% lower in the 18-week HFD group compared with CTL (p < 0.001) and 10% lower in 18 weeks HFD-E compared with CTL-E (p < 0.001). Increased Ucp1 and Pgc1a in exercised muscle of running mice suggests that a beige/brown fat phenotype develops, which differs from the fat phenotype that induces insulin resistance in high fat feeding. This suggests that increased muscle lipid may develop a "brown" phenotype in the setting of endurance exercise training, a shift that is further promoted by HFD.
Collapse
Affiliation(s)
- Tiffany L. Morton
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kornelia Galior
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cody McGrath
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xin Wu
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gunes Uzer
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Guniz Bas Uzer
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Buer Sen
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhihui Xie
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David Tyson
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janet Rubin
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maya Styner
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- *Correspondence: Maya Styner,
| |
Collapse
|
24
|
Sasaki H, Ishibashi A, Tsuchiya Y, Shimura N, Kurihara T, Ebi K, Goto K. A 3-day high-fat/low-carbohydrate diet does not alter exercise-induced growth hormone response in healthy males. Growth Horm IGF Res 2015; 25:304-311. [PMID: 26387476 DOI: 10.1016/j.ghir.2015.07.005] [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: 03/14/2015] [Revised: 07/03/2015] [Accepted: 07/13/2015] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The purpose of the present study was to examine the effects of 3 days isoenergetic high-fat/low-carbohydrate diet (HF-LC) relative to low-fat/high-carbohydrate diet (LF-HC) on the exercise-induced growth hormone (GH) response in healthy male subjects. DESIGN Ten healthy young males participated in this study. Each subject consumed the HF-LC (18±1% protein, 61±2% fat, 21±1% carbohydrate, 2720 kcal per day) for 3 consecutive days after consuming the LF-HC (18±1% protein, 20±1% fat, 62±1% carbohydrate, 2755 kcal per day) for 3 consecutive days. After each dietary intervention period, the hormonal and metabolic responses to an acute exercise (30 min of continuous pedaling at 60% of V˙O2max) were compared. The intramyocellular lipid (IMCL) contents in the vastus lateralis, soleus, and tibialis anterior were evaluated by proton magnetic resonance spectroscopy. RESULTS Serum GH concentrations increased significantly during the exercise after both the HF-LC and LF-HC periods (P<0.05). However, the exercise-induced GH response was not significantly different between the two periods. Fat utilization and lipolytic responses during the exercise were enhanced significantly after the HF-LC period compared with the LF-HC period. IMCL content did not differ significantly in any portion of muscle after the dietary interventions. CONCLUSIONS We could not show that short-term HF-LC consumption changed significantly exercise-induced GH response or IMCL content in healthy young males.
Collapse
Affiliation(s)
- Hiroto Sasaki
- Graduate School of Sport and Health Science, Ritsumeikan University, Shiga, Japan.
| | - Aya Ishibashi
- Graduate School of Sport and Health Science, Ritsumeikan University, Shiga, Japan.
| | - Yoshihumi Tsuchiya
- Graduate School of Sport and Health Science, Ritsumeikan University, Shiga, Japan.
| | - Nobuhiro Shimura
- Graduate School of Sport and Health Science, Ritsumeikan University, Shiga, Japan.
| | - Toshiyuki Kurihara
- Graduate School of Sport and Health Science, Ritsumeikan University, Shiga, Japan; Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan.
| | - Kumiko Ebi
- Graduate School of Sport and Health Science, Ritsumeikan University, Shiga, Japan; Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan.
| | - Kazushige Goto
- Graduate School of Sport and Health Science, Ritsumeikan University, Shiga, Japan; Faculty of Sport and Health Science, Ritsumeikan University, Shiga, Japan.
| |
Collapse
|
25
|
Stinkens R, Goossens GH, Jocken JWE, Blaak EE. Targeting fatty acid metabolism to improve glucose metabolism. Obes Rev 2015; 16:715-57. [PMID: 26179344 DOI: 10.1111/obr.12298] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/23/2015] [Accepted: 05/10/2015] [Indexed: 12/15/2022]
Abstract
Disturbances in fatty acid metabolism in adipose tissue, liver, skeletal muscle, gut and pancreas play an important role in the development of insulin resistance, impaired glucose metabolism and type 2 diabetes mellitus. Alterations in diet composition may contribute to prevent and/or reverse these disturbances through modulation of fatty acid metabolism. Besides an increased fat mass, adipose tissue dysfunction, characterized by an altered capacity to store lipids and an altered secretion of adipokines, may result in lipid overflow, systemic inflammation and excessive lipid accumulation in non-adipose tissues like liver, skeletal muscle and the pancreas. These impairments together promote the development of impaired glucose metabolism, insulin resistance and type 2 diabetes mellitus. Furthermore, intrinsic functional impairments in either of these organs may contribute to lipotoxicity and insulin resistance. The present review provides an overview of fatty acid metabolism-related pathways in adipose tissue, liver, skeletal muscle, pancreas and gut, which can be targeted by diet or food components, thereby improving glucose metabolism.
Collapse
Affiliation(s)
- R Stinkens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - G H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - J W E Jocken
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - E E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
26
|
de Ligt M, Timmers S, Schrauwen P. Resveratrol and obesity: Can resveratrol relieve metabolic disturbances? Biochim Biophys Acta Mol Basis Dis 2015; 1852:1137-44. [DOI: 10.1016/j.bbadis.2014.11.012] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 10/31/2014] [Accepted: 11/11/2014] [Indexed: 12/25/2022]
|
27
|
Wulan SN, Schrauwen-Hinderling VB, Westerterp KR, Plasqui G. Liver fat accumulation in response to overfeeding with a high-fat diet: a comparison between South Asian and Caucasian men. Nutr Metab (Lond) 2015; 12:18. [PMID: 27408613 PMCID: PMC4940726 DOI: 10.1186/s12986-015-0015-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/09/2015] [Indexed: 12/04/2022] Open
Abstract
Background South Asians were reported to have a higher liver fat content as compared to BMI-matched Caucasians. This study compared the increase in liver fat content in response to overfeeding with a high fat diet in South Asian and Caucasian men when matched for body fat percentage. Methods Ten South Asian men (BMI 18–29 kg/m2) and 10 Caucasian men (BMI 22–33 kg/m2), aged 20–40 y, matched for body fat percentage, were included. A weight maintenance diet was given for 3 days based on the individual energy requirement. Individual energy requirement of the subjects was calculated based on their body composition (measured by hydro densitometry and deuterium dilution) and activity counts (accelerometer). Liver fat content was measured before and after 4 days of overfeeding (50 % excess energy need) with a high fat diet (60 % energy from fat). Fat distribution was measured by anthropometry and an MRI scan of the abdomen while liver fat content using 1H-MRS. Results While having a similar body fat % (P = 0.58), South Asians had a lower BMI (P = 0.04) than Caucasians. Liver fat content at baseline did not differ between ethnicities (P = 0.48) and was associated with visceral fat area (P = 0.002, R2 = 0.56) but not with ethnicity (P = 0.13). Overfeeding with a high fat diet significantly increased liver fat (P = 0.01) but the increase did not differ between ethnicities (P = 0.47). There was no difference in the total abdominal fat area (P = 0.37), subcutaneous abdominal fat area (P = 0.18) and visceral fat area (VAT, P = 0.32). However as a percentage of the total abdominal fat area, VAT was higher in South Asians (P = 0.003). Conclusion Despite a relatively higher percentage of visceral fat area, liver fat increased similarly in South Asian and Caucasian men in response to overfeeding with a high fat diet. Trial registration The study was registered in the public trial registry www.ccmo.nl No. NL31217.068.10.
