101
|
Ji Q. Treatment Strategy for Type 2 Diabetes with Obesity: Focus on Glucagon-like Peptide-1 Receptor Agonists. Clin Ther 2017; 39:1244-1264. [PMID: 28526416 DOI: 10.1016/j.clinthera.2017.03.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 03/08/2017] [Accepted: 03/15/2017] [Indexed: 12/18/2022]
Abstract
PURPOSE The progressive nature of type 2 diabetes mellitus (T2DM) calls for step-wise intensification of therapy for maintaining normal glycemic levels and lowering cardiovascular (CV) risk. Because obesity is a prominent risk factor and comorbidity of T2DM, it further elevates the CV risk in T2DM. Therefore, it is vital to manage weight, obesity, and glycemic parameters for effective T2DM management. Few oral antidiabetic drugs (sulfonylureas and thiazolidinediones) and insulin are not suitable for obese patients with T2DM because these drugs cause weight gain. The present review discusses the place of glucagon-like peptide-1 receptor agonists (GLP-1RAs) in the treatment of obese patients with T2DM and the significance of these drugs in the prevention of future CV risk in patients with T2DM. METHODS A literature search of PubMed and EMBASE was conducted by using the search terms T2DM, GLP-1RAs, obesity, and cardiovascular complication. Randomized controlled trials measuring the effect of GLP-1RAs versus that of placebo on CV outcomes were included in the review. FINDINGS GLP-1RAs have emerged as a therapeutic alternative; these drugs exert their actions by providing glycemic control, improving insulin resistance and ö̇-cell function, and reducing weight. The risk of hypoglycemia with GLP-1RAs is minimal; however, GLP-1RAs are associated with gastrointestinal adverse events and raise concerns regarding pancreatitis. Combining GLP-1RAs with insulin analogues results in higher efficacy, a lowered insulin dose, and reduced insulin-related hypoglycemia and weight gain. Longer acting GLP-1RAs are also associated with improvement in medication adherence. Improvement in CV risk factors such as blood pressure and lipid profile further increases their usability for improving CV outcomes. IMPLICATIONS Overall, the properties of GLP-1RAs make them suitable for combination with oral antidiabetic drugs in the early stages of T2DM and with insulins in the later stages for optimizing comprehensive management of the disease.
Collapse
Affiliation(s)
- Qiuhe Ji
- Department of Endocrinology, Xijing Hospital, The First Affiliated Hospital of the Fourth Military Medical University, Xian, People's Republic of China.
| |
Collapse
|
102
|
Knop FK, Brønden A, Vilsbøll T. Exenatide: pharmacokinetics, clinical use, and future directions. Expert Opin Pharmacother 2017; 18:555-571. [PMID: 28085521 DOI: 10.1080/14656566.2017.1282463] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The first-in-class glucagon-like peptide-1 receptor agonist (GLP-1RA) exenatide, which was initially approved in 2005, is available in twice-daily (BID) and once-weekly (QW) formulations. Clinical trial data suggest both formulations are effective and safe for patients with type 2 diabetes (T2D), both as monotherapy and as part of combination therapy. Since exenatide was approved, several other GLP-1RAs have become available for clinical use. Areas covered: Many ongoing clinical trials involving exenatide BID and exenatide QW are investigating new indications (exenatide BID) and new end points and combination therapies (exenatide QW). This review provides an overview of the delivery and pharmacokinetics of both formulations of exenatide, reviews existing data in T2D, and summarizes ongoing investigations. Expert opinion: Exenatide BID and QW have substantial clinical benefits. Comparisons with other GLP-1RAs demonstrate some differences in efficacy and safety profiles that make assessment of benefit:risk ratios complex. Head-to-head comparisons of QW GLP-1RA formulations may assist in the ranking of GLP-1RAs according to efficacy and safety. Results on the impact of exenatide QW on cardiovascular outcomes are eagerly awaited. The potential clinical utility of exenatide BID in other indications will clarify whether exenatide holds clinical promise in diagnoses other than T2D.
Collapse
Affiliation(s)
- Filip K Knop
- a Center for Diabetes Research, Gentofte Hospital , University of Copenhagen , Hellerup , Denmark.,b Department of Clinical Medicine, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark.,c The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark
| | - Andreas Brønden
- a Center for Diabetes Research, Gentofte Hospital , University of Copenhagen , Hellerup , Denmark.,b Department of Clinical Medicine, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark
| | - Tina Vilsbøll
- a Center for Diabetes Research, Gentofte Hospital , University of Copenhagen , Hellerup , Denmark.,b Department of Clinical Medicine, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
103
|
Cernea S, Cahn A, Raz I. Pharmacological management of nonalcoholic fatty liver disease in type 2 diabetes. Expert Rev Clin Pharmacol 2017; 10:535-547. [PMID: 28276774 DOI: 10.1080/17512433.2017.1300059] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The prevalence of nonalcoholic fatty liver disease (NAFLD) in patients with type 2 diabetes (T2D) is high and it is associated with poor prognosis. Hepatic steatosis results as a consequence of excessive hepatic lipid accumulation which correlates with insulin resistance and lipotoxicity, with subsequent oxidative stress, inflammation, apoptosis and fibrosis. Areas covered: This article presents the main pathophysiologic mechanisms and currently available drugs evaluated for their therapeutic effects on NAFLD/nonalcoholic steatohepatitis (NASH) and drugs under development that target relevant pathogenetic pathways. However, to date there is no particular drug approved for treatment of NAFLD in patients with T2D. Expert commentary: Early recognition and intervention are essential to ameliorate disease progression. Specific recommendations are still needed for NAFLD/NASH screening and diagnosis and therapeutic algorithm in patients with T2D. Lifestyle optimization with significant weight loss is a key intervention in patients with NAFLD and T2D. Pioglitazone, liraglutide, vitamin E, OCA and pentoxifylline have proven some histological improvements in NASH and omega 3-PUFAs were shown to decrease liver fat, but no specific recommendation can be made for treatment of NASH. Perhaps a combination of agents that target different pathogenic pathways are needed to better control disease progression, but more robust evidence for these agents is still needed.
Collapse
Affiliation(s)
- Simona Cernea
- a Department M3/Internal Medicine IV , University of Medicine and Pharmacy , Târgu Mureş , Romania.,b Diabetes, Nutrition and Metabolic Diseases Outpatient Unit , Emergency County Clinical Hospital , Târgu Mureş , Romania
| | - Avivit Cahn
- c Diabetes Unit, Department of Internal Medicine , Hadassah Hebrew University Hospital , Jerusalem , Israel.,d Endocrinology and Metabolism Unit, Department of Internal Medicine , Hadassah University Hospital , Jerusalem , Israel
| | - Itamar Raz
- c Diabetes Unit, Department of Internal Medicine , Hadassah Hebrew University Hospital , Jerusalem , Israel
| |
Collapse
|
104
|
GLP-1 analogue-induced weight loss does not improve obesity-induced AT dysfunction. Clin Sci (Lond) 2017; 131:343-353. [DOI: 10.1042/cs20160803] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 12/21/2022]
Abstract
Liraglutide shows limited impact on adipose tissue (AT) independent of the associated weight loss. However, despite weight loss and better glycaemic control than with diet alone, Liraglutide increases inflammation and creates a pro-fibrotic environment in subcutaneous AT (SCAT).
Collapse
|
105
|
Rajeev SP, Sprung VS, Roberts C, Harrold JA, Halford JCG, Stancak A, Boyland EJ, Kemp GJ, Cuthbertson DJ, Wilding JPH. Compensatory changes in energy balance during dapagliflozin treatment in type 2 diabetes mellitus: a randomised double-blind, placebo-controlled, cross-over trial (ENERGIZE)-study protocol. BMJ Open 2017; 7:e013539. [PMID: 28132008 PMCID: PMC5278268 DOI: 10.1136/bmjopen-2016-013539] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Sodium glucose cotransporter 2 (SGLT2) inhibitors are effective blood-glucose-lowering medications with beneficial effects on body weight in patients with type 2 diabetes mellitus (T2DM). However, observed weight loss is less than that predicted from quantified glycosuria, suggesting a compensatory increase in energy intake or a decrease in energy expenditure. Studies using dual-energy X-ray absorptiometry (DEXA) have suggested most body weight change is due to loss of adipose tissue, but organ-specific changes in fat content (eg, liver, skeletal muscle) have not been determined. In this randomised, double-blind, placebo-controlled crossover study, we aim to study the compensatory changes in energy intake, eating behaviour and energy expenditure accompanying use of the SGLT2 inhibitor, dapagliflozin. Additionally, we aim to quantify changes in fat distribution using MRI, in liver fat using proton magnetic resonance spectroscopy (1H-MRS) and in central nervous system (CNS) responses to food images using blood oxygen level dependent (BOLD) functional MRI (fMRI). METHODS AND ANALYSIS This outpatient study will evaluate the effect of dapagliflozin (10 mg), compared with placebo, on food intake and energy expenditure at 7 days and 12 weeks. 52 patients with T2DM will be randomised to dapagliflozin or placebo for short-term and long-term trial interventions in a within participants, crossover design. The primary outcome is the difference in energy intake during a test meal between dapagliflozin and placebo. Intake data are collected automatically using a customised programme operating a universal eating monitor (UEM). Secondary outcomes include (1) measures of appetite regulation including rate of eating, satiety quotient, appetite ratings (between and within meals), changes in CNS responses to food images measured using BOLD-fMRI, (2) measures of energy expenditure and (3) changes in body composition including changes in liver fat and abdominal visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT). ETHICAL APPROVAL This study has been approved by the North West Liverpool Central Research Ethics Committee (14/NW/0340) and is conducted in accordance with the Declaration of Helsinki and the Good Clinical Practice (GCP). TRIAL REGISTRATION NUMBER ISRCTN14818531. EUDRACT number 2013-004264-60.
Collapse
Affiliation(s)
- Surya Panicker Rajeev
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Diabetes and Endocrinology Research Group, Clinical Sciences Centre, Aintree University Hospital NHS Foundation Trust, Liverpool, UK
| | - Victoria S Sprung
- Diabetes and Endocrinology Research Group, Clinical Sciences Centre, Aintree University Hospital NHS Foundation Trust, Liverpool, UK
- Department of Musculoskeletal Biology II, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Carl Roberts
- Department of Psychological Sciences, Institute of Psychology, Health and Society, University of Liverpool, Liverpool, UK
| | - Jo A Harrold
- Department of Psychological Sciences, Institute of Psychology, Health and Society, University of Liverpool, Liverpool, UK
| | - Jason C G Halford
- Department of Psychological Sciences, Institute of Psychology, Health and Society, University of Liverpool, Liverpool, UK
| | - Andrej Stancak
- Department of Psychological Sciences, Institute of Psychology, Health and Society, University of Liverpool, Liverpool, UK
| | - Emma J Boyland
- Department of Psychological Sciences, Institute of Psychology, Health and Society, University of Liverpool, Liverpool, UK
| | - Graham J Kemp
- Department of Musculoskeletal Biology II, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Magnetic Resonance and Image Analysis Research Centre (MARIARC), University of Liverpool, Liverpool, UK
| | - Daniel J Cuthbertson
- Diabetes and Endocrinology Research Group, Clinical Sciences Centre, Aintree University Hospital NHS Foundation Trust, Liverpool, UK
- Department of Musculoskeletal Biology II, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - John P H Wilding
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Diabetes and Endocrinology Research Group, Clinical Sciences Centre, Aintree University Hospital NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
106
|
Lundkvist P, Sjöström CD, Amini S, Pereira MJ, Johnsson E, Eriksson JW. Dapagliflozin once-daily and exenatide once-weekly dual therapy: A 24-week randomized, placebo-controlled, phase II study examining effects on body weight and prediabetes in obese adults without diabetes. Diabetes Obes Metab 2017; 19:49-60. [PMID: 27550386 PMCID: PMC5215525 DOI: 10.1111/dom.12779] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/18/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022]
Abstract
AIMS To explore the effects of dual therapy with dapagliflozin and exenatide on body weight, body composition, glycaemic variables and systolic blood pressure (SBP) in obese adults without diabetes. MATERIALS AND METHODS In this single-centre, double-blind trial, we randomized 50 obese adults without diabetes (aged 18-70 years; body mass index 30-45 kg/m2 ) to oral dapagliflozin 10 mg once daily plus subcutaneous long-acting exenatide 2 mg once weekly or placebo. MRI was used to assess change in body composition. Participants were instructed to follow a balanced diet and exercise moderately. RESULTS Of 25 dapagliflozin/exenatide- and 25 placebo-treated participants, 23 (92.0%) and 20 (80.0%) completed 24 weeks of treatment, respectively. At baseline, the mean participant age was 52 years, 61% were female, the mean body weight was 104.6 kg, and 73.5% of participants had prediabetes (impaired fasting glucose or impaired glucose tolerance). After 24 weeks, for dapagliflozin/exenatide versus placebo: the difference in body weight change was -4.13 kg (95% confidence interval -6.44, -1.81; P < .001), which was mostly attributable to adipose tissue reduction without lean tissue change; 36.0% versus 4.2% of participants achieved ≥5% body weight loss, respectively; and prediabetes was less frequent with active treatment (34.8% vs 85.0%, respectively; P < .01). The difference in SBP change for dapagliflozin/exenatide versus placebo was -6.7 mm Hg. As expected, nausea and injection-site reactions were more frequent with dapagliflozin/exenatide than with placebo. Only two and three participants, respectively, discontinued because of adverse events. CONCLUSIONS Compared with placebo, dapagliflozin/exenatide dual therapy reduced body weight, frequency of prediabetes and SBP over 24 weeks and was well tolerated in obese adults without diabetes.