Collapse
Affiliation(s)
- Siti N Wulan
- Department of Human Biology, Nutrition and Toxicology Research Institute (NUTRIM) - School for Nutrition Toxicology and Metabolism, Maastricht University Medical Center (MUMC+), Universiteitssingel 50, PO.Box 616, 6200MD Maastricht, The Netherlands ; Laboratory of Food Quality and Nutrition, Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Brawijaya University, Malang, East Java Indonesia
| | - Vera B Schrauwen-Hinderling
- Department of Human Biology, Nutrition and Toxicology Research Institute (NUTRIM) - School for Nutrition Toxicology and Metabolism, Maastricht University Medical Center (MUMC+), Universiteitssingel 50, PO.Box 616, 6200MD Maastricht, The Netherlands ; Department of Radiology, Maastricht Academic Hospital, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Klaas R Westerterp
- Department of Human Biology, Nutrition and Toxicology Research Institute (NUTRIM) - School for Nutrition Toxicology and Metabolism, Maastricht University Medical Center (MUMC+), Universiteitssingel 50, PO.Box 616, 6200MD Maastricht, The Netherlands
| | - Guy Plasqui
- Department of Human Biology, Nutrition and Toxicology Research Institute (NUTRIM) - School for Nutrition Toxicology and Metabolism, Maastricht University Medical Center (MUMC+), Universiteitssingel 50, PO.Box 616, 6200MD Maastricht, The Netherlands
| |
Collapse
|
28
|
Ahmed MM, Samir ESA, El-Shehawi AM, Alkafafy ME. Anti-obesity effects of Taif and Egyptian pomegranates: molecular study. Biosci Biotechnol Biochem 2015; 79:598-609. [DOI: 10.1080/09168451.2014.982505] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Abstract
The present study investigated the anti-obesity effects of pomegranate (Punica granatum) juices from the two Saudi Arabian, Taif red, Taif white, and Egyptian pomegranates in high-fat diet (HFD)-induced obese rats. Administrating any of the used juices decreased the body weight gain, food consumption, and serum levels of lipid, leptin, and glucose, while it increased serum insulin level. Histologically, all types of juices decreased the number and size of lipid droplets in hepatocytes compared to the obese, non-treated animals. All juices types upregulated the hepatic mRNA expression of hormone-sensitive lipase, pyruvate kinase, and adiponectin in obese rats; the genes were all suppressed by HFD feeding. Additionally, the expression of fatty acid synthase, sterol regulatory element-binding protein-1c, and acetyl-CoA carboxylase1 was also upregulated by all types of juices. Conversely, ghrelin mRNA expression was downregulated by all used juices’ types. These findings demonstrate that all types of tested juices protect against the HFD-induced obesity in rats.
Collapse
Affiliation(s)
- Mohamed M Ahmed
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
- Department of Biochemistry, College of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - El-Shazly A Samir
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
- Department of Biochemistry, College of Veterinary Medicine, Kaferelsheikh University, Kaferelsheikh, Egypt
| | - Ahmed M El-Shehawi
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
- Faculty of Agriculture, Department of Genetics, University of Alexandria, Alexandria, Egypt
| | - Mohamed E Alkafafy
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
- Department of Cytology and Histology, College of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| |
Collapse
|
29
|
Lindeboom L, Nabuurs CI, Hesselink MKC, Wildberger JE, Schrauwen P, Schrauwen-Hinderling VB. Proton magnetic resonance spectroscopy reveals increased hepatic lipid content after a single high-fat meal with no additional modulation by added protein. Am J Clin Nutr 2015; 101:65-71. [PMID: 25527751 DOI: 10.3945/ajcn.114.094730] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Fat accumulation in nonadipose tissue is linked to insulin resistance and metabolic diseases. Earlier studies have shown that hepatic lipid accumulation can occur after 4 d of a high-fat diet in humans, and this fat accumulation can be blunted by the ingestion of additional proteins. OBJECTIVES In this study, we explored whether a single high-fat meal increased the lipid content in liver and skeletal muscle as measured by using in vivo proton magnetic resonance spectroscopy (¹H-MRS) and whether the addition of protein can modulate the postprandial ectopic lipid storage. DESIGN Intrahepatic lipid (IHL) and intramyocellular lipid (IMCL) concentrations were determined by using ¹H-MRS before and 3 and 5 h after a high-fat with added protein meal (61.5% of energy from fat) or a high-fat without added protein meal (mean ± SEM: 51.1 ± 7.9 g of protein; 191.9 ± 9.9 kcal added) in a randomized crossover study. IHL and IMCL concentrations were converted to absolute concentrations (g/kg wet weight) by using water as an internal reference. RESULTS Nine lean, healthy subjects [6 men and 3 women; mean (±SD) age: 22.7 ± 3.0 y; mean body mass index (in kg/m²): 21.8 ± 1.8] were included in this study. IHL concentrations increased ∼20% (P < 0.01) at 3 h after the meal and did not further increase after 5 h. In contrast, IMCL concentrations were not altered during the postprandial period (P = 0.74). The addition of protein to a single high-fat meal did not change the postprandial accumulation of fat in the liver (P = 0.93) or skeletal muscle (P = 0.84). CONCLUSIONS In this study, we showed that a single energy-dense, high-fat meal induced net lipid accumulation in the liver, which was detected by using in vivo ¹H-MRS. This noninvasive approach might bring new opportunities to study postprandial hepatic lipid dynamics. The addition of protein did not change the ectopic lipid retention after a single high-fat meal.
Collapse
Affiliation(s)
- Lucas Lindeboom
- From the Departments of Radiology (LL, CIN, JEW, and VBS-H), Human Biology (LL, PS, and VBS-H), and Human Movement Sciences (CIN and MKCH), NUTRIM, Maastricht University Medical Center, Maastricht, The Netherlands, and Top Institute Food and Nutrition (LL, CIN, PS, and VBS-H), Wageningen, The Netherlands
| | - Christine I Nabuurs
- From the Departments of Radiology (LL, CIN, JEW, and VBS-H), Human Biology (LL, PS, and VBS-H), and Human Movement Sciences (CIN and MKCH), NUTRIM, Maastricht University Medical Center, Maastricht, The Netherlands, and Top Institute Food and Nutrition (LL, CIN, PS, and VBS-H), Wageningen, The Netherlands
| | - Matthijs K C Hesselink
- From the Departments of Radiology (LL, CIN, JEW, and VBS-H), Human Biology (LL, PS, and VBS-H), and Human Movement Sciences (CIN and MKCH), NUTRIM, Maastricht University Medical Center, Maastricht, The Netherlands, and Top Institute Food and Nutrition (LL, CIN, PS, and VBS-H), Wageningen, The Netherlands
| | - Joachim E Wildberger
- From the Departments of Radiology (LL, CIN, JEW, and VBS-H), Human Biology (LL, PS, and VBS-H), and Human Movement Sciences (CIN and MKCH), NUTRIM, Maastricht University Medical Center, Maastricht, The Netherlands, and Top Institute Food and Nutrition (LL, CIN, PS, and VBS-H), Wageningen, The Netherlands
| | - Patrick Schrauwen
- From the Departments of Radiology (LL, CIN, JEW, and VBS-H), Human Biology (LL, PS, and VBS-H), and Human Movement Sciences (CIN and MKCH), NUTRIM, Maastricht University Medical Center, Maastricht, The Netherlands, and Top Institute Food and Nutrition (LL, CIN, PS, and VBS-H), Wageningen, The Netherlands
| | - Vera B Schrauwen-Hinderling
- From the Departments of Radiology (LL, CIN, JEW, and VBS-H), Human Biology (LL, PS, and VBS-H), and Human Movement Sciences (CIN and MKCH), NUTRIM, Maastricht University Medical Center, Maastricht, The Netherlands, and Top Institute Food and Nutrition (LL, CIN, PS, and VBS-H), Wageningen, The Netherlands
| |
Collapse
|
30
|
MacPherson REK, Castelli LM, Miotto PM, Frendo-Cumbo S, Milburn A, Roy BD, LeBlanc PJ, Ward WE, Peters SJ. A maternal high fat diet has long-lasting effects on skeletal muscle lipid and PLIN protein content in rat offspring at young adulthood. Lipids 2015; 50:205-17. [PMID: 25552350 DOI: 10.1007/s11745-014-3985-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 12/16/2014] [Indexed: 01/08/2023]
Abstract
A maternal high fat diet (HFD) can have adverse effects on skeletal muscle development. Skeletal muscle PLIN proteins (PLIN2, 3 and 5) are thought to play critical roles in lipid metabolism, however effects of HFD on PLIN and lipases (HSL, ATGL, CGI-58) in mothers as well as their offspring have yet to be investigated. The primary objective of this study was to determine whether maternal HFD would influence skeletal muscle lipase and PLIN protein content in offspring at weaning (19 d) and young adulthood (3 mo). Female rats (28 d old, n = 9/group) were fed control (CON, AIN93G, 7% soybean oil) or HFD (AIN93G, 20% lard) for 10 weeks prior to mating and throughout pregnancy and lactation. All offspring were weaned to CON [n = 18/group, 1 female and 1 male pup per litter were studied at weaning (19 d) and 3 mo of age]. There was no effect of sex for the main outcomes measured in plantaris, therefore male and female data was combined. Maternal HFD resulted in higher triacylglycerol content in pups at 3 mo (p < 0.05), as well as in the dams (p = 0.015). Maternal HFD resulted in higher PLIN5 content in pups at weaning and 3 mo (p = 0.05). PLIN2 and PLIN5 content decreased at 3 mo versus weaning (p < 0.001). HFD dams had a higher PLIN3 content (p = 0.016). Diet had no effect on ATGL, CGI-58, or HSL content. In conclusion, exposure to a maternal HFD resulted in higher skeletal muscle lipid and PLIN5 content in plantaris of offspring through to young adulthood.
Collapse
Affiliation(s)
- Rebecca E K MacPherson
- Faculty of Applied Health Sciences, Center for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada,
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yokoyama Y, Barnard ND, Levin SM, Watanabe M. Vegetarian diets and glycemic control in diabetes: a systematic review and meta-analysis. Cardiovasc Diagn Ther 2014; 4:373-82. [PMID: 25414824 DOI: 10.3978/j.issn.2223-3652.2014.10.04] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 10/08/2014] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Previous studies have suggested an association between vegetarian diets and improvements in glycemic control in diabetes, although this relationship is not well established. No meta-analysis of these studies has been performed. METHODS To conduct a systematic review and meta-analysis of controlled clinical trials examining the association between vegetarian diets and glycemic control in type 2 diabetes. DATA SOURCE The electronic databases Medline, Web of Science, Excerpta Medica Database (EMBASE), and Cochrane Central Register of Controlled Trials were searched for articles published in any language through December 9, 2013. STUDY SELECTION The following criteria were used for study inclusion: (I) age of participants >20 years; (II) vegetarian diet as intervention; (III) mean difference in hemoglobin A1c (HbA1c) and/or fasting blood glucose levels used as outcomes; and (IV) controlled trials, duration ≥4 weeks. Exclusion criteria were: (I) not an original investigation; (II) duplicate samples; (III) diabetes other than type 2; (IV) multiple interventions; and (V) uncontrolled studies. DATA EXTRACTION AND SYNTHESIS The data collected included study design, baseline population characteristics, dietary data, and outcomes. Data were pooled using a random-effects model. MAIN OUTCOMES AND MEASURES Differences in HbA1c and fasting blood glucose levels associated with vegetarian diets were assessed. RESULTS Of 477 studies identified, six met the inclusion criteria (n=255, mean age 42.5 years). Consumption of vegetarian diets was associated with a significant reduction in HbA1c [-0.39 percentage point; 95% confidence interval (CI), -0.62 to -0.15; P=0.001; I(2)=3.0; P for heterogeneity =0.389], and a non-significant reduction in fasting blood glucose concentration (-0.36 mmol/L; 95% CI, -1.04 to 0.32; P=0.301; I(2)=0; P for heterogeneity =0.710), compared with consumption of comparator diets. CONCLUSIONS Consumption of vegetarian diets is associated with improved glycemic control in type 2 diabetes. PROSPERO registration number is CRD42013004370.
Collapse
Affiliation(s)
- Yoko Yokoyama
- 1 Graduate School of Media and Governance, Keio University, 5322 Endo, Fujisawa, Kanagawa, 252-0882, Japan ; 2 Department of Medicine, The George Washington University School of Medicine, Washington, DC, USA ; 3 Physicians Committee for Responsible Medicine, Washington, DC, USA ; 4 Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan ; 5 Health Science Laboratory, Graduate School of Media and Governance, Faculty of Environment and Information Studies, Keio University, 5322 Endo, Fujisawa, Kanagawa, 252-0882, Japan
| | - Neal D Barnard
- 1 Graduate School of Media and Governance, Keio University, 5322 Endo, Fujisawa, Kanagawa, 252-0882, Japan ; 2 Department of Medicine, The George Washington University School of Medicine, Washington, DC, USA ; 3 Physicians Committee for Responsible Medicine, Washington, DC, USA ; 4 Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan ; 5 Health Science Laboratory, Graduate School of Media and Governance, Faculty of Environment and Information Studies, Keio University, 5322 Endo, Fujisawa, Kanagawa, 252-0882, Japan
| | - Susan M Levin
- 1 Graduate School of Media and Governance, Keio University, 5322 Endo, Fujisawa, Kanagawa, 252-0882, Japan ; 2 Department of Medicine, The George Washington University School of Medicine, Washington, DC, USA ; 3 Physicians Committee for Responsible Medicine, Washington, DC, USA ; 4 Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan ; 5 Health Science Laboratory, Graduate School of Media and Governance, Faculty of Environment and Information Studies, Keio University, 5322 Endo, Fujisawa, Kanagawa, 252-0882, Japan
| | - Mitsuhiro Watanabe
- 1 Graduate School of Media and Governance, Keio University, 5322 Endo, Fujisawa, Kanagawa, 252-0882, Japan ; 2 Department of Medicine, The George Washington University School of Medicine, Washington, DC, USA ; 3 Physicians Committee for Responsible Medicine, Washington, DC, USA ; 4 Department of Internal Medicine, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan ; 5 Health Science Laboratory, Graduate School of Media and Governance, Faculty of Environment and Information Studies, Keio University, 5322 Endo, Fujisawa, Kanagawa, 252-0882, Japan
| |
Collapse
|
32
|
Ageing, adipose tissue, fatty acids and inflammation. Biogerontology 2014; 16:235-48. [PMID: 25367746 DOI: 10.1007/s10522-014-9536-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 10/20/2014] [Indexed: 12/22/2022]
Abstract
A common feature of ageing is the alteration in tissue distribution and composition, with a shift in fat away from lower body and subcutaneous depots to visceral and ectopic sites. Redistribution of adipose tissue towards an ectopic site can have dramatic effects on metabolic function. In skeletal muscle, increased ectopic adiposity is linked to insulin resistance through lipid mediators such as ceramide or DAG, inhibiting the insulin receptor signalling pathway. Additionally, the risk of developing cardiovascular disease is increased with elevated visceral adipose distribution. In ageing, adipose tissue becomes dysfunctional, with the pathway of differentiation of preadipocytes to mature adipocytes becoming impaired; this results in dysfunctional adipocytes less able to store fat and subsequent fat redistribution to ectopic sites. Low grade systemic inflammation is commonly observed in ageing, and may drive the adipose tissue dysfunction, as proinflammatory cytokines are capable of inhibiting adipocyte differentiation. Beyond increased ectopic adiposity, the effect of impaired adipose tissue function is an elevation in systemic free fatty acids (FFA), a common feature of many metabolic disorders. Saturated fatty acids can be regarded as the most detrimental of FFA, being capable of inducing insulin resistance and inflammation through lipid mediators such as ceramide, which can increase risk of developing atherosclerosis. Elevated FFA, in particular saturated fatty acids, maybe a driving factor for both the increased insulin resistance, cardiovascular disease risk and inflammation in older adults.