Collapse
Affiliation(s)
- Per Lundkvist
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | | | - Sam Amini
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | | | | | - Jan W. Eriksson
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| |
Collapse
|
107
|
Petta S, Gastaldelli A, Rebelos E, Bugianesi E, Messa P, Miele L, Svegliati-Baroni G, Valenti L, Bonino F. Pathophysiology of Non Alcoholic Fatty Liver Disease. Int J Mol Sci 2016; 17:2082. [PMID: 27973438 PMCID: PMC5187882 DOI: 10.3390/ijms17122082] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 12/18/2022] Open
Abstract
The physiopathology of fatty liver and metabolic syndrome are influenced by diet, life style and inflammation, which have a major impact on the severity of the clinicopathologic outcome of non-alcoholic fatty liver disease. A short comprehensive review is provided on current knowledge of the pathophysiological interplay among major circulating effectors/mediators of fatty liver, such as circulating lipids, mediators released by adipose, muscle and liver tissues and pancreatic and gut hormones in relation to diet, exercise and inflammation.
Collapse
Affiliation(s)
- Salvatore Petta
- Gastroenterology, Di.Bi.M.I.S Policlinic Paolo Giaccone Hospital, University of Palermo, PC 90127, Palermo, Italy.
| | - Amalia Gastaldelli
- Cardiometabolic Risk Unit-Institute of Clinical Physiology, CNR, PC 56124, Pisa, Italy.
| | - Eleni Rebelos
- Department of Clinical and Experimental Medicine, University of Pisa, PC 56122, Pisa, Italy.
| | - Elisabetta Bugianesi
- Gastroenterology and Hepatology, Department of Medical Sciences, Città della, Salute e della Scienza di Torino Hospital, University of Turin, PC 10122, Turin, Italy.
| | - Piergiorgio Messa
- Department of Nephrology, Urology and Renal Transplant-Fondazione IRCCS Ca', Granda, PC 20122 Milano, Italy.
| | - Luca Miele
- Institute of Internal Medicine, Gastroenterology and Liver Diseases Unit, Fondazione Policlinico Gemelli, Catholic University of Rome, PC 00168, Rome, Italy.
| | - Gianluca Svegliati-Baroni
- Department of Gastroenterology 1 and Obesity 2, Polytechnic University of Marche, PC 60121, Ancona, Italy.
| | - Luca Valenti
- Metabolic Liver Diseases-Università degli Studi Milano-Fondazione IRCCS Ca', Granda via F Sforza 35, PC 20122 Milano, Italy.
| | - Ferruccio Bonino
- Department of Clinical and Experimental Medicine, University of Pisa, PC 56122, Pisa, Italy.
- Institute for Health, PC 53042, Chianciano Terme, Italy.
| |
Collapse
|
108
|
Steven S, Hollingsworth KG, Small PK, Woodcock SA, Pucci A, Aribasala B, Al-Mrabeh A, Batterham RL, Taylor R. Calorie restriction and not glucagon-like peptide-1 explains the acute improvement in glucose control after gastric bypass in Type 2 diabetes. Diabet Med 2016; 33:1723-1731. [PMID: 27589584 DOI: 10.1111/dme.13257] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/31/2016] [Indexed: 12/15/2022]
Abstract
AIMS To compare directly the impact of glucagon-like peptide-1 secretion on glucose metabolism in individuals with Type 2 diabetes listed for Roux-en-Y gastric bypass surgery, randomized to be studied before and 7 days after undergoing Roux-en-Y gastric bypass or after following a very-low-calorie diet. METHODS A semi-solid meal test was used to investigate glucose, insulin and glucagon-like peptide-1 response. Insulin secretion in response to intravenous glucose and arginine stimulus was measured. Hepatic and pancreatic fat content was quantified using magnetic resonance imaging. RESULTS The decrease in fat mass was almost identical in the Roux-en-Y gastric bypass and the very-low-calorie diet groups (3.0±0.3 and 3.0±0.7kg). The early rise in plasma glucose level and in acute insulin secretion were greater after Roux-en-Y gastric bypass than after a very-low-calorie diet; however, the early rise in glucagon-like peptide-1 was disproportionately greater (sevenfold) after Roux-en-Y gastric bypass than after a very-low-calorie diet. This did not translate into a greater improvement in fasting glucose level or area under the curve for glucose. The reduction in liver fat was greater after Roux-en-Y gastric bypass (29.8±3.7 vs 18.6±4.0%) and the relationships between weight loss and reduction in liver fat differed between the Roux-en-Y gastric bypass group and the very-low-calorie diet group. CONCLUSIONS This study shows that gastroenterostomy increases the rate of nutrient absorption, bringing about a commensurately rapid rise in insulin level; however, there was no association with the large post-meal rise in glucagon-like peptide-1, and post-meal glucose homeostasis was similar in the Roux-en-Y gastric bypass and very-low-calorie diet groups. (Clinical trials registry number: ISRCTN11969319.).
Collapse
Affiliation(s)
- S Steven
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - K G Hollingsworth
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - P K Small
- Department of Surgery, Sunderland Royal Hospital, Sunderland, UK
| | - S A Woodcock
- Department of Surgery, North Tyneside General Hospital, North Shields, UK
| | - A Pucci
- Centre for Obesity Research, University College London, London, UK
| | - B Aribasala
- Computer Science Department, Faculty of Science, Lagos State University, Lagos, Nigeria
| | - A Al-Mrabeh
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - R L Batterham
- Centre for Obesity Research, University College London, London, UK
| | - R Taylor
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
109
|
Smits MM, Tonneijck L, Muskiet MHA, Kramer MHH, Pouwels PJW, Pieters-van den Bos IC, Hoekstra T, Diamant M, van Raalte DH, Cahen DL. Twelve week liraglutide or sitagliptin does not affect hepatic fat in type 2 diabetes: a randomised placebo-controlled trial. Diabetologia 2016; 59:2588-2593. [PMID: 27627981 PMCID: PMC6518065 DOI: 10.1007/s00125-016-4100-7] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 08/19/2016] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS Glucagon-like peptide (GLP)-1-based therapies have been suggested to improve hepatic steatosis. We assessed the effects of the GLP-1 receptor agonist liraglutide and the dipeptidyl peptidase (DPP)-4 inhibitor sitagliptin on hepatic steatosis and fibrosis in patients with type 2 diabetes. METHODS In this 12 week, parallel, randomised, placebo-controlled trial, performed at the VU University Medical Center between July 2013 and August 2015, 52 overweight patients with type 2 diabetes treated with metformin and/or sulphonylurea agent ([mean ± SD] age 62.7 ± 6.9 years, HbA1c 7.3 ± 0.7% or 56 ± 1 mmol/mol) were allocated to once daily liraglutide 1.8 mg (n = 17), sitagliptin 100 mg (n = 18) or matching placebos (n = 17) by computer generated numbers. Both participants and researchers were blinded to group assignment. Hepatic fat content was measured using proton magnetic resonance spectroscopy (1H-MRS). Hepatic fibrosis was estimated using three validated formulae. RESULTS One patient dropped out in the sitagliptin group owing to dizziness, but no serious adverse events occurred. At week 12, no between-group differences in hepatic steatosis were found. Liraglutide reduced steatosis by 10% (20.9 ± 3.4% to 18.8 ± 3.3%), sitagliptin reduced steatosis by 12.1% (23.9 ± 3.0% to 21.0 ± 2.7%) and placebo lessened it by 9.5% (18.7 ± 2.7% to 16.9 ± 2.7%). Neither drug affected hepatic fibrosis scores compared with placebo. CONCLUSIONS/INTERPRETATION Twelve-week liraglutide or sitagliptin treatment does not reduce hepatic steatosis or fibrosis in type 2 diabetes. TRIAL REGISTRATION ClinicalTrials.gov NCT01744236 FUNDING : Funded by the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 282521 - the SAFEGUARD project.
Collapse
Affiliation(s)
- Mark M Smits
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, De Boelelaan 1117 (Room ZH 4A65), 1081 HV, Amsterdam, the Netherlands.
| | - Lennart Tonneijck
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, De Boelelaan 1117 (Room ZH 4A65), 1081 HV, Amsterdam, the Netherlands
| | - Marcel H A Muskiet
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, De Boelelaan 1117 (Room ZH 4A65), 1081 HV, Amsterdam, the Netherlands
| | - Mark H H Kramer
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, De Boelelaan 1117 (Room ZH 4A65), 1081 HV, Amsterdam, the Netherlands
| | - Petra J W Pouwels
- Department of Physics and Medical Technology, VU University Medical Center and Neuroscience Campus Amsterdam, Amsterdam, the Netherlands
| | | | - Trynke Hoekstra
- Department of Health Sciences and the EMGO Institute for Health and Care Research, VU University Amsterdam, Amsterdam, the Netherlands
- Department of Epidemiology and Biostatistics, VU University Medical Center, Amsterdam, the Netherlands
| | - Michaela Diamant
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, De Boelelaan 1117 (Room ZH 4A65), 1081 HV, Amsterdam, the Netherlands
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, VU University Medical Center, De Boelelaan 1117 (Room ZH 4A65), 1081 HV, Amsterdam, the Netherlands
| | - Djuna L Cahen
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
110
|
Mitra S, Fernandez-Del-Valle M, Hill JE. The role of MRI in understanding the underlying mechanisms in obesity associated diseases. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1115-1131. [PMID: 27639834 DOI: 10.1016/j.bbadis.2016.09.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 02/07/2023]
Abstract
Obesity and its possible association with diseases including diabetes and cardiovascular diseases have been studied for decades for its impact on healthcare. Recent studies clearly indicate the need for developing accurate and reproducible methodologies for assessing body fat content and distribution. Body fat distribution plays a significant role in developing an insight in the underlying mechanisms in which adipose tissue is linked with various diseases. Among imaging technologies including computerized axial tomography (CAT or CT), magnetic resonance imaging (MRI), and magnetic resonance spectroscopy (MRS), MRI and MRS seem to be the best emerging techniques and together are being considered as the gold standard for body fat content and distribution. This paper reviews studies up to the present time involving different methodologies of these two emerging technologies and presents the basic concepts of MRI and MRS with required novel image analysis techniques in accurate, quantitative, and direct assessment of body fat content and distribution. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
| | | | - Jason E Hill
- Texas Tech University, Lubbock, TX, United States
| |
Collapse
|
111
|
Jin T, Weng J. Hepatic functions of GLP-1 and its based drugs: current disputes and perspectives. Am J Physiol Endocrinol Metab 2016; 311:E620-7. [PMID: 27507553 DOI: 10.1152/ajpendo.00069.2016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 08/08/2016] [Indexed: 12/17/2022]
Abstract
GLP-1 and its based drugs possess extrapancreatic metabolic functions, including that in the liver. These direct hepatic metabolic functions explain their therapeutic efficiency for subjects with insulin resistance. The direct hepatic functions could be mediated by previously assumed "degradation" products of GLP-1 without involving canonic GLP-1R. Although GLP-1 analogs were created as therapeutic incretins, extrapancreatic functions of these drugs, as well as native GLP-1, have been broadly recognized. Among them, the hepatic functions are particularly important. Postprandial GLP-1 release contributes to insulin secretion, which represses hepatic glucose production. This indirect effect of GLP-1 is known as the gut-pancreas-liver axis. Great efforts have been made to determine whether GLP-1 and its analogs possess direct metabolic effects on the liver, as the determination of the existence of direct hepatic effects may advance the therapeutic theory and clinical practice on subjects with insulin resistance. Furthermore, recent investigations on the metabolic beneficial effects of previously assumed "degradation" products of GLP-1 in the liver and elsewhere, including GLP-128-36 and GLP-132-36, have drawn intensive attention. Such investigations may further improve the development and the usage of GLP-1-based drugs. Here, we have reviewed the current advancement and the existing controversies on the exploration of direct hepatic functions of GLP-1 and presented our perspectives that the direct hepatic metabolic effects of GLP-1 could be a GLP-1 receptor-independent event involving Wnt signaling pathway activation.
Collapse
Affiliation(s)
- Tianru Jin
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada; Banting and Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; and
| | - Jianping Weng
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| |
Collapse
|
112
|
Dutour A, Abdesselam I, Ancel P, Kober F, Mrad G, Darmon P, Ronsin O, Pradel V, Lesavre N, Martin JC, Jacquier A, Lefur Y, Bernard M, Gaborit B. Exenatide decreases liver fat content and epicardial adipose tissue in patients with obesity and type 2 diabetes: a prospective randomized clinical trial using magnetic resonance imaging and spectroscopy. Diabetes Obes Metab 2016; 18:882-91. [PMID: 27106272 DOI: 10.1111/dom.12680] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/11/2016] [Accepted: 04/20/2016] [Indexed: 12/24/2022]
Abstract
AIM To conduct a prospective randomized trial to investigate the effect of glucagon-like peptide-1 (GLP-1) analogues on ectopic fat stores. METHODS A total of 44 obese subjects with type 2 diabetes uncontrolled on oral antidiabetic drugs were randomly assigned to receive exenatide or reference treatment according to French guidelines. Epicardial adipose tissue (EAT), myocardial triglyceride content (MTGC), hepatic triglyceride content (HTGC) and pancreatic triglyceride content (PTGC) were assessed 45 min after a standardized meal with 3T magnetic resonance imaging and proton magnetic resonance spectroscopy before and after 26 weeks of treatment. RESULTS The study population had a mean glycated haemoglobin (HbA1c) level of 7.5 ± 0.2% and a mean body mass index of 36.1 ± 1.1 kg/m(2) . Ninety five percent had hepatic steatosis at baseline (HTGC ≥ 5.6%). Exenatide and reference treatment led to a similar improvement in HbA1c (-0.7 ± 0.3% vs. -0.7 ± 0.4%; p = 0.29), whereas significant weight loss was observed only in the exenatide group (-5.5 ± 1.2 kg vs. -0.2 ± 0.8 kg; p = 0.001 for the difference between groups). Exenatide induced a significant reduction in EAT (-8.8 ± 2.1%) and HTGC (-23.8 ± 9.5%), compared with the reference treatment (EAT: -1.2 ± 1.6%, p = 0.003; HTGC: +12.5 ± 9.6%, p = 0.007). No significant difference was observed in other ectopic fat stores, PTGC or MTGC. In the group treated with exenatide, reductions in liver fat and EAT were not associated with homeostatic model assessment of insulin resistance index, adiponectin, HbA1c or fructosamin change, but were significantly related to weight loss (r = 0.47, p = 0.03, and r = 0.50, p = 0.018, respectively). CONCLUSION Our data indicate that exenatide is an effective treatment to reduce liver fat content and epicardial fat in obese patients with type 2 diabetes, and these effects are mainly weight loss dependent.
Collapse
Affiliation(s)
- A Dutour
- Inserm U1062, Inra U1260, Faculté de Médecine, 13385, Marseille, France
- Aix Marseille Université, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pole Endo, Marseille, France
| | - I Abdesselam
- Inserm U1062, Inra U1260, Faculté de Médecine, 13385, Marseille, France
- Aix Marseille Université, Marseille, France
- Centre de Résonance Magnétique Biologique et Médicale, CNRS UMR 7339, Marseille, France
| | - P Ancel
- Inserm U1062, Inra U1260, Faculté de Médecine, 13385, Marseille, France
- Aix Marseille Université, Marseille, France
| | - F Kober
- Aix Marseille Université, Marseille, France
- Centre de Résonance Magnétique Biologique et Médicale, CNRS UMR 7339, Marseille, France
| | - G Mrad
- Inserm U1062, Inra U1260, Faculté de Médecine, 13385, Marseille, France
| | - P Darmon
- Inserm U1062, Inra U1260, Faculté de Médecine, 13385, Marseille, France
- Aix Marseille Université, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pole Endo, Marseille, France
| | - O Ronsin
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pole Endo, Marseille, France
| | - V Pradel
- Aix Marseille Université, Marseille, France
- Statistics Department, Assistance Publique Hôpitaux Marseille, CHU Sainte Marguerite, Marseille, France
| | - N Lesavre
- Aix Marseille Université, Marseille, France
- Centre d'investigation Clinique, 1409, Assistance Publique Hôpitaux de Marseille, CHU Nord, Marseille, France
| | - J C Martin
- Inserm U1062, Inra U1260, Faculté de Médecine, 13385, Marseille, France
- Aix Marseille Université, Marseille, France
| | - A Jacquier
- Aix Marseille Université, Marseille, France
- Centre de Résonance Magnétique Biologique et Médicale, CNRS UMR 7339, Marseille, France
- Radiology Department, CHU La Timone, Marseille, France
| | - Y Lefur
- Aix Marseille Université, Marseille, France
- Centre de Résonance Magnétique Biologique et Médicale, CNRS UMR 7339, Marseille, France
| | - M Bernard
- Aix Marseille Université, Marseille, France
- Centre de Résonance Magnétique Biologique et Médicale, CNRS UMR 7339, Marseille, France
| | - B Gaborit
- Inserm U1062, Inra U1260, Faculté de Médecine, 13385, Marseille, France
- Aix Marseille Université, Marseille, France
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pole Endo, Marseille, France
| |
Collapse
|
113
|
Noureddin M, Zhang A, Loomba R. Promising therapies for treatment of nonalcoholic steatohepatitis. Expert Opin Emerg Drugs 2016; 21:343-57. [PMID: 27501374 DOI: 10.1080/14728214.2016.1220533] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) has become the most common etiology for abnormal aminotransferase levels and chronic liver disease. Its growing prevalence is largely linked to the presence of metabolic syndrome, particularly diabetes and insulin resistance. It is estimated that 60-80% of the type 2 diabetic population has NAFLD. NAFLD encompasses a range of conditions ranging from simple steatosis to non-alcoholic steatohepatitis (NASH). A subset of patients with hepatic steatosis progress to NASH, while 15-20% of patients with NASH develop cirrhosis. This progression is thought to be multifactorial, and there are currently no FDA-approved medications for the treatment of NASH. AREAS COVERED We review drugs currently in Phase II and III clinical trials for treatment of NAFLD and NASH, including their mechanisms of action, relationship to the pathophysiology of NASH, and rationale for their development. EXPERT OPINION The treatment of NASH is complex and necessitates targeting a number of different pathways. Combination therapy, preferably tailored toward the disease stage and severity, will be needed to achieve maximum therapeutic effect. With multiple agents currently being developed, there may soon be an ability to effectively slow or even reverse the disease process in many NAFLD/NASH patients.
Collapse
Affiliation(s)
- Mazen Noureddin
- a Fatty Liver Program, Division of Digestive and Liver Diseases, Comprehensive Transplant Center , Cedars-Sinai Medical Center , Los Angeles , CA , USA.,b Department of Medicine , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| | - Alice Zhang
- b Department of Medicine , Cedars-Sinai Medical Center , Los Angeles , CA , USA
| | - Rohit Loomba
- c Division of Gastroenterology and Division of Epidemiology , University of California, San Diego , La Jolla , CA , USA
| |
Collapse
|
114
|
Barb D, Portillo-Sanchez P, Cusi K. Pharmacological management of nonalcoholic fatty liver disease. Metabolism 2016; 65:1183-95. [PMID: 27301803 DOI: 10.1016/j.metabol.2016.04.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 04/16/2016] [Accepted: 04/18/2016] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) affects one-third of the population and two-thirds of patients with obesity or type 2 diabetes (T2DM). Its more aggressive form is known as nonalcoholic steatohepatitis (NASH) and is characterized by hepatocyte necrosis, inflammation and often fibrosis. The presence of fibrosis indicates a more aggressive course and may lead to cirrhosis. Premature mortality in NASH is related to both hepatic (cirrhosis and hepatocellular carcinoma) and extra-hepatic complications, largely cardiovascular disease (CVD). Many therapeutic agents have been tested, but still none approved specifically for NASH. Treatment of NAFLD includes aggressive management of diabetes and cardiovascular risk factors, although the role of controlling hyperglycemia per se in patients with T2DM and NASH remains unknown. Agents tested with some success in non-diabetic patients with NASH include pioglitazone, liraglutide, vitamin E and to a lesser degree, pentoxiphylline. In patients with T2DM and NASH only pioglitazone has shown to significantly improve liver histology, with only a handful of patients with diabetes having been studied with other modalities. This review focuses on available agents for NASH to assist clinicians in the management of these complex patients. Many novel compounds are being studied and will likely make combination therapy for NASH a reality in the future.
Collapse
Affiliation(s)
- Diana Barb
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA; Division of Endocrinology, Diabetes and Metabolism at Malcom Randall Veterans Affairs Medical Center, Gainesville, FL, USA
| | - Paola Portillo-Sanchez
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA; Division of Endocrinology, Diabetes and Metabolism at Malcom Randall Veterans Affairs Medical Center, Gainesville, FL, USA
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA; Division of Endocrinology, Diabetes and Metabolism at Malcom Randall Veterans Affairs Medical Center, Gainesville, FL, USA.
| |
Collapse
|
115
|
Bozzetto L, Annuzzi G, Ragucci M, Di Donato O, Della Pepa G, Della Corte G, Griffo E, Anniballi G, Giacco A, Mancini M, Rivellese AA. Insulin resistance, postprandial GLP-1 and adaptive immunity are the main predictors of NAFLD in a homogeneous population at high cardiovascular risk. Nutr Metab Cardiovasc Dis 2016; 26:623-629. [PMID: 27134062 DOI: 10.1016/j.numecd.2016.01.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 11/06/2015] [Accepted: 01/25/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS The role of the different factors associated with fatty liver is still poorly defined. We evaluated the relationships between liver fat content (LF) and metabolic, inflammatory and nutritional factors in a homogeneous cohort of individuals at high cardio-metabolic risk. METHODS AND RESULTS In 70 individuals with high waist circumference and at least one more criterion for metabolic syndrome enrolled in a nutritional intervention study, LF was evaluated at baseline by hepatic/renal echo intensity ratio (H/R), together with dietary habits (7-day dietary record), insulin sensitivity and β-cell function (fasting and OGTT-derived indices), fasting and postprandial plasma GLP-1 and lipoproteins, and plasma inflammatory markers. H/R correlated positively with fasting and OGTT plasma glucose and insulin concentrations, HOMA-IR and β-cell function, and IL-4, IL-17, IFN-γ, TNF-α, FGF and GCSF plasma concentrations (p < 0.05 for all), and negatively with insulin sensitivity (OGIS), dietary, polyphenols and fiber (p < 0.05 for all). By multiple stepwise regression analysis, the best predictors of H/R were OGIS (β = -0.352 p = 0.001), postprandial GLP-1 (β = -0.344; p = 0.001), HDL-cholesterol (β = -0.323; p = 0.002) and IFN-γ (β = 0.205; p = 0.036). CONCLUSION A comprehensive evaluation of factors associated with liver fat, in a homogeneous population at high cardio-metabolic risk, indicated a pathogenic combination of the same pathways underlying the atherosclerotic process, namely whole body insulin sensitivity and inflammation. The higher predictive value of postprandial variables suggests that liver fat is essentially a postprandial phenomenon, with a relevant role possibly played by GLP-1. REGISTRATION NUMBER FOR CLINICAL TRIALS NCT01154478.