Collapse
|
33
|
Lee HS, Nam Y, Chung YH, Kim HR, Park ES, Chung SJ, Kim JH, Sohn UD, Kim HC, Oh KW, Jeong JH. Beneficial effects of phosphatidylcholine on high-fat diet-induced obesity, hyperlipidemia and fatty liver in mice. Life Sci 2014; 118:7-14. [PMID: 25445436 DOI: 10.1016/j.lfs.2014.09.027] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 02/07/2023]
Abstract
AIMS Soybean-derived PC is an essential cell membrane phospholipid that is composed of unsaturated fatty acids, including oleic acid. The present study aimed to evaluate the potential alleviation effects of soybean PC on high fat diet (HFD)-induced obesity and its related complications. MAIN METHODS We fed C57BL/6 mice a HFD for 12 weeks and administered PC orally for 8 or 12 weeks at different doses. At the end of the experiment, blood was prepared for biochemical analysis and leptin ELISA. Aorta, epididymal and mesenteric fat and liver were removed surgically, weighed and observed for histological or immunohistochemical changes. KEY FINDINGS PC significantly prevented body weight gain and lipid accumulation and alleviated hyperlipidemia by decreasing triglyceride (TG) and total cholesterol (TC) levels and the atherogenic index in serum or by increasing the HDL/TC ratio. Aortic apoE expression and serum leptin levels were suppressed by PC treatment in the HFD-induced obese mouse model. Elevated serum aspartate aminotransferase and alanine aminotransferase levels in HFD-fed mice were decreased in the PC groups. PC treatment significantly decreased HFD-induced liver weight and hepatic lipid accumulation. SIGNIFICANCE PC treatment alleviated HFD-induced obese status and obesity-related complications such as hyperlipidemic changes that induce cardiovascular disease and NAFLD.
Collapse
Affiliation(s)
- Ho Sung Lee
- Department of Pharmacology, College of Medicine, Chung-ang University, Seoul 156-756, Republic of Korea
| | - Yunsung Nam
- Department of Pharmacology, College of Medicine, Chung-ang University, Seoul 156-756, Republic of Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-ang University, Seoul 156-756, Republic of Korea
| | - Hak Rim Kim
- Department of Pharmacology, College of Medicine, Dankook University, Chungnam, Republic of Korea
| | - Eon Sub Park
- Departments of Pathology, College of Medicine, Chung-ang University, Seoul 156-756, Republic of Korea
| | - Su Jin Chung
- Department of Pharmacology, College of Medicine, Chung-ang University, Seoul 156-756, Republic of Korea
| | - Jung Hyo Kim
- Department of Pharmacology, College of Pharmacy, Chung-ang University, Seoul 156-756, Republic of Korea
| | - Uy Dong Sohn
- Department of Pharmacology, College of Pharmacy, Chung-ang University, Seoul 156-756, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, Republic of Korea
| | - Keon Woong Oh
- Departments of Pathology, College of Medicine, Chung-ang University, Seoul 156-756, Republic of Korea; Christmas clinic, Sin-gal-dong, Giheung-gu, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-ang University, Seoul 156-756, Republic of Korea.
| |
Collapse
|
34
|
Jordy AB, Serup AK, Karstoft K, Pilegaard H, Kiens B, Jeppesen J. Insulin sensitivity is independent of lipid binding protein trafficking at the plasma membrane in human skeletal muscle: effect of a 3-day, high-fat diet. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1136-45. [PMID: 25163924 DOI: 10.1152/ajpregu.00124.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of the present study was to investigate lipid-induced regulation of lipid binding proteins in human skeletal muscle and the impact hereof on insulin sensitivity. Eleven healthy male subjects underwent a 3-day hypercaloric and high-fat diet regime. Muscle biopsies were taken before and after the diet intervention, and giant sarcolemmal vesicles were prepared. The high-fat diet induced decreased insulin sensitivity, but this was not associated with a relocation of FAT/CD36 or FABPpm protein to the sarcolemma. However, FAT/CD36 and FABPpm mRNA, but not the proteins, were upregulated by increased fatty acid availability. This suggests a time dependency in the upregulation of FAT/CD36 and FABPpm protein during high availability of plasma fatty acids. Furthermore, we did not detect FATP1 and FATP4 protein in giant sarcolemmal vesicles obtained from human skeletal muscle. In conclusion, this study shows that a short-term lipid-load increases mRNA content of key lipid handling proteins in human muscle. However, decreased insulin sensitivity after a high-fat diet is not accompanied with relocation of FAT/CD36 or FABPpm protein to the sarcolemma. Finally, FATP1 and FATP4 protein was located intracellularly but not at the sarcolemma in humans.
Collapse
Affiliation(s)
- Andreas B Jordy
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Annette K Serup
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Karstoft
- The Centre of Inflammation and Metabolism and The Centre for Physical Activity Research, Department of Infectious Diseases, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; and
| | - Henriette Pilegaard
- The Centre of Inflammation and Metabolism and The Centre for Physical Activity Research, Department of Infectious Diseases, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; and CFAS, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Bente Kiens
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark;
| | - Jacob Jeppesen
- Section of Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
35
|
Rinnankoski-Tuikka R, Hulmi JJ, Torvinen S, Silvennoinen M, Lehti M, Kivelä R, Reunanen H, Kujala UM, Kainulainen H. Lipid droplet-associated proteins in high-fat fed mice with the effects of voluntary running and diet change. Metabolism 2014; 63:1031-40. [PMID: 24972504 DOI: 10.1016/j.metabol.2014.05.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 05/12/2014] [Accepted: 05/22/2014] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The relation between lipid accumulation and influence of exercise on insulin sensitivity is not straightforward. A proper balance between lipid droplet synthesis, lipolysis, and oxidative metabolism would ensure low local intramyocellular fatty acid levels, thereby possibly protecting against lipotoxicity-associated insulin resistance. This study investigated whether the accumulation of triglycerides and lipid droplets in response to high availability of fatty acids after high-fat feeding would parallel the abundance of intramyocellular perilipin proteins, especially PLIN5. The effects on these variables after diet change or voluntary running exercise intervention in skeletal muscle were also investigated. METHODS During a 19-week experiment, C57BL/6J mice were studied in six different groups: low-fat diet sedentary, low-fat diet active, high-fat diet sedentary, high-fat diet active and two groups which were high-fat sedentary for nine weeks, after which divided into low-fat sedentary or low-fat active groups. Myocellular triglyceride concentration and perilipin protein expression levels were assessed. RESULTS We show that, concurrently with impaired insulin sensitivity, the expression level of PLIN5 and muscular triglyceride concentration increased dramatically after high-fat diet. These adaptations were reversible after the diet change intervention with no additional effect of exercise. CONCLUSIONS After high-fat diet, lipid droplets become larger providing more surface area for PLIN5. We suggest that PLIN5 is an important regulator of lipid droplet turnover in altered conditions of fatty acid supply and consumption. Imbalances in lipid droplet metabolism and turnover might lead to lipotoxicity-related insulin resistance.