Collapse
Affiliation(s)
- L Bozzetto
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - G Annuzzi
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - M Ragucci
- Department of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - O Di Donato
- Department of Advanced Biomedical Sciences, University Federico II, Naples, Italy
| | - G Della Pepa
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - G Della Corte
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - E Griffo
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - G Anniballi
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - A Giacco
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - M Mancini
- Institute of Biostructures and Bioimaging, National Research Council, Naples, Italy
| | - A A Rivellese
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.
| |
Collapse
|
116
|
GLP-1 analogue improves hepatic lipid accumulation by inducing autophagy via AMPK/mTOR pathway. Biochem Biophys Res Commun 2016; 476:196-203. [PMID: 27208776 DOI: 10.1016/j.bbrc.2016.05.086] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 05/16/2016] [Indexed: 01/27/2023]
Abstract
The incidence of nonalcoholic fatty liver disease (NAFLD) keeps rising year by year, and NAFLD is rapidly becoming the most common liver disease worldwide. Clinical studies have found that glucagon-like peptide-1 (GLP-1) analogue, liraglutide (LRG), cannot only reduce glucose levels, but also improve hepatic lipase, especially in patients also with type 2 diabetes mellitus (T2DM). In addition, enhancing autophagy decreases lipid accumulation in hepatocytes. The aim of the present study is to explore the effect of LRG on hepatocyte steatosis and the possible role of autophagy. We set up an obesity mouse model with a high-fat diet (HFD) and induced hepatocyte steatosis with free fatty acids (FFA) in human L-O2 cells. LRG and two inhibitors of autophagy, Chloroquine (CQ) and bafilomycin A1 (Baf), were added into each group, respectively. The lipid profiles and morphological modifications of each group were tested. Immunohistochemistry, immunofluorescence staining and transmission electron microscopy (TEM) were used to measure autophagy in this study. The autophagy protein expression of SQSTM1 (P62), and LC3B, along with the signaling pathway proteins of mTOR, phosphorylated mTOR (p-mTOR), AMPK, phosphorylated AMPK (p-AMPK) and Beclin1, were evaluated by western blot. Our results showed that LRG improved hepatocyte steatosis by inducing autophagy, and the AMPK/mTOR pathway is involved. These findings suggest an important mechanism for the positive effects of LRG on hepatic steatosis, and provide new evidence for clinical use of LRG in NAFLD.
Collapse
|
117
|
Naftalin RJ. A computer model simulating human glucose absorption and metabolism in health and metabolic disease states. F1000Res 2016; 5:647. [PMID: 27347379 PMCID: PMC4909112 DOI: 10.12688/f1000research.8299.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/31/2016] [Indexed: 12/16/2022] Open
Abstract
A computer model designed to simulate integrated glucose-dependent changes in splanchnic blood flow with small intestinal glucose absorption, hormonal and incretin circulation and hepatic and systemic metabolism in health and metabolic diseases e.g. non-alcoholic fatty liver disease, (NAFLD), non-alcoholic steatohepatitis, (NASH) and type 2 diabetes mellitus, (T2DM) demonstrates how when glucagon-like peptide-1, (GLP-1) is synchronously released into the splanchnic blood during intestinal glucose absorption, it stimulates superior mesenteric arterial (SMA) blood flow and by increasing passive intestinal glucose absorption, harmonizes absorption with its distribution and metabolism. GLP-1 also synergises insulin-dependent net hepatic glucose uptake (NHGU). When GLP-1 secretion is deficient post-prandial SMA blood flow is not increased and as NHGU is also reduced, hyperglycaemia follows. Portal venous glucose concentration is also raised, thereby retarding the passive component of intestinal glucose absorption. Increased pre-hepatic sinusoidal resistance combined with portal hypertension leading to opening of intrahepatic portosystemic collateral vessels are NASH-related mechanical defects that alter the balance between splanchnic and systemic distributions of glucose, hormones and incretins.The model reveals the latent contribution of portosystemic shunting in development of metabolic disease. This diverts splanchnic blood content away from the hepatic sinuses to the systemic circulation, particularly during the glucose absorptive phase of digestion, resulting in inappropriate increases in insulin-dependent systemic glucose metabolism. This hastens onset of hypoglycaemia and thence hyperglucagonaemia. The model reveals that low rates of GLP-1 secretion, frequently associated with T2DM and NASH, may be also be caused by splanchnic hypoglycaemia, rather than to intrinsic loss of incretin secretory capacity. These findings may have therapeutic implications on GLP-1 agonist or glucagon antagonist usage.
Collapse
Affiliation(s)
- Richard J Naftalin
- Departments of Physiology and Vascular Biology, BHF centre of research excellence, King's College London School of Medicine, London, UK
| |
Collapse
|
118
|
Portillo-Sanchez P, Cusi K. Treatment of Nonalcoholic Fatty Liver Disease (NAFLD) in patients with Type 2 Diabetes Mellitus. Clin Diabetes Endocrinol 2016; 2:9. [PMID: 28702244 PMCID: PMC5471954 DOI: 10.1186/s40842-016-0027-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/28/2016] [Indexed: 02/08/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is believed to be the most common chronic liver disease, affecting at least one-third of the population worldwide. The more aggressive form is known as nonalcoholic steatohepatitis (NASH) and characterized by hepatocyte necrosis and inflammation. The presence of fibrosis is not uncommon. Fibrosis indicates a more aggressive course and patients with NASH that are at high-risk of cirrhosis and premature mortality, as well as at increased risk of hepatocellular carcinoma (HCC). Patients with type 2 diabetes mellitus (T2DM) are at the highest risk for the development of NASH, even in the setting of normal plasma aminotransferase levels. The presence of dysfunctional adipose tissue in most overweight and obese subjects, combined with insulin resistance, hyperglycemia, and atherogenic dyslipidemia, contribute to their increased cardiovascular risk. Many therapeutic agents have been tested for the treatment of NASH but few studies have focused in patients with T2DM. At the present moment, the only FDA-approved agents that in controlled studies have shown to significantly improve liver histology in patients with diabetes are pioglitazone and liraglutide. Current research efforts are centering on the mechanisms for intrahepatic triglyceride accumulation and for the development of steatohepatitis, the role of mitochondrial dysfunction in NASH, and the impact of improving glycemic control per se on the natural history of the disease. This brief review summarizes our current knowledge on the pharmacological agents available for the treatment of NASH to assist healthcare providers in the management of these challenging patients.
Collapse
Affiliation(s)
- Paola Portillo-Sanchez
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, 1600 SW Archer Road, room H-2, Gainesville, FL 32610 USA
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, University of Florida, 1600 SW Archer Road, room H-2, Gainesville, FL 32610 USA
- Division of Endocrinology, Diabetes, and Metabolism, Malcom Randall Veterans Affairs Medical Center, Gainesville, FL 32608 USA
| |
Collapse
|
119
|
Vanderheiden A, Harrison LB, Warshauer JT, Adams-Huet B, Li X, Yuan Q, Hulsey K, Dimitrov I, Yokoo T, Jaster AW, Pinho DF, Pedrosa I, Lenkinski RE, Pop LM, Lingvay I. Mechanisms of Action of Liraglutide in Patients With Type 2 Diabetes Treated With High-Dose Insulin. J Clin Endocrinol Metab 2016; 101:1798-806. [PMID: 26909799 DOI: 10.1210/jc.2015-3906] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
CONTEXT The mechanisms of action of incretin mimetics in patients with long-standing type 2 diabetes (T2D) and high insulin requirements have not been studied. OBJECTIVE To evaluate changes in β-cell function, glucagon secretion, and fat distribution after addition of liraglutide to high-dose insulin. DESIGN A single-center, randomized, double-blind, placebo-controlled trial. SETTING University of Texas Southwestern and Parkland Memorial Hospital clinics. PATIENTS Seventy-one patients with long-standing (median, 17 years) T2D requiring high-dose insulin treatment (>1.5 U/kg/d; average, 2.2 ± 0.9 U/kg/d). INTERVENTION Patients were randomized to liraglutide 1.8 mg/d or matching placebo for 6 months. MAIN OUTCOME MEASURES We measured changes in insulin and glucagon secretion using a 4-hour mixed-meal challenge test. Magnetic resonance-based techniques were used to estimate sc and visceral fat in the abdomen and ectopic fat in the liver and pancreas. RESULTS Glycosylated hemoglobin improved significantly with liraglutide treatment, with an end-of-trial estimated treatment difference between groups of −0.9% (95% confidence interval, −1.5, −0.4%) (P = .002). Insulin secretion improved in the liraglutide group vs placebo, as measured by the area under the curve of C-peptide (P = .002) and the area under the curves ratio of C-peptide to glucose (P = .003). Insulin sensitivity (Matsuda index) and glucagon secretion did not change significantly between groups. Liver fat and sc fat decreased in the liraglutide group vs placebo (P = .0006 and P = .01, respectively), whereas neither visceral nor pancreatic fat changed significantly. CONCLUSIONS Treatment with liraglutide significantly improved insulin secretion, even in patients with long-standing T2D requiring high-dose insulin treatment. Liraglutide also decreased liver and sc fat, but it did not alter glucagon secretion.
Collapse
Affiliation(s)
- Anna Vanderheiden
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Lindsay B Harrison
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Jeremy T Warshauer
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Beverley Adams-Huet
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Xilong Li
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Qing Yuan
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Keith Hulsey
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Ivan Dimitrov
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Takeshi Yokoo
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Adam W Jaster
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Daniella F Pinho
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Ivan Pedrosa
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Robert E Lenkinski
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Laurentiu M Pop
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Ildiko Lingvay
- Department of Internal Medicine (A.V., L.B.H., J.T.W., B.A.-H., L.M.P., I.L.), Division of Endocrinology, University of Texas Southwestern Medical Center, Dallas, Texas 75390; Texas Diabetes & Endocrinology (L.B.H.), Austin, Texas 78749; Departments of Clinical Sciences (B.A.-H., X.L., I.L.) and Radiology (Q.Y., K.H., T.Y., A.W.J., D.F.P., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390; Philips Medical Systems (I.D.), Cleveland, Ohio 44143; and Advanced Imaging Research Center (T.Y., I.P., R.E.L.), University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
120
|
Smits MM, van Raalte DH, Tonneijck L, Muskiet MHA, Kramer MHH, Cahen DL. GLP-1 based therapies: clinical implications for gastroenterologists. Gut 2016; 65:702-11. [PMID: 26786687 DOI: 10.1136/gutjnl-2015-310572] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/23/2015] [Indexed: 12/13/2022]
Abstract
The gut-derived incretin hormone, glucagon-like peptide 1 (GLP-1) lowers postprandial blood glucose levels by stimulating insulin and inhibiting glucagon secretion. Two novel antihyperglycaemic drug classes augment these effects; GLP-1 receptor agonists and inhibitors of the GLP-1 degrading enzyme dipeptidyl peptidase 4. These so called GLP-1 based or incretin based drugs are increasingly used to treat type 2 diabetes, because of a low risk of hypoglycaemia and favourable effect on body weight, blood pressure and lipid profiles. Besides glucose control, GLP-1 functions as an enterogastrone, causing a wide range of GI responses. Studies have shown that endogenous GLP-1 and its derived therapies slow down digestion by affecting the stomach, intestines, exocrine pancreas, gallbladder and liver. Understanding the GI actions of GLP-1 based therapies is clinically relevant; because GI side effects are common and need to be recognised, and because these drugs may be used to treat GI disease.
Collapse
Affiliation(s)
- Mark M Smits
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Daniel H van Raalte
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Lennart Tonneijck
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Marcel H A Muskiet
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Mark H H Kramer
- Department of Internal Medicine, Diabetes Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Djuna L Cahen
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
121
|
García Díaz E, Guagnozzi D, Gutiérrez V, Mendoza C, Maza C, Larrañaga Y, Perdomo D, Godoy T, Taleb G. Effect of incretin therapies compared to pioglitazone and gliclazide in non-alcoholic fatty liver disease in diabetic patients not controlled on metformin alone: An observational, pilot study. ACTA ACUST UNITED AC 2016; 63:194-201. [PMID: 26976710 DOI: 10.1016/j.endonu.2016.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 12/31/2015] [Accepted: 01/04/2016] [Indexed: 02/07/2023]
Abstract
AIM To compare the effect of different hypoglycemic drugs on laboratory and ultrasonographic markers of non-alcoholic fatty liver disease (NAFLD) in patients with type 2 diabetes not controlled on metformin alone. METHODS Prospective study of diabetic patients treated with metformin in combination with gliclazide, pioglitazone, sitagliptin, exenatide, or liraglutide. NAFLD was assessed by abdominal ultrasound and NAFLD fibrosis score was calculated at baseline and 6 months. RESULTS Fifty-eight patients completed 6 months of follow-up: 15 received gliclazide, 13 pioglitazone, 15 sitagliptin, 7 exenatide, and 8 liraglutide. NAFLD affected 57.8% of patients at baseline, and its ultrasonographic course varied depending on changes in weight (P=.009) and waist circumference (P=.012). The proportions of patients who experienced ultrasonographic improvement in the different treatment groups were: 33.3% with gliclazide, 37.5% with pioglitazone, 45.5% with sitagliptin, 80% with exenatide, and 33% with liraglutide (P=.28). CONCLUSIONS Qualitative ultrasonographic NAFLD improvement in diabetic patients treated with metformin in combination with other hypoglycemic drugs is associated to change over time in weight and waist circumference. Long-term clinical trials are needed to assess whether incretin therapies result in better liver outcomes than other hypoglycemic therapies.
Collapse
Affiliation(s)
- Eduardo García Díaz
- Unidad de Endocrinología, Hospital Dr. José Molina Orosa, Lanzarote, Canarias, España.
| | - Danila Guagnozzi
- Unidad de Gastroenterología, Hospital Vall d'Hebron, Barcelona, España
| | - Verónica Gutiérrez
- Unidad de Atención Primaria, Gerencia de Servicios Sanitarios de Lanzarote, Lanzarote, Canarias, España
| | - Carmen Mendoza
- Unidad de Atención Primaria, Gerencia de Servicios Sanitarios de Lanzarote, Lanzarote, Canarias, España
| | - Cristina Maza
- Unidad de Atención Primaria, Gerencia de Servicios Sanitarios de Lanzarote, Lanzarote, Canarias, España
| | - Yulene Larrañaga
- Unidad de Atención Primaria, Gerencia de Servicios Sanitarios de Lanzarote, Lanzarote, Canarias, España
| | - Dolores Perdomo
- Unidad de Atención Primaria, Gerencia de Servicios Sanitarios de Lanzarote, Lanzarote, Canarias, España
| | - Teresa Godoy
- Unidad de Atención Primaria, Gerencia de Servicios Sanitarios de Lanzarote, Lanzarote, Canarias, España
| | - Ghalli Taleb
- Unidad de Atención Primaria, Gerencia de Servicios Sanitarios de Lanzarote, Lanzarote, Canarias, España
| |
Collapse
|
122
|
Savvidou S, Karatzidou K, Tsakiri K, Gagalis A, Hytiroglou P, Goulis J. Circulating adiponectin levels in type 2 diabetes mellitus patients with or without non-alcoholic fatty liver disease: Results of a small, open-label, randomized controlled intervention trial in a subgroup receiving short-term exenatide. Diabetes Res Clin Pract 2016; 113:125-34. [PMID: 26803355 DOI: 10.1016/j.diabres.2015.12.003] [Citation(s) in RCA: 14] [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: 07/10/2015] [Revised: 10/19/2015] [Accepted: 12/27/2015] [Indexed: 02/08/2023]
Abstract
AIM Diabetes mellitus type 2 (DMT2) and non-alcoholic fatty liver disease (NAFLD) are both characterized by decreased circulating adiponectin. Recently, glucagon-like peptide-1 receptor agonists have been shown to induce adiponectin's expression. However, their interaction on clinical grounds needs to be further elucidated. METHODS DMT2 patients with abnormal aminotransferases were screened for NAFLD and subjected to liver biopsy (group A, n=17). A subgroup of patients (n=110), after assessed for eligibility criteria, was blindly randomized to receive either 6-month exenatide supplementation on glargine insulin (group B) or intense, self-regulated, insulin therapy alone (group C). RESULTS Baseline patient characteristics: 49(38.6%) males, aged 63.1 ± 7.5 years-old, BMI 32.9 ± 4.9 kg/m(2), HbA1c 8.1 ± 1.2% (65 ± 14 mmol/mol), median ALT 23 U/L (range 5-126), AST 20 U/L (7-72). Group A had biopsy-proven NAFLD with a median Activity Score of 5 and fibrosis stage 3. Presence of NAFLD was accompanied by a significant decline in adiponectin (p<0.001), which was negatively correlated with the degree of ALT in all groups (Spearman's correlation, rs=-0.644, p<0.001). In the subgroup intervention trial, adiponectin was significantly raised in both groups B and C (t-Student for paired samples, p=0.001) by Δ=+24.2% (interquartile range 14.8-53.2%). This elevation was not associated with the type of intervention but with weight loss, glycemic control and reduction of C-reactive protein (one-way ANCOVA). CONCLUSION Supplementation of exenatide to glargine insulin compared to standard insulin was: (i) effective in inducing weight loss, (ii) non-inferior in lowering HbA1c and (iii) non-inferior in increasing circulating adiponectin. Higher adiponectin was associated with lower ALT, suggesting a hepato-protective role for this cytokine.
Collapse
Affiliation(s)
- Savvoula Savvidou
- Medical Center of Diabetes Mellitus, 1st Department of Internal Medicine, "Papageorgiou" University Hospital of Thessaloniki, Greece; Hepatology Outpatients' Clinic, "Papageorgiou" University Hospital of Thessaloniki, Greece; 4th Department of Internal Medicine, Hippocration University Hospital of Thessaloniki, Greece.
| | - Kyparissia Karatzidou
- Medical Center of Diabetes Mellitus, 1st Department of Internal Medicine, "Papageorgiou" University Hospital of Thessaloniki, Greece
| | - Kalliopi Tsakiri
- Medical Center of Diabetes Mellitus, 1st Department of Internal Medicine, "Papageorgiou" University Hospital of Thessaloniki, Greece
| | - Asterios Gagalis
- Hepatology Outpatients' Clinic, "Papageorgiou" University Hospital of Thessaloniki, Greece
| | - Prodromos Hytiroglou
- Department of Pathology, Medical School of Aristotle University of Thessaloniki, Greece
| | - John Goulis
- 4th Department of Internal Medicine, Hippocration University Hospital of Thessaloniki, Greece
| |
Collapse
|
123
|
A Comprehensive Updated Review of Pharmaceutical and Nonpharmaceutical Treatment for NAFLD. Gastroenterol Res Pract 2016; 2016:7109270. [PMID: 27006654 PMCID: PMC4781972 DOI: 10.1155/2016/7109270] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/27/2016] [Indexed: 02/08/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease in the western world with prevalence of 20–33%. NAFLD comprises a pathological spectrum. Nonalcoholic fatty liver (NAFL) is at one end and consists of simple hepatic steatosis. On the contrary, nonalcoholic steatohepatitis (NASH) consists of steatosis, inflammation, and ballooning degeneration and can progress to cirrhosis. Despite the rising incidence, definitive treatment for NAFLD, specifically NASH, has not yet been established. Lifestyle modification with dietary changes combined with regular aerobic exercise, along with multidisciplinary approach including cognitive behavior therapy, has been shown to be an effective therapeutic option, even without a significant weight loss. Pioglitazone and vitamin E have shown to be most effective in NASH patients. Surgery and weight loss medication are effective means of weight loss but can potentially worsen NASH related fibrosis. Other agents such as n-3 polyunsaturated fatty acids, probiotics, and pentoxifylline along with herbal agent such as milk thistle as well as daily intake of coffee have shown potential benefits, but further well organized studies are definitely warranted. This review focuses on the available evidence on pharmaceutical and nonpharmaceutical therapy in the treatment and the prevention of NAFLD, primarily NASH.
Collapse
|
124
|
Rajeev SP, Cuthbertson DJ, Wilding JPH. Energy balance and metabolic changes with sodium-glucose co-transporter 2 inhibition. Diabetes Obes Metab 2016; 18:125-34. [PMID: 26403227 DOI: 10.1111/dom.12578] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 08/26/2015] [Accepted: 09/18/2015] [Indexed: 12/25/2022]
Abstract
Sodium-glucose co-transporter 2 (SGLT2) inhibitors are the latest addition to the class of oral glucose-lowering drugs. They have been rapidly adopted into clinical practice because of therapeutic advantages, including weight loss and reduction in blood pressure, in addition to glycaemic benefits and a low intrinsic risk of hypoglycaemia. Although there are extensive data on the clinical effects of SGLT2 inhibition, the metabolic effects of inhibiting renal glucose reabsorption have not been fully described. Recent studies have identified compensatory metabolic effects, such as an increase in endogenous glucose production, and have also shown an increase in glucagon secretion during SGLT2 inhibition. In addition, there is a discrepancy between the expected and observed weight loss found in clinical studies on SGLT2 inhibitors, probably as a result of changes in energy balance with this treatment approach. SGLT2 inhibition is likely to have intriguing effects on whole body metabolism which have not been fully elucidated, and which, if explained, might help optimize the use of this new class of medicines.
Collapse
Affiliation(s)
- S P Rajeev
- Obesity and Endocrinology Research Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Aintree University Hospital NHS Foundation Trust, Liverpool, UK
| | - D J Cuthbertson
- Obesity and Endocrinology Research Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Aintree University Hospital NHS Foundation Trust, Liverpool, UK
| | - J P H Wilding
- Obesity and Endocrinology Research Group, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Aintree University Hospital NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
125
|
Tang W, Xu Q, Hong T, Tong G, Feng W, Shen S, Bi Y, Zhu D. Comparative efficacy of anti-diabetic agents on nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus: a systematic review and meta-analysis of randomized and non-randomized studies. Diabetes Metab Res Rev 2016; 32:200-16. [PMID: 26381272 DOI: 10.1002/dmrr.2713] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 07/10/2015] [Accepted: 07/23/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) has a high prevalence in patients with type 2 diabetes mellitus (T2DM). In this study, we sought to provide a comprehensive assessment regarding the effects of anti-diabetic agents on NAFLD in patients with T2DM. METHODS MEDLINE, EMBASE and the Cochrane Central Register of Controlled Trials were searched for randomized controlled trials (RCTs) with different anti-diabetic agents in T2DM. Observational trials were also recruited to expand our population. Hepatic fat content and liver histology were evaluated as primary outcomes. Pooled estimates were calculated using a fixed effect model. RESULTS One thousand one hundred ninety-six participants in 19 RCTs and 14 non-randomized studies were included. Evidence from RCTs and observational studies suggested that greater hepatic fat content reduction and improved liver histology were seen in thiazolidinediones for 12-72 weeks; glucagon-like peptide-1 receptor agonists had beneficial effects on hepatic fat content after 26-50 weeks intervention, and insulin/metformin combination with 3-7 months improved hepatic fat content. Initiating metformin or dapagliflozin showed no benefit on hepatic fat content or liver histology in 16-48 weeks. Besides, nateglinide for 18 months was reported in a small sample-size RCT to improve hepatic fat content and liver histology. Sitagliptin therapy of 1 year also provided benefit on nonalcoholic steatohepatitis score in an observational study. CONCLUSIONS For T2DM with NAFLD, administrating thiazolidinediones and glucagon-like peptide-1 receptor agonists seems to provide more identified advances in attenuating hepatic fat content. Further RCTs are warranted to assess the efficacy of various hypoglycemic agents on clinical outcomes associated with NAFLD in T2DM. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Wenjuan Tang
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Qianyue Xu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Ting Hong
- Department of Endocrinology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Guoyu Tong
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Wenhuan Feng
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Shanmei Shen
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
| | - Dalong Zhu
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, China
- Department of Endocrinology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
126
|
Gastaldelli A, Marchesini G. Time for Glucagon like peptide-1 receptor agonists treatment for patients with NAFLD? J Hepatol 2016; 64:262-264. [PMID: 26643784 DOI: 10.1016/j.jhep.2015.11.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 11/26/2015] [Accepted: 11/27/2015] [Indexed: 01/12/2023]
|
127
|
Carbone LJ, Angus PW, Yeomans ND. Incretin-based therapies for the treatment of non-alcoholic fatty liver disease: A systematic review and meta-analysis. J Gastroenterol Hepatol 2016; 31:23-31. [PMID: 26111358 DOI: 10.1111/jgh.13026] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 06/02/2015] [Accepted: 06/09/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIM Non-alcoholic fatty liver disease (NAFLD) is a leading cause of chronic liver disease in Western societies. Despite its significance, there are no well-proven pharmacological treatments. Two novel classes of potential pharmacotherapies are the glucagon-like peptide-1 receptor agonists (GLP-1 RA) and dipeptidyl peptidase-4 inhibitors (DPP-4I), collectively known as incretin-based therapies. These have several metabolic and anti-inflammatory actions that may be of benefit in NAFLD. The aim of this meta-analysis was to evaluate their efficacy via a structured retrieval and pooled analysis of relevant studies. METHODS Studies were sourced from electronic databases and meeting abstracts. Main inclusion criteria were original studies investigating treatment of adults with NAFLD using GLP-1 RA/DPP-4I. Key outcomes were a change in serum alanine transaminase (ALT), as a marker of liver inflammation, and improvement in disease status measured by imaging or histology. RESULTS Initial searching retrieved 1357 peer-reviewed articles and abstracts. Four studies met all inclusion and exclusion criteria. There were a total of 136 participants with NAFLD and concomitant type 2 diabetes mellitus (T2DM). Meta-analysis (random-effects model) revealed a significant decrease in serum ALT following treatment (mean reduction 14.1 IU/L, 95% confidence intervals [CI] 8.3-19.8, P < 0.0001). In two studies with imaging and tissue data, treatment was found to significantly reduce steatosis, inflammation, and fibrosis. CONCLUSION The significant decrease in a key biochemical marker of hepatic inflammation following treatment with incretin-based therapies, as well as improvements in imaging and histology, suggests these agents may be effective options for managing NAFLD with comorbid T2DM.