Collapse
Affiliation(s)
- Rita Rinnankoski-Tuikka
- Neuromuscular Research Center, Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland
| | - Juha J Hulmi
- Neuromuscular Research Center, Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland
| | - Sira Torvinen
- Neuromuscular Research Center, Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland
| | - Mika Silvennoinen
- Neuromuscular Research Center, Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland
| | - Maarit Lehti
- LIKES Research Center for Sport and Health Sciences, Jyväskylä, Finland
| | - Riikka Kivelä
- Neuromuscular Research Center, Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland
| | - Hilkka Reunanen
- Department of Biological and Environmental Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Urho M Kujala
- Department of Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Heikki Kainulainen
- Neuromuscular Research Center, Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland.
| |
Collapse
|
36
|
Sakurai Y, Tamura Y, Takeno K, Kumashiro N, Sato F, Kakehi S, Ikeda S, Ogura Y, Saga N, Naito H, Katamoto S, Fujitani Y, Hirose T, Kawamori R, Watada H. Determinants of intramyocellular lipid accumulation after dietary fat loading in non-obese men. J Diabetes Investig 2014; 2:310-7. [PMID: 24843504 PMCID: PMC4014973 DOI: 10.1111/j.2040-1124.2010.00091.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Aims/Introduction: Accumulation of intramyocellular lipid (IMCL) is associated with insulin resistance. However, the factors affecting the change in IMCL remain to be elucidated. The aim of the present study was to determine the factors that influence the change in IMCL level after high‐fat loading. Materials and Methods: The study subjects were 37 non‐obese men. Each subject consumed a high‐fat diet for 3 days after a normal‐fat diet for 3 days. After each diet program, IMCL levels in the tibialis anterior (TA‐IMCL) and soleus (SOL‐IMCL) were measured by proton magnetic resonance spectroscopy. Glucose infusion rate (GIR) was evaluated by euglycemic hyperinsulinemic clamp as an index of peripheral insulin sensitivity. Results: The high‐fat diet significantly increased TA‐IMCL and SOL‐IMCL by ∼30 and ∼20%, respectively (P < 0.05), whereas it did not significantly alter GIR. The increase in SOL‐IMCL, but not in TA‐IMCL, negatively correlated with serum high molecular weight (HMW)‐adiponectin (r = −0.36, P < 0.05) and HMW‐/total‐adiponectin ratio (r = −0.46, P < 0.05). Although high‐fat diet‐related changes in SOL‐IMCL showed high inter‐individual variations, in subjects doing exercise, changes in SOL‐IMCL (r = 0.55, P < 0.05) and TA‐IMCL (r = 0.61, P < 0.05) positively correlated with daily physical activity level. In contrast, in sedentary subjects, changes in SOL‐IMCL (r = −0.50, P < 0.01) and TA‐IMCL (r = −0.48, P < 0.05) negatively correlated with daily physical activity. Conclusions: HMW‐adiponectin and daily physical activity are determinants of IMCL accumulation by a high‐fat diet. Intriguingly, the effect of daily physical activity on the change in IMCL depends on the level of regular exercise. (J Diabetes Invest,doi: 10.1111/j.2040‐1124.2010.00091.x, 2011)
Collapse
Affiliation(s)
- Yuko Sakurai
- Department of Medicine, Metabolism and Endocrinology
| | - Yoshifumi Tamura
- Department of Medicine, Metabolism and Endocrinology ; Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo
| | | | | | - Fumihiko Sato
- Department of Medicine, Metabolism and Endocrinology
| | - Saori Kakehi
- Department of Medicine, Metabolism and Endocrinology ; Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo
| | - Shinichi Ikeda
- Department of Medicine, Metabolism and Endocrinology ; Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo
| | - Yuji Ogura
- Institute of Health and Sports Science and Medicine ; Department of Exercise Physiology, Graduate School of Health and Sports Science, Juntendo University, Chiba
| | - Norio Saga
- Institute of Health and Sports Science and Medicine ; Department of Exercise Physiology, Graduate School of Health and Sports Science, Juntendo University, Chiba
| | - Hisashi Naito
- Institute of Health and Sports Science and Medicine ; Department of Exercise Physiology, Graduate School of Health and Sports Science, Juntendo University, Chiba
| | - Shizuo Katamoto
- Institute of Health and Sports Science and Medicine ; Department of Exercise Physiology, Graduate School of Health and Sports Science, Juntendo University, Chiba
| | | | | | - Ryuzo Kawamori
- Department of Medicine, Metabolism and Endocrinology ; Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo ; Center for Therapeutic Innovations in Diabetes ; Center for Beta Cell Biology and Regeneration, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Hirotaka Watada
- Department of Medicine, Metabolism and Endocrinology ; Sportology Center, Graduate School of Medicine, Juntendo University, Tokyo
| |
Collapse
|
37
|
Metabolic profile before and after short-term overfeeding with a high-fat diet: a comparison between South Asian and White men. Br J Nutr 2014; 111:1853-61. [PMID: 24506887 DOI: 10.1017/s0007114514000014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
For the same BMI, South Asians have a higher body fat percentage and an adverse metabolic profile compared with whites. The objective of the present study was to determine the metabolic profiles of South Asian and white men matched for body fat percentage in response to short-term overfeeding with a high-fat diet. A total of ten healthy non-diabetic South Asian men matched for body fat percentage with ten white men were included in the study. A weight-maintenance diet (containing 30 % fat, 55 % carbohydrate and 15 % protein) was provided for 3 d followed by 4 d of overfeeding (150 % of energy requirement) with a high-fat diet (60 % fat, 25 % carbohydrate and 15 % protein). Before and after the overfeeding period, plasma glucose, insulin, TAG, NEFA, total cholesterol and HDL-cholesterol concentrations were determined. Glucose clearance was calculated using a 2 h oral glucose tolerance test. The results revealed that in South Asian and white men, respectively, overfeeding with a high-fat diet decreased plasma TAG concentrations by 0·4 (sd 0·6) and 0·4 (sd 0·5) mmol/l (P diet= 0·008; P ethnicity= 0·24), increased HDL-cholesterol concentrations by 0·12 (sd 0·1) and 0·14 (sd 0·2) mmol/l (P diet= 0·001; P ethnicity= 0·06) and decreased glucose clearance by 48·8 (sd 53·5) and 37·2 (sd 34·2) ml/min per m2 body surface (P diet= 0·004; P ethnicity= 0·18). There was a significant interaction between diet and ethnicity with regard to the changes in total and LDL-cholesterol concentrations (P= 0·01 and 0·007, respectively), which trended towards a larger increase in South Asian subjects than in white subjects. Despite a similar body fat percentage, short-term overfeeding with a high-fat diet had more adverse effects on the lipid profile of South Asians than on that of whites.
Collapse
|
38
|
Ross JS, Hu W, Rosen B, Snider AJ, Obeid LM, Cowart LA. Sphingosine kinase 1 is regulated by peroxisome proliferator-activated receptor α in response to free fatty acids and is essential for skeletal muscle interleukin-6 production and signaling in diet-induced obesity. J Biol Chem 2013; 288:22193-206. [PMID: 23766515 PMCID: PMC3829312 DOI: 10.1074/jbc.m113.477786] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/06/2013] [Indexed: 12/25/2022] Open
Abstract
We previously demonstrated that sphingosine kinase 1 (Sphk1) expression and activity are up-regulated by exogenous palmitate (PAL) in a skeletal muscle model system and in diet-induced obesity in mice; however, potential functions and in vivo relevance of this have not been addressed. Here, we aimed to determine the mechanism by which PAL regulates SphK1 in muscle, and to determine potential roles for its product, sphingosine-1-phosphate (S1P), in muscle biology in the context of obesity. Cloning and analysis of the mouse Sphk1 promoter revealed a peroxisome proliferator-activated receptor (PPAR) α cis-element that mediated activation of a reporter under control of the Sphk1 promoter; direct interaction of PPARα was demonstrated by chromatin immunoprecipitation. PAL treatment induced the proinflammatory cytokine interleukin (IL)-6 in a manner dependent on SphK1, and this was attenuated by inhibition of the sphingosine-1-phosphate receptor 3 (S1PR3). Diet-induced obesity in mice demonstrated that IL-6 expression in muscle, but not adipose tissue, increased in obesity, but this was attenuated in Sphk1(-/-) mice. Moreover, plasma IL-6 levels were significantly decreased in obese Sphk1(-/-) mice relative to obese wild type mice, and muscle, but not adipose tissue IL-6 signaling was activated. These data indicate that PPARα regulates Sphk1 expression in the context of fatty acid oversupply and links PAL to muscle IL-6 production. Moreover, this function of SphK1 in diet-induced obesity suggests a potential role for SphK1 in obesity-associated pathological outcomes.