Collapse
Affiliation(s)
- Laura J Carbone
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Peter W Angus
- Department of Gastroenterology and Hepatology, Austin Health, Melbourne, Victoria, Australia
| | - Neville D Yeomans
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
128
|
Li D, Xu X, Zhang Y, Zhu J, Ye L, Lee KO, Ma J. Liraglutide treatment causes upregulation of adiponectin and downregulation of resistin in Chinese type 2 diabetes. Diabetes Res Clin Pract 2015; 110:224-228. [PMID: 26376464 DOI: 10.1016/j.diabres.2015.05.051] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 05/13/2015] [Accepted: 05/28/2015] [Indexed: 01/02/2023]
Abstract
AIMS To assess the effect of 16 weeks of liraglutide administration on the plasma levels of adiponectin and resistin in Chinese patients diagnosed with type 2 diabetes mellitus (T2DM). METHODS Forty-four subjects were recruited and randomly assigned to once-a-day dosage of either liraglutide, or glimepiride (4 mg) in a double-blinded double-dummy active-controlled study. All treatments were administered in combination with metformin (1 g, twice daily). The efficacy of liraglutide was estimated by measuring and comparing the levels of HbA1c, adiponectin and resistin in the plasma before and after the 16-week treatment. RESULTS The plasma level of adiponectin was significantly increased (0.74±0.19 ng/ml, p<0.05) and resistin was significantly lowered (-1.34±0.34 pg/ml, p<0.05) in a dose-dependent manner in the liraglutide group when compared with the glimepiride group (-0.44±0.09 ng/ml of adiponectin and 0.14±0.41 pg/ml of resistin). In contrast, we found no differences in the decrease in HbA1c between the two groups (8.3±1.2% to 7.2±1.1% in NGSP units vs. 8.3±1.0% to 7.3±1.2% in NGSP units; 67±13 mmol/mol to 55±12 mmol/mol vs. 67±11 mmol/mol to 56±13 mmol/mol in IFCC units). CONCLUSIONS In Chinese T2DM patients, liraglutide treatment led to increased adiponectin and decreased resistin levels compared to glimepiride-treated subjects, while inducing similar glycemic changes.
Collapse
Affiliation(s)
- Dongmei Li
- Department of Endocrinology, Nanjing Medical University Affiliated Nanjing First Hospital, No. 68 Changle Road, Nanjing, China
| | - Xiaohua Xu
- Department of Endocrinology, Nanjing Medical University Affiliated Nanjing First Hospital, No. 68 Changle Road, Nanjing, China
| | - Ying Zhang
- Department of Endocrinology, Nanjing Medical University Affiliated Nanjing First Hospital, No. 68 Changle Road, Nanjing, China
| | - Jian Zhu
- Department of Endocrinology, Nanjing Medical University Affiliated Nanjing First Hospital, No. 68 Changle Road, Nanjing, China
| | - Lei Ye
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Kok Onn Lee
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Jianhua Ma
- Department of Endocrinology, Nanjing Medical University Affiliated Nanjing First Hospital, No. 68 Changle Road, Nanjing, China.
| |
Collapse
|
129
|
DiNicolantonio JJ, Bhutani J, O'Keefe JH. Acarbose: safe and effective for lowering postprandial hyperglycaemia and improving cardiovascular outcomes. Open Heart 2015; 2:e000327. [PMID: 26512331 PMCID: PMC4620230 DOI: 10.1136/openhrt-2015-000327] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 09/23/2015] [Accepted: 09/27/2015] [Indexed: 12/17/2022] Open
Abstract
α-Glucosidase inhibitors (AGIs) are a class of oral glucose-lowering drugs used exclusively for treatment or prevention of type 2 diabetes mellitus. AGIs act by altering the intestinal absorption of carbohydrates through inhibition of their conversion into simple sugars (monosaccharides) and thus decrease the bioavailability of carbohydrates in the body, significantly lowering blood glucose levels. The three AGIs used in clinical practice are acarbose, voglibose and miglitol. This review will focus on the cardiovascular properties of acarbose. The current available data suggest that AGIs (particularly acarbose) may be safe and effective for the treatment of prediabetes and diabetes.
Collapse
Affiliation(s)
| | | | - James H O'Keefe
- Saint Luke's Mid America Heart Institute , Kansas City, Missouri , USA
| |
Collapse
|
130
|
Dissociation between exercise-induced reduction in liver fat and changes in hepatic and peripheral glucose homoeostasis in obese patients with non-alcoholic fatty liver disease. Clin Sci (Lond) 2015; 130:93-104. [PMID: 26424731 DOI: 10.1042/cs20150447] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/30/2015] [Indexed: 12/22/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is associated with multi-organ (hepatic, skeletal muscle, adipose tissue) insulin resistance (IR). Exercise is an effective treatment for lowering liver fat but its effect on IR in NAFLD is unknown. We aimed to determine whether supervised exercise in NAFLD would reduce liver fat and improve hepatic and peripheral (skeletal muscle and adipose tissue) insulin sensitivity. Sixty nine NAFLD patients were randomized to 16 weeks exercise supervision (n=38) or counselling (n=31) without dietary modification. All participants underwent MRI/spectroscopy to assess changes in body fat and in liver and skeletal muscle triglyceride, before and following exercise/counselling. To quantify changes in hepatic and peripheral insulin sensitivity, a pre-determined subset (n=12 per group) underwent a two-stage hyperinsulinaemic euglycaemic clamp pre- and post-intervention. Results are shown as mean [95% confidence interval (CI)]. Fifty participants (30 exercise, 20 counselling), 51 years (IQR 40, 56), body mass index (BMI) 31 kg/m(2) (IQR 29, 35) with baseline liver fat/water % of 18.8% (IQR 10.7, 34.6) completed the study (12/12 exercise and 7/12 counselling completed the clamp studies). Supervised exercise mediated a greater reduction in liver fat/water percentage than counselling [Δ mean change 4.7% (0.01, 9.4); P<0.05], which correlated with the change in cardiorespiratory fitness (r=-0.34, P=0.0173). With exercise, peripheral insulin sensitivity significantly increased (following high-dose insulin) despite no significant change in hepatic glucose production (HGP; following low-dose insulin); no changes were observed in the control group. Although supervised exercise effectively reduced liver fat, improving peripheral IR in NAFLD, the reduction in liver fat was insufficient to improve hepatic IR.
Collapse
|
131
|
Dobson R, Burgess MI, Sprung VS, Irwin A, Hamer M, Jones J, Daousi C, Adams V, Kemp GJ, Shojaee-Moradie F, Umpleby M, Cuthbertson DJ. Metabolically healthy and unhealthy obesity: differential effects on myocardial function according to metabolic syndrome, rather than obesity. Int J Obes (Lond) 2015; 40:153-61. [PMID: 26271188 DOI: 10.1038/ijo.2015.151] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/09/2015] [Accepted: 08/03/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND The term 'metabolically healthy obese (MHO)' is distinguished using body mass index (BMI), yet BMI is a poor index of adiposity. Some epidemiological data suggest that MHO carries a lower risk of cardiovascular disease (CVD) or mortality than being normal weight yet metabolically unhealthy. OBJECTIVES We aimed to undertake a detailed phenotyping of individuals with MHO by using imaging techniques to examine ectopic fat (visceral and liver fat deposition) and myocardial function. We hypothesised that metabolically unhealthy individuals (irrespective of BMI) would have adverse levels of ectopic fat and myocardial dysfunction compared with MHO individuals. SUBJECTS Individuals were categorised as non-obese or obese (BMI ⩾30 kg m(-2)) and as metabolically healthy or unhealthy according to the presence or absence of metabolic syndrome. METHODS Sixty-seven individuals (mean±s.d.: age 49±11 years) underwent measurement of (i) visceral, subcutaneous and liver fat using magnetic resonance imaging and proton magnetic resonance spectroscopy, (ii) components of metabolic syndrome, (iii) cardiorespiratory fitness and (iv) indices of systolic and diastolic function using tissue Doppler echocardiography. RESULTS Cardiorespiratory fitness was similar between all groups; abdominal and visceral fat was highest in the obese groups. Compared with age- and BMI-matched metabolically healthy counterparts, the unhealthy (lean or obese) individuals had higher liver fat and decreased early diastolic strain rate, early diastolic tissue velocity and systolic strain indicative of subclinical systolic and diastolic dysfunction. The magnitude of dysfunction correlated with the number of components of metabolic syndrome but not with BMI or with the degree of ectopic (visceral or liver) fat deposition. CONCLUSIONS Myocardial dysfunction appears to be related to poor metabolic health rather than simply BMI or fat mass. These data may partly explain the epidemiological evidence on CVD risk relating to the different obesity phenotypes.
Collapse
Affiliation(s)
- R Dobson
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - M I Burgess
- Department of Cardiology, University Hospital Aintree, Liverpool, UK
| | - V S Sprung
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - A Irwin
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - M Hamer
- National Centre Sport and Exercise Medicine, Loughborough University, Loughborough, UK
| | - J Jones
- Department of Cardiology, University Hospital Aintree, Liverpool, UK
| | - C Daousi
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - V Adams
- Magnetic Resonance and Image Analysis Research Centre, University of Liverpool, Liverpool, UK
| | - G J Kemp
- Magnetic Resonance and Image Analysis Research Centre, University of Liverpool, Liverpool, UK.,Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - F Shojaee-Moradie
- Diabetes and Metabolic Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - M Umpleby
- Diabetes and Metabolic Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - D J Cuthbertson
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| |
Collapse
|
132
|
Rizvi AA, Patti AM, Giglio RV, Nikolic D, Amato A, Al-Busaidi N, Al-Rasadi K, Soresi M, Banach M, Montalto G, Rizzo M. Liraglutide improves carotid intima-media thickness in patients with type 2 diabetes and non-alcoholic fatty liver disease: an 8-month prospective pilot study. Expert Opin Biol Ther 2015; 15:1391-1397. [PMID: 26195184 DOI: 10.1517/14712598.2015.1067299] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To explore the effects of the glucagon-like peptide-1 receptor analogue liraglutide on subclinical atherosclerosis in diabetic subjects with non-alcoholic fatty liver disease (NAFLD). RESEARCH DESIGN AND METHODS In this 8-month prospective study, 29 subjects with type 2 diabetes (T2DM) and NAFLD (16 men and 13 women, mean age: 61 ± 10 years) were matched for age and gender with 29 subjects with T2DM without NAFLD (16 men and 13 women, mean age: 61 ± 8 years). Liraglutide 0.6 mg/day for 2 weeks, followed by 1.2 mg/day, was given in addition to metformin. MAIN OUTCOME MEASURES Anthropometric variables, glucometabolic parameters and carotid intima-media thickness (IMT) using B-mode real-time ultrasound were assessed at baseline and 4 and 8 months. RESULTS Glycated hemoglobin reduced significantly in both groups. No significant changes were found in body weight, waist circumference and lipids. Carotid IMT decreased significantly in the T2DM patients with NAFLD (from 0.96 ± 0.27 to 0.82 ± 0.17 to 0.85 ± 0.12 mm, p = 0.0325), but not in the T2DM patients without NAFLD (from 0.91 ± 0.23 to 0.88 ± 0.17 to 0.85 ± 0.15 mm, p = 0.4473). CONCLUSION Eight months of liraglutide use in patients with T2DM and NAFLD significantly reduced carotid IMT, a surrogate marker of atherosclerosis, independently of glucometabolic changes.