Collapse
Affiliation(s)
- Jessica S. Ross
- From the Departments of Biochemistry and Molecular Biology and
- Molecular and Cellular Biology and Pathobiology Program, and
| | - Wei Hu
- From the Departments of Biochemistry and Molecular Biology and
| | - Bess Rosen
- the Boston University School of Medicine, Center for Regenerative Medicine, Boston, Massachusetts 02118
| | - Ashley J. Snider
- Medicine, Medical University of South Carolina, Charleston, South Carolina 29425
- the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| | - Lina M. Obeid
- the Department of Medicine, Stony Brook University, Stony Brook, New York 11790
- the Northport Veterans Affairs Medical Center, Northpoint, New York 11768, and
| | - L. Ashley Cowart
- From the Departments of Biochemistry and Molecular Biology and
- the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| |
Collapse
|
39
|
Marjańska M, Eberly LE, Adriany G, Verdoliva SN, Garwood M, Chow L. Influence of foot orientation on the appearance and quantification of 1H magnetic resonance muscle spectra obtained from the soleus and the vastus lateralis. Magn Reson Med 2012; 68:1731-7. [PMID: 22298295 PMCID: PMC3381854 DOI: 10.1002/mrm.24198] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 01/09/2011] [Accepted: 01/12/2011] [Indexed: 12/21/2022]
Abstract
Muscle (1)H magnetic resonance spectroscopy noninvasively measures intramyocellular lipid levels, which correlate with obesity, insulin resistance, and type 2 diabetes. The appearance of muscle magnetic resonance spectra is influenced by bulk magnetic susceptibility and residual dipolar couplings which depend on the angle between the muscle fibers and the main magnetic field. This study used a 4 T magnet to examine the influence of foot orientation on the appearance and quantification of muscle magnetic resonance spectra from the soleus and the vastus lateralis. For each individual, intramyocellular lipid, extramyocellular lipid, and creatine concentrations were quantified in the soleus and the vastus lateralis during one session. Foot orientation was found to influence the appearance of muscle spectra from the soleus but not from the vastus lateralis. It was concluded that quantifying intramyocellular lipid by the standard LCModel using a water reference may be more appropriate than using a creatine reference in the presence of residual dipolar couplings.
Collapse
Affiliation(s)
- Małgorzata Marjańska
- Center for Magnetic Resonance Research and Department of Radiology, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Hill BG, Benavides GA, Lancaster JR, Ballinger S, Dell’Italia L, Zhang J, Darley-Usmar VM. Integration of cellular bioenergetics with mitochondrial quality control and autophagy. Biol Chem 2012; 393:1485-1512. [PMID: 23092819 PMCID: PMC3594552 DOI: 10.1515/hsz-2012-0198] [Citation(s) in RCA: 361] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Accepted: 06/22/2012] [Indexed: 02/06/2023]
Abstract
Bioenergetic dysfunction is emerging as a cornerstone for establishing a framework for understanding the pathophysiology of cardiovascular disease, diabetes,cancer and neurodegeneration. Recent advances in cellular bioenergetics have shown that many cells maintain a substantial bioenergetic reserve capacity, which is a prospective index of ‘ healthy ’ mitochondrial populations.The bioenergetics of the cell are likely regulated by energy requirements and substrate availability. Additionally,the overall quality of the mitochondrial population and the relative abundance of mitochondria in cells and tissues also impinge on overall bioenergetic capacity and resistance to stress. Because mitochondria are susceptible to damage mediated by reactive oxygen/nitrogen and lipid species, maintaining a ‘ healthy ’ population of mitochondria through quality control mechanisms appears to be essential for cell survival under conditions of pathological stress. Accumulating evidence suggest that mitophagy is particularly important for preventing amplification of initial oxidative insults, which otherwise would further impair the respiratory chain or promote mutations in mitochondrial DNA (mtDNA). The processes underlying the regulation of mitophagy depend on several factors, including the integrity of mtDNA, electron transport chain activity, and the interaction and regulation of the autophagic machinery. The integration and interpretation of cellular bioenergetics in the context of mitochondrial quality control and genetics is the theme of this review.
Collapse
Affiliation(s)
- Bradford G. Hill
- Diabetes and Obesity Center, Institute of Molecular Cardiology, and Department of Medicine, University of Louisville, Louisville, KY
- Departments of Biochemistry and Molecular Biology and Physiology and Biophysics, University of Louisville, Louisville, KY
| | - Gloria A. Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jack R. Lancaster
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Medicine, Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Lou Dell’Italia
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Medicine, Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Victor M. Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Medicine, Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
41
|
Jacobsen SC, Brøns C, Bork-Jensen J, Ribel-Madsen R, Yang B, Lara E, Hall E, Calvanese V, Nilsson E, Jørgensen SW, Mandrup S, Ling C, Fernandez AF, Fraga MF, Poulsen P, Vaag A. Effects of short-term high-fat overfeeding on genome-wide DNA methylation in the skeletal muscle of healthy young men. Diabetologia 2012; 55:3341-9. [PMID: 22961225 DOI: 10.1007/s00125-012-2717-8] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 08/08/2012] [Indexed: 10/27/2022]
Abstract
AIMS/HYPOTHESIS Energy-dense diets that are high in fat are associated with a risk of metabolic diseases. The underlying molecular mechanisms could involve epigenetics, as recent data show altered DNA methylation of putative type 2 diabetes candidate genes in response to high-fat diets. We examined the effect of a short-term high-fat overfeeding (HFO) diet on genome-wide DNA methylation patterns in human skeletal muscle. METHODS Skeletal muscle biopsies were obtained from 21 healthy young men after ingestion of a short-term HFO diet and a control diet, in a randomised crossover setting. DNA methylation was measured in 27,578 CpG sites/14,475 genes using Illumina's Infinium Bead Array. Candidate gene expression was determined by quantitative real-time PCR. RESULTS HFO introduced widespread DNA methylation changes affecting 6,508 genes (45%), with a maximum methylation change of 13.0 percentage points. The HFO-induced methylation changes were only partly and non-significantly reversed after 6-8 weeks. Alterations in DNA methylation levels primarily affected genes involved in inflammation, the reproductive system and cancer. Few gene expression changes were observed and these had poor correlation to DNA methylation. CONCLUSIONS/INTERPRETATION The genome-wide DNA methylation changes induced by the short-term HFO diet could have implications for our understanding of transient epigenetic regulation in humans and its contribution to the development of metabolic diseases. The slow reversibility suggests a methylation build-up with HFO, which over time may influence gene expression levels.
Collapse
|
42
|
Hoeks J, Schrauwen P. Muscle mitochondria and insulin resistance: a human perspective. Trends Endocrinol Metab 2012; 23:444-50. [PMID: 22726362 DOI: 10.1016/j.tem.2012.05.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/16/2012] [Accepted: 05/21/2012] [Indexed: 01/07/2023]
Abstract
Reduced mitochondrial capacity in skeletal muscle has been suggested to underlie the development of insulin resistance and type 2 diabetes mellitus (T2DM). However, data obtained from human subjects concerning this putative relation indicate that the mitochondrial defect observed in diabetic muscle might be secondary to the insulin-resistant state instead of being a causal factor. Nonetheless, diminished mitochondrial function, even secondary to insulin resistance, may accelerate lipid deposition in non-adipose tissues and aggravate insulin resistance. Indeed, improving mitochondrial capacity via exercise training and calorie restriction is associated with positive metabolic health effects. Here we review muscle mitochondrial dysfunction in humans and propose that targeting muscle mitochondria to improve muscle oxidative capacity should be considered as a strategy for improving metabolic health.
Collapse
Affiliation(s)
- Joris Hoeks
- NUTRIM - School for Nutrition, Toxicology and Metabolism, Department of Human Biology, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
| | | |
Collapse
|
43
|
Sions JM, Tyrell CM, Knarr BA, Jancosko A, Binder-Macleod SA. Age- and stroke-related skeletal muscle changes: a review for the geriatric clinician. J Geriatr Phys Ther 2012; 35:155-61. [PMID: 22107952 PMCID: PMC3290755 DOI: 10.1519/jpt.0b013e318236db92] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Independently, aging and stroke each have a significant negative impact on skeletal muscle, but the potential cumulative effects of aging and stroke have not been explored. Optimal interventions for individuals post stroke may include those that specifically target skeletal muscle. Addressing changes in muscles may minimize activity limitations and enhance participation post stroke. This article reviews the impact of aging and stroke on muscle morphology and composition, including fiber atrophy, reductions in muscle cross-sectional area, changes in muscle fiber distributions, and increases in intramuscular fat. Relationships between changes in muscle structure, muscle function, and physical mobility are reviewed. Clinical recommendations that preserve and enhance skeletal muscle in the aging adult and individuals post stroke are discussed. Future research directions that include systematic comparison of the differences in skeletal muscle between younger and older adults who have sustained a stroke are suggested.
Collapse
Affiliation(s)
- Jaclyn Megan Sions
- Biomechanics and Movement Science Program, University of Delaware, Newark, USA.
| | | | | | | | | |
Collapse
|
44
|
Gjelstad IMF, Haugen F, Gulseth HL, Norheim F, Jans A, Bakke SS, Raastad T, Tjønna AE, Wisløff U, Blaak EE, Risérus U, Gaster M, Roche HM, Birkeland KI, Drevon CA. Expression of perilipins in human skeletal muscle in vitro and in vivo in relation to diet, exercise and energy balance. Arch Physiol Biochem 2012; 118:22-30. [PMID: 22117101 DOI: 10.3109/13813455.2011.630009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The perilipin proteins enclose intracellular lipid droplets. We describe the mRNA expression of the five perilipins in human skeletal muscle in relation to fatty acid supply, exercise and energy balance. We observed that all perilipins were expressed in skeletal muscle biopsies with the highest mRNA levels of perilipin 2, 4 and 5. Cultured myotubes predominantly expressed perilipin 2 and 3. In vitro, incubation of myotubes with fatty acids enhanced mRNA expression of perilipin 1, 2 and 4. In vivo, low fat diet increased mRNA levels of perilipin 3 and 4. Endurance training, but not strength training, enhanced the expression of perilipin 2 and 3. Perilipin 1 mRNA correlated positively with body fat mass, whereas none of the perilipins were associated with insulin sensitivity. In conclusion, all perilipins mRNAs were expressed in human skeletal muscle. Diet as well as endurance exercise modulated the expression of perilipins.
Collapse
Affiliation(s)
- I M F Gjelstad
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Postprandial metabolism of meal triglyceride in humans. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1821:721-6. [PMID: 22281699 DOI: 10.1016/j.bbalip.2012.01.006] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 01/05/2012] [Accepted: 01/06/2012] [Indexed: 12/20/2022]
Abstract
The intake of dietary fat above energy needs has contributed to the growing rates of obesity worldwide. The concept of disease development occurring in the fed state now has much support and dysregulation of substrate flux may occur due to poor handling of dietary fat in the immediate postprandial period. The present paper will review recent observations implicating cephalic phase events in the control of enterocyte lipid transport, the impact of varying the composition of meals on subsequent fat metabolism, and the means by which dietary lipid carried in chylomicrons can lead to elevated postprandial non-esterified fatty acid concentrations. This discussion is followed by an evaluation of the data on quantitative meal fat oxidation at the whole body level and an examination of dietary fat clearance to peripheral tissues - with particular attention paid to skeletal muscle and liver given the role of ectopic lipid deposition in insulin resistance. Estimates derived from data of dietary-TG clearance show good agreement with clearance to the liver equaling 8-12% of meal fat in lean subjects and this number appears higher (10-16%) in subjects with diabetes and fatty liver disease. Finally, we discuss new methods with which to study dietary fatty acid partitioning in vivo. Future research is needed to include a more comprehensive understanding of 1) the potential for differential oxidation of saturated versus unsaturated fatty acids which might lead to meaningful energy deficit and whether this parameter varies based on insulin sensitivity, 2) whether compartmentalization exists for diet-derived fatty acids within tissues vs. intracellular pools, and 3) the role of reduced peripheral fatty acid clearance in the development of fatty liver disease. Further advancements in the quantitation of dietary fat absorption and disposal will be central to the development of therapies designed to treat diet-induced obesity. This article is part of a Special Issue entitled Triglyceride Metabolism and Disease.
Collapse
|
46
|
Bergouignan A, Gozansky WS, Barry DW, Leitner W, MacLean PS, Hill JO, Draznin B, Melanson EL. Increasing dietary fat elicits similar changes in fat oxidation and markers of muscle oxidative capacity in lean and obese humans. PLoS One 2012; 7:e30164. [PMID: 22253914 PMCID: PMC3257254 DOI: 10.1371/journal.pone.0030164] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Accepted: 12/11/2011] [Indexed: 12/16/2022] Open
Abstract
In lean humans, increasing dietary fat intake causes an increase in whole-body fat oxidation and changes in genes that regulate fat oxidation in skeletal muscle, but whether this occurs in obese humans is not known. We compared changes in whole-body fat oxidation and markers of muscle oxidative capacity differ in lean (LN) and obese (OB) adults exposed to a 2-day high-fat (HF) diet. Ten LN (BMI = 22.5±2.5 kg/m², age = 30±8 yrs) and nine OB (BMI = 35.9±4.93 kg/m², 38±5 yrs, Mean±SD) were studied in a room calorimeter for 24hr while consuming isocaloric low-fat (LF, 20% of energy) and HF (50% of energy) diets. A muscle biopsy was obtained the next morning following an overnight fast. 24h respiratory quotient (RQ) did not significantly differ between groups (LN: 0.91±0.01; OB: 0.92±0.01) during LF, and similarly decreased during HF in LN (0.86±0.01) and OB (0.85±0.01). The expression of pyruvate dehydrogenase kinase 4 (PDK4) and the fatty acid transporter CD36 increased in both LN and OB during HF. No other changes in mRNA or protein were observed. However, in both LN and OB, the amounts of acetylated peroxisome proliferator-activated receptor γ coactivator-1-α (PGC1-α) significantly decreased and phosphorylated 5-AMP-activated protein kinase (AMPK) significantly increased. In response to an isoenergetic increase in dietary fat, whole-body fat oxidation similarly increases in LN and OB, in association with a shift towards oxidative metabolism in skeletal muscle, suggesting that the ability to adapt to an acute increase in dietary fat is not impaired in obesity.
Collapse
Affiliation(s)
- Audrey Bergouignan
- Center for Human Nutrition, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
| | - Wendolyn S. Gozansky
- Division of Geriatric Medicine, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
| | - Daniel W. Barry
- Division of General Internal Medicine, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
| | - Wayne Leitner
- Division of Geriatric Medicine, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
| | - Paul S. MacLean
- Center for Human Nutrition, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
| | - James O. Hill
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
- Section of Nutrition, Department of Pediatrics, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
| | - Boris Draznin
- Division of Geriatric Medicine, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
| | - Edward L. Melanson
- Center for Human Nutrition, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
- Division of Endocrinology, Metabolism, and Diabetes, School of Medicine, University of Colorado Denver, Denver, Colorado, United States of America
- * E-mail:
| |
Collapse
|
47
|
Abstract
OBJECTIVE The skeletal muscle of obese humans is characterized by an inability to appropriately respond to alterations in substrate availability. The purpose of this study was to determine if this metabolic inflexibility with obesity is retained in mitochondria of human skeletal muscle cells raised in culture (HSkMC) and to identify potential mechanisms involved. DESIGN Mitochondrial respiration was measured in permeabilized myotubes cultured from lean and obese individuals before and after a 24-h lipid incubation. RESULTS Mitochondrial respiration (state 3) in the presence of lipid substrate (palmitoyl carnitine) increased by almost twofold after lipid incubation in HSkMC from lean, but not obese subjects, indicative of metabolic inflexibility with obesity. The 24-h lipid incubation increased mitochondrial DNA (mtDNA) copy number in HSkMC from lean subjects by +16% (P<0.05); conversely, mtDNA copy number decreased in myotubes cultured from obese individuals (-13%, P=0.06). When respiration data were normalized to mtDNA copy number and other indices of mitochondrial content (COX-IV protein content and CS activity), the significant treatment effects of lipid incubation persisted in the lean subjects, suggesting concomitant alterations in mitochondrial function; no similar adjustment was evident in HSkMC from obese individuals. CONCLUSION These data indicate that the skeletal muscle of obese individuals inherently lacks metabolic flexibility in response to lipid exposure, which consists of an inability to increase mitochondrial respiration in the presence of lipid substrate and perhaps by an inability to induce mitochondrial proliferation.