Collapse
Affiliation(s)
- Ali A Rizvi
- a 1 University of South Carolina School of Medicine, Division of Endocrinology, Diabetes and Metabolism , Columbia, SC, USA
| | - Angelo Maria Patti
- b 2 University of Palermo, Biomedical Department of Internal Medicine and Medical Specialties , Via del Vespro, 141, 90127, Palermo, Italy +39 091 6552945 ; +39 091 6552945 ;
| | - Rosaria Vincenza Giglio
- b 2 University of Palermo, Biomedical Department of Internal Medicine and Medical Specialties , Via del Vespro, 141, 90127, Palermo, Italy +39 091 6552945 ; +39 091 6552945 ;
| | - Dragana Nikolic
- b 2 University of Palermo, Biomedical Department of Internal Medicine and Medical Specialties , Via del Vespro, 141, 90127, Palermo, Italy +39 091 6552945 ; +39 091 6552945 ;
| | - Antonella Amato
- c 3 University of Palermo, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies , Palermo, Italy
| | - Noor Al-Busaidi
- d 4 Sultan Qaboos University Hospital, Department of Clinical Biochemistry , Muscat, Oman
| | - Khalid Al-Rasadi
- d 4 Sultan Qaboos University Hospital, Department of Clinical Biochemistry , Muscat, Oman
| | - Maurizio Soresi
- b 2 University of Palermo, Biomedical Department of Internal Medicine and Medical Specialties , Via del Vespro, 141, 90127, Palermo, Italy +39 091 6552945 ; +39 091 6552945 ;
| | - Maciej Banach
- e 5 University of Lodz, Department of Hypertension, Chair of Nephrology and Hypertension , Lodz, Poland
| | - Giuseppe Montalto
- b 2 University of Palermo, Biomedical Department of Internal Medicine and Medical Specialties , Via del Vespro, 141, 90127, Palermo, Italy +39 091 6552945 ; +39 091 6552945 ;
| | - Manfredi Rizzo
- a 1 University of South Carolina School of Medicine, Division of Endocrinology, Diabetes and Metabolism , Columbia, SC, USA
- b 2 University of Palermo, Biomedical Department of Internal Medicine and Medical Specialties , Via del Vespro, 141, 90127, Palermo, Italy +39 091 6552945 ; +39 091 6552945 ;
| |
Collapse
|
133
|
Kato H, Nagai Y, Ohta A, Tenjin A, Nakamura Y, Tsukiyama H, Sasaki Y, Fukuda H, Ohshige T, Terashima Y, Sada Y, Kondo A, Sasaoka T, Tanaka Y. Effect of sitagliptin on intrahepatic lipid content and body fat in patients with type 2 diabetes. Diabetes Res Clin Pract 2015; 109:199-205. [PMID: 25934525 DOI: 10.1016/j.diabres.2015.04.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 02/07/2015] [Accepted: 04/12/2015] [Indexed: 12/18/2022]
Abstract
AIMS To evaluate the effect of the DPP-4 inhibitor sitagliptin on intrahepatic lipid (IHL) content and body fat in overweight Japanese patients with type 2 diabetes. METHODS A prospective, 24-week, single-center, open-label comparative study enrolled 20 Japanese patients with type 2 diabetes (male: 11, female: 9) with a BMI≥25 kg/m(2) or fatty liver. Subjects were randomly assigned to receive treatment with sitagliptin (25 mg titrated up to 50 mg: S) or glimepiride (0.5 mg titrated up to 1 mg: G). After starting each treatment, IHL and total fat mass were evaluated by (1)H-magnetic resonance spectroscopy ((1)H-MRS) and dual energy X-ray absorptiometry (DEXA), respectively at baseline and at 12 weeks and 24 weeks. RESULTS After 24 weeks, HbA1c levels showed a similar significant decrease in both groups from 7.2 (7.0, 7.5) to 6.6 (6.4, 6.8)%, (54 (53, 56) to 48(47, 49) mmol/mol) with S and 7.3(6.8, 7.4) to 6.6 (6.3, 6.7)%, (55 (51, 56) to 48 (46, 49) mmol/mol) with G, median (interquartile range), p<0.05 vs. baseline, with no significant differences between the two groups. The IHL and total body fat mass were decreased in S group from 24.5(18.9, 36.6) to 20.5 (14.6, 28.5)% (p=0.009) and 22.5 (20.6, 33.7) to 21.6 (19.7, 32.4)kg (p=0.028), respectively, but not in G group. CONCLUSIONS Our findings indicate that sitagliptin and glimepiride achieved similar glycemic control, but only sitagliptin reduced IHL and total body fat (UMIN: 000013356).
Collapse
Affiliation(s)
- Hiroyuki Kato
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Yoshio Nagai
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan.
| | - Akio Ohta
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Ayumi Tenjin
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Yuta Nakamura
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Hidekazu Tsukiyama
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Yosuke Sasaki
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Hisashi Fukuda
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Toshihiko Ohshige
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Yuko Terashima
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Yukiyoshi Sada
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Akihiko Kondo
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| | - Toshiyasu Sasaoka
- Department of Clinical Pharmacology, University School of Toyama, 930-0194 Sugitani, Toyama, Japan
| | - Yasushi Tanaka
- Department of Internal Medicine, Division of Metabolism and Endocrinology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan
| |
Collapse
|
134
|
Tang A, Rabasa-Lhoret R, Castel H, Wartelle-Bladou C, Gilbert G, Massicotte-Tisluck K, Chartrand G, Olivié D, Julien AS, de Guise J, Soulez G, Chiasson JL. Effects of Insulin Glargine and Liraglutide Therapy on Liver Fat as Measured by Magnetic Resonance in Patients With Type 2 Diabetes: A Randomized Trial. Diabetes Care 2015; 38:1339-46. [PMID: 25813773 DOI: 10.2337/dc14-2548] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/09/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE This study determined the effects of insulin versus liraglutide therapy on liver fat in patients with type 2 diabetes inadequately controlled with oral agents therapy, including metformin. RESEARCH DESIGN AND METHODS Thirty-five patients with type 2 diabetes inadequately controlled on metformin monotherapy or in combination with other oral antidiabetic medications were randomized to receive insulin glargine or liraglutide therapy for 12 weeks. The liver proton density fat fraction (PDFF) was measured by MRS. The mean liver PDFF, the total liver volume, and the total liver fat index were measured by MRI. The Student t test, the Fisher exact test, and repeated-measures ANOVA were used for statistical analysis. RESULTS Insulin treatment was associated with a significant improvement in glycated hemoglobin (7.9% to 7.2% [62.5 to 55.2 mmol/mol], P = 0.005), a trend toward a decrease in MRS-PDFF (12.6% to 9.9%, P = 0.06), and a significant decrease in liver mean MRI-PDFF (13.8% to 10.6%, P = 0.005), liver volume (2,010.6 to 1,858.7 mL, P = 0.01), and the total liver fat index (304.4 vs. 209.3 % ⋅ mL, P = 0.01). Liraglutide treatment was also associated with a significant improvement in glycated hemoglobin (7.6% to 6.7% [59.8 to 50.2 mmol/mol], P < 0.001) but did not change MRS-PDFF (P = 0.80), liver mean MRI-PDFF (P = 0.15), liver volume (P = 0.30), or the total liver fat index (P = 0.39). CONCLUSIONS The administration of insulin glargine therapy reduced the liver fat burden in patients with type 2 diabetes. However, the improvements in the liver fat fraction and glycemia control were not significantly different from those in the liraglutide group.
Collapse
Affiliation(s)
- An Tang
- Department of Radiology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Rémi Rabasa-Lhoret
- Institut de Recherches Cliniques de Montréal, Montréal, Québec, Canada Department of Medicine and Endocrinology, Centre Hospitalier de l'Université de Montréal (CHUM), Hôpital Hotel-Dieu, Montréal, Québec, Canada
| | - Hélène Castel
- Department of Gastroenterology and Hepatology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Claire Wartelle-Bladou
- Department of Gastroenterology and Hepatology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Guillaume Gilbert
- Department of Radiology, Centre Hospitalier de l'Université de Montréal (CHUM), Hôpital Notre-Dame, Montréal, Québec, Canada MR Clinical Science, Philips Healthcare Canada, Markham, Ontario, Canada
| | - Karine Massicotte-Tisluck
- Department of Radiology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Gabriel Chartrand
- Imaging and Orthopaedics Research Laboratory (LIO), École de Technologie Supérieure, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Damien Olivié
- Department of Radiology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Anne-Sophie Julien
- Department of Mathematics and Statistics, Pavillon André-Aisenstadt, Université de Montréal, Montréal, Québec, Canada
| | - Jacques de Guise
- Imaging and Orthopaedics Research Laboratory (LIO), École de Technologie Supérieure, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Gilles Soulez
- Department of Radiology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, Québec, Canada Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Jean-Louis Chiasson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada Department of Medicine and Endocrinology, Centre Hospitalier de l'Université de Montréal (CHUM), Hôpital Hotel-Dieu, Montréal, Québec, Canada
| |
Collapse
|
135
|
Abstract
The rising prevalence of nonalcoholic fatty liver disease (NAFLD) is associated with the increasing global pandemic of obesity. These conditions cluster with type II diabetes mellitus and the metabolic syndrome to result in obesity-associated liver disease. The benefits of bariatric procedures on diabetes and the metabolic syndrome have been recognized for some time, and there is now mounting evidence to suggest that bariatric procedures improve liver histology and contribute to the beneficial resolution of NAFLD in obese patients. These beneficial effects derive from a number of weight-dependent and weight-independent mechanisms including surgical BRAVE actions (bile flow changes, restriction of stomach size, anatomical gastrointestinal rearrangement, vagal manipulation, enteric hormonal modulation) and subsequent effects such as reduced lipid intake, adipocytokine secretion, modulation of gut flora, improvements in insulin resistance and reduced inflammation. Here, we review the clinical investigations on bariatric procedures for NAFLD, in addition to the mounting mechanistic data supporting these findings. Elucidating the mechanisms by which bariatric procedures may resolve NAFLD can help enhance surgical approaches for metabolic hepatic dysfunction and also contribute toward developing the next generation of therapies aimed at reducing the burden of obesity-associated liver disease.
Collapse
|
136
|
Li CL, Zhao LJ, Zhou XL, Wu HX, Zhao JJ. Review on the effect of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors for the treatment of non-alcoholic fatty liver disease. ACTA ACUST UNITED AC 2015; 35:333-336. [PMID: 26072069 DOI: 10.1007/s11596-015-1433-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 03/20/2015] [Indexed: 01/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common liver disease and it represents the hepatic manifestation of metabolic syndrome, which includes type 2 diabetes mellitus (T2DM), dyslipidemia, central obesity and hypertension. Glucagon-like peptide-1 (GLP-1) analogues and dipeptidyl peptidase-4 (DPP-4) inhibitors were widely used to treat T2DM. These agents improve glycemic control, promote weight loss and improve lipid metabolism. Recent studies have demonstrated that the GLP-1 receptor (GLP-1R) is present and functional in human and rat hepatocytes. In this review, we present data from animal researches and human clinical studies that showed GLP-1 analogues and DPP-4 inhibitors can decrease hepatic triglyceride (TG) content and improve hepatic steatosis, although some effects could be a result of improvements in metabolic parameters. Multiple hepatocyte signal transduction pathways and mRNA from key enzymes in fatty acid metabolism appear to be activated by GLP-1 and its analogues. Thus, the data support the need for more rigorous prospective clinical trials to further investigate the potential of incretin therapies to treat patients with NAFLD.