Collapse
|
48
|
Van Proeyen K, Szlufcik K, Nielens H, Deldicque L, Van Dyck R, Ramaekers M, Hespel P. High-fat diet overrules the effects of training on fiber-specific intramyocellular lipid utilization during exercise. J Appl Physiol (1985) 2011; 111:108-16. [PMID: 21551007 DOI: 10.1152/japplphysiol.01459.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In this study, we compared the effects of endurance training in the fasted state (F) vs. the fed state [ample carbohydrate intake (CHO)] on exercise-induced intramyocellular lipid (IMCL) and glycogen utilization during a 6-wk period of a hypercaloric (∼+30% kcal/day) fat-rich diet (HFD; 50% of kcal). Healthy male volunteers (18-25 yrs) received a HFD in conjunction with endurance training (four times, 60-90 min/wk) either in F (n = 10) or with CHO before and during exercise sessions (n = 10). The control group (n = 7) received a HFD without training and increased body weight by ∼3 kg (P < 0.001). Before and after a HFD, the subjects performed a 2-h constant-load bicycle exercise test in F at ∼70% maximal oxygen uptake rate. A HFD, both in the absence (F) or presence (CHO) of training, elevated basal IMCL content by ∼50% in type I and by ∼75% in type IIa fibers (P < 0.05). Independent of training in F or CHO, a HFD, as such, stimulated exercise-induced net IMCL breakdown by approximately twofold in type I and by approximately fourfold in type IIa fibers. Furthermore, exercise-induced net muscle glycogen breakdown was not significantly affected by a HFD. It is concluded that a HFD stimulates net IMCL degradation by increasing basal IMCL content during exercise in type I and especially IIa fibers. Furthermore, a hypercaloric HFD provides adequate amounts of carbohydrates to maintain high muscle glycogen content during training and does not impair exercise-induced muscle glycogen breakdown.
Collapse
Affiliation(s)
- Karen Van Proeyen
- Research Centre for Exercise and Health, Department of Biomedical Kinesiology, Katholieke Universiteit Leuven, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
49
|
van Herpen NA, Schrauwen-Hinderling VB, Schaart G, Mensink RP, Schrauwen P. Three weeks on a high-fat diet increases intrahepatic lipid accumulation and decreases metabolic flexibility in healthy overweight men. J Clin Endocrinol Metab 2011; 96:E691-5. [PMID: 21252252 DOI: 10.1210/jc.2010-2243] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
CONTEXT In rodents, high-fat diets increase intrahepatic lipid (IHL), but human studies are scarce. OBJECTIVE Our objective was to examine whether high-fat diets influence IHL, intramyocellular lipids (IMCL), and insulin resistance. DESIGN Twenty overweight men were randomly allocated to low- or high-fat groups (age, 54.0 ± 2.3 and 56.4 ± 2.5 yr; body mass index, 29.3 ± 0.6 and 28.3 ± 0.5 kg/m(2), respectively). Both groups started with a 3-wk low-fat diet [15% energy (En%) as protein, 65 En% as carbohydrates, 20 En% as fat], after which half of the subjects switched to a 3-wk isocaloric high-fat diet (15 En% protein, 30 En% carbohydrates, 55 En% fat). After 3 and 6 wk, IHL and IMCL content were assessed by (1)H magnetic resonance spectroscopy and a muscle biopsy, and insulin sensitivity was studied using a hyperinsulinemic-euglycemic clamp. An additional liver scan was performed after 1 wk in the high-fat group. RESULTS IHL decreased by 13% in the low-fat group and increased by 17% in high-fat group (P = 0.047). IMCL content was unaffected (P = 0.304). Insulin sensitivity was unaffected. At wk 3, IHL correlated negatively with insulin sensitivity (r = -0.584; P = 0.009, all subjects combined). Metabolic flexibility, defined as change in respiratory quotient upon insulin stimulation, was decreased after 3 wk of the high-fat diet (change in respiratory quotient was +0.02 ± 0.02 vs. -0.05 ± 0.1 in low-fat vs. high-fat group, P = 0.009). Basal plasma glucose increased after the high-fat diet (P = 0.038). Plasma parameters insulin, free fatty acids, high-sensitivity C-reactive protein, and liver enzymes and body weight were unaffected by diet. CONCLUSION A 3-wk high-fat diet leads to IHL accumulation and a decreased metabolic flexibility, but insulin sensitivity is unaffected.
Collapse
Affiliation(s)
- Noud A van Herpen
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology, and Metabolism, Maastricht University Medical Centre, 6200 MD Maastricht, The Netherlands
| | | | | | | | | |
Collapse
|
50
|
Van Proeyen K, Szlufcik K, Nielens H, Pelgrim K, Deldicque L, Hesselink M, Van Veldhoven PP, Hespel P. Training in the fasted state improves glucose tolerance during fat-rich diet. J Physiol 2011; 588:4289-302. [PMID: 20837645 DOI: 10.1113/jphysiol.2010.196493] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A fat-rich energy-dense diet is an important cause of insulin resistance. Stimulation of fat turnover in muscle cells during dietary fat challenge may contribute to maintenance of insulin sensitivity. Exercise in the fasted state markedly stimulates energy provision via fat oxidation. Therefore, we investigated whether exercise training in the fasted state is more potent than exercise in the fed state to rescue whole-body glucose tolerance and insulin sensitivity during a period of hyper-caloric fat-rich diet. Healthy male volunteers (18-25 y) received a hyper-caloric (∼+30% kcal day(-1)) fat-rich (50% of kcal) diet for 6 weeks. Some of the subjects performed endurance exercise training (4 days per week) in the fasted state (F; n = 10), whilst the others ingested carbohydrates before and during the training sessions (CHO; n = 10). The control group did not train (CON; n = 7). Body weight increased in CON (+3.0 ± 0.8 kg) and CHO (+1.4 ± 0.4 kg) (P < 0.01), but not in F (+0.7 ± 0.4 kg, P = 0.13). Compared with CON, F but not CHO enhanced whole-body glucose tolerance and the Matsuda insulin sensitivity index (P < 0.05). Muscle GLUT4 protein content was increased in F (+28%) compared with both CHO (P = 0.05) and CON (P < 0.05). Furthermore, only training in F elevated AMP-activated protein kinase α phosphorylation (+25%) as well as up-regulated fatty acid translocase/CD36 and carnitine palmitoyltransferase 1 mRNA levels compared with CON (∼+30%). High-fat diet increased intramyocellular lipid but not diacylglycerol and ceramide contents, either in the absence or presence of training. This study for the first time shows that fasted training is more potent than fed training to facilitate adaptations in muscle and to improve whole-body glucose tolerance and insulin sensitivity during hyper-caloric fat-rich diet.
Collapse
Affiliation(s)
- Karen Van Proeyen
- Research Centre for Exercise and Health, Department of Biomedical Kinesiology, K.U. Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|