Collapse
Affiliation(s)
- Chao-Lin Li
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
- Department of Endocrinology, Jinan, China
| | - Lu-Jie Zhao
- Hemodialysis Center, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Xin-Li Zhou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Hui-Xiao Wu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Jia-Jun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China.
| |
Collapse
|
137
|
Scheen AJ. Pharmacokinetics in patients with chronic liver disease and hepatic safety of incretin-based therapies for the management of type 2 diabetes mellitus. Clin Pharmacokinet 2015; 53:773-85. [PMID: 25091053 DOI: 10.1007/s40262-014-0157-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Patients with type 2 diabetes mellitus have an increased risk of chronic liver disease (CLD) such as non-alcoholic fatty liver disease and steatohepatitis, and about one-third of cirrhotic patients have diabetes. However, the use of several antidiabetic agents, such as metformin and sulphonylureas, may be a concern in case of hepatic impairment (HI). New glucose-lowering agents targeting the incretin system are increasingly used for the management of type 2 diabetes. Incretin-based therapies comprise oral inhibitors of dipeptidyl peptidase-4 (DPP-4) (gliptins) or injectable glucagon-like peptide-1 (GLP-1) receptor agonists. This narrative review summarises the available data regarding the use of both incretin-based therapies in patients with HI. In contrast to old glucose-lowering agents, they were evaluated in specifically designed acute pharmacokinetic studies in patients with various degrees of HI and their hepatic safety was carefully analysed in large clinical trials. Only mild changes in pharmacokinetic characteristics of DPP-4 inhibitors were observed in patients with different degrees of HI, presumably without major clinical relevance. GLP-1 receptor agonists have a renal excretion rather than liver metabolism. Specific pharmacokinetic data in patients with HI are only available for liraglutide. No significant changes in liver enzymes were reported with DPP-4 inhibitors or GLP-1 receptor agonists, alone or in combination with various other glucose-lowering agents, in clinical trials up to 2 years in length. On the contrary, preliminary data suggested that incretin-based therapies may be beneficial in patients with CLD, more particularly in the presence of non-alcoholic fatty liver disease. Nevertheless, caution should be recommended, especially in patients with advanced cirrhosis, because of a lack of clinical experience with incretin-based therapies in these vulnerable patients.
Collapse
Affiliation(s)
- André J Scheen
- Division of Diabetes, Nutrition and Metabolic Disorders, Department of Medicine, University of Liège, Liège, Belgium,
| |
Collapse
|
138
|
Jordy AB, Kraakman MJ, Gardner T, Estevez E, Kammoun HL, Weir JM, Kiens B, Meikle PJ, Febbraio MA, Henstridge DC. Analysis of the liver lipidome reveals insights into the protective effect of exercise on high-fat diet-induced hepatosteatosis in mice. Am J Physiol Endocrinol Metab 2015; 308:E778-91. [PMID: 25714675 DOI: 10.1152/ajpendo.00547.2014] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 02/15/2015] [Indexed: 01/14/2023]
Abstract
The accumulation of lipid at ectopic sites, including the skeletal muscle and liver, is a common consequence of obesity and is associated with tissue-specific and whole body insulin resistance. Exercise is well known to improve insulin resistance by mechanisms not completely understood. We performed lipidomic profiling via mass spectrometry in liver and skeletal muscle samples from exercise-trained mice to decipher the lipid changes associated with exercise-induced improvements in whole body glucose metabolism. Obesity and insulin resistance were induced in C57BL/6J mice by high-fat feeding for 4 wk. Mice then underwent an exercise training program (treadmill running) 5 days/wk (Ex) for 4 wk or remained sedentary (Sed). Compared with Sed, Ex displayed improved (P < 0.01) whole body metabolism as measured via an oral glucose tolerance test. Deleterious lipid species such as diacylglycerol (P < 0.05) and cholesterol esters (P < 0.01) that accumulate with high-fat feeding were decreased in the liver of trained mice. Furthermore, the ratio of phosphatidylcholine (PC) to phosphatidylethanolamine (PE) (the PC/PE ratio), which is associated with membrane integrity and linked to hepatic disease progression, was increased by training (P < 0.05). These findings occurred without corresponding changes in the skeletal muscle lipidome. A concomitant decrease (P < 0.05) was observed for the fatty acid transporters CD36 and FATP4 in the liver, suggesting that exercise stimulates a coordinated reduction in fatty acid entry into hepatocytes. Given the important role of the liver in the regulation of whole body glucose homeostasis, hepatic lipid regression may be a key component by which exercise can improve metabolism.
Collapse
Affiliation(s)
- Andreas B Jordy
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; and Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Kraakman
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; and
| | - Tim Gardner
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; and
| | - Emma Estevez
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; and
| | - Helene L Kammoun
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; and
| | - Jacqui M Weir
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; and
| | - Bente Kiens
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Peter J Meikle
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; and
| | - Mark A Febbraio
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia; and
| | | |
Collapse
|
139
|
Liu J, Wang G, Jia Y, Xu Y. GLP-1 receptor agonists: effects on the progression of non-alcoholic fatty liver disease. Diabetes Metab Res Rev 2015; 31:329-35. [PMID: 25066109 DOI: 10.1002/dmrr.2580] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 06/15/2014] [Accepted: 07/06/2014] [Indexed: 12/21/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common liver diseases, and its incidence has been increasing recently. In addition to hepatic complications, NAFLD is also recognized as an independent risk factor for cardiovascular disease. Unfortunately, the current therapies for NAFLD display variable efficacy; a novel and effective drug is urgently needed. Glucagon-like peptide-1 (GLP-1), a receptor agonist is a new drug approved for treating type 2 diabetes. Recently, these types of agents have shown a novel therapeutic effect on NAFLD. However, the mechanisms of GLP-1 receptor agonists on the treatment of NAFLD have not yet been explained precisely. Recent studies have demonstrated that GLP-1 reverses the progression of NAFLD not only indirectly through an incretin effect that improves key parameters involved in NAFLD, but also a direct effect on lipid metabolism of hepatocytes and inflammation in liver. In this review, we provided an overview of the role and mechanisms of GLP-1 in the therapy of NAFLD.
Collapse
Affiliation(s)
- Jia Liu
- Department of Endocrinology, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, 100020, People's Republic of China
| | | | | | | |
Collapse
|
140
|
Lonardo A, Ballestri S, Targher G, Loria P. Diagnosis and management of cardiovascular risk in nonalcoholic fatty liver disease. Expert Rev Gastroenterol Hepatol 2015; 9:629-650. [PMID: 25327387 DOI: 10.1586/17474124.2015.965143] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is increasingly recognized as an important cardiovascular risk (CVR) factor. This is a narrative clinical review aimed at answering how diagnosis and management of CVR should be conducted in the individual patient with NAFLD. To this end, the authors performed an extensive search of the existing literature on PubMed (1993-2014) using pertinent keywords. To date, CVR among patients with NAFLD might be assessed with the Framingham risk score equation or other risk calculators, to be adapted to the true CVR in the specific population being assessed; however, the use of these CVR calculators needs to be validated by future studies in larger cohorts of NAFLD patients of various ethnic backgrounds in order to substantiate their clinical relevance as a foundation for the primary prevention of cardiovascular diseases in this group of patients. Early and aggressive drug treatment of CVR should be started in NAFLD patients with a history of cardiovascular events, established diabetes or who are at high (calculated) CVR. Whether such an aggressive pharmacological approach is also justified in patients with NAFLD, who are at intermediate or low CVR, remains debatable. Currently, there are no clinical trials showing that the treatment of NAFLD per se (either associated or unassociated with traditional CVR factors) will result in decreased risk of cardiovascular events. Accordingly, drug treatment should be better individualized, aiming at correcting all the coexisting cardio-metabolic risk factors of the individual patient with NAFLD. To this end, an overview of the lifestyle interventions and the available drugs is offered, emphasis being conveyed to statins and metformin, which promise to cover worrying complications of NAFLD such as the risk of developing hepatocellular carcinoma.
Collapse
Affiliation(s)
- Amedeo Lonardo
- Department of Medicine, Division of Internal Medicine, Pavullo Hospital, Pavullo 41026, Italy
| | | | | | | |
Collapse
|
141
|
Liu YS, Huang ZW, Wang L, Liu XX, Wang YM, Zhang Y, Zhang M. Sitagliptin alleviated myocardial remodeling of the left ventricle and improved cardiac diastolic dysfunction in diabetic rats. J Pharmacol Sci 2015; 127:260-74. [PMID: 25837922 DOI: 10.1016/j.jphs.2014.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 12/11/2014] [Accepted: 12/12/2014] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Sitagliptin, a dipeptidyl peptidase IV (DPP-Ⅳ) inhibitor, has a biological role in improving the serum levels of glucagon-like peptide 1 (GLP-1). Hence, we sought to determine the effect of sitagliptin on myocardial inflammation, collagen metabolism, lipid content and myocardial apoptosis in diabetic rats. MATERIALS AND METHODS The type 2 diabetic rat model was induced by low-dose streptozotocin and a high-fat diet. Characteristics of diabetic rats were evaluated by electrocardiography, echocardiography and blood analysis. Cardiac inflammation, fibrosis, cardiomyocyte density, lipid accumulation, and receptor-interacting protein kinase 3 (RIP3) level, related to apoptosis, were detected by histopathologic analysis, RT-PCR and western blot analysis to evaluate the effects of sitagliptin on myocardial remodeling of the left ventricle. RESULTS Diabetic rats showed myocardial hypertrophy or apoptosis, inflammation, lipid accumulation, myocardial fibrosis, elevated collagen content, RIP3 overexpression, and left-ventricular dysfunction. Sitagliptin could reverse the overexpression of RIP3 and alleviate cellular apoptosis in myocardial tissues. It could significantly improve left-ventricular systolic pressure and +dp/dt max, reduce the E/E' ratio, left ventricular end diastolic pressure, -dp/dt max and Tau in diabetic rats. CONCLUSIONS Sitagliptin might have a myocardial protective effect by inhibiting apoptosis, inflammation, lipid accumulation and myocardial fibrosis in diabetic rats, for a potential role in improving left-ventricular function in diabetes.
Collapse
Affiliation(s)
- Yu-Sheng Liu
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China; Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Zhi-Wei Huang
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Lin Wang
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Xin-Xin Liu
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Yong-Mei Wang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Yun Zhang
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| | - Mei Zhang
- Department of Cardiology, The Qilu Hospital of Shandong University, Jinan, Shandong, 250033, PR China.
| |
Collapse
|
142
|
Liu J, Xu Y, Hu Y, Wang G. The role of fibroblast growth factor 21 in the pathogenesis of non-alcoholic fatty liver disease and implications for therapy. Metabolism 2015; 64:380-90. [PMID: 25516477 DOI: 10.1016/j.metabol.2014.11.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/06/2014] [Accepted: 11/25/2014] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) includes a cluster of liver disorders ranging from simple fatty liver to non-alcoholic steatohepatitis (NASH) and cirrhosis. Due to its liver and vascular complications, NAFLD has become a public health problem with high morbidity and mortality. The pathogenesis of NAFLD is considered a "multi-hit hypothesis" that involves lipotoxicity, oxidative stress, endoplasmic reticulum stress, a chronic inflammatory state and mitochondrial dysfunction. Fibroblast growth factor 21 (FGF21) is a member of the fibroblast growth factor family with multiple metabolic functions. FGF21 directly regulates lipid metabolism and reduces hepatic lipid accumulation in an insulin-independent manner. Several studies have shown that FGF21 can ameliorate the "multi-hits" in the pathogenesis of NAFLD. The administration of FGF21 reverses hepatic steatosis, counteracts obesity and alleviates insulin resistance in rodents and nonhuman primates. Using several strategies, we show that the reversal of simple fatty liver and NASH is mediated by activation of the FGF21 signaling pathway. In this review, we describe the molecular mechanisms involved in the onset and/or progression of NAFLD, and review the current literature to highlight the therapeutic procedures associated with the FGF21 signaling pathway for simple fatty liver and NASH, which are the two most important types of NAFLD.
Collapse
Affiliation(s)
- Jia Liu
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China
| | - Yuan Xu
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China
| | - Yanjin Hu
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-yang Hospital, Capital Medical University, No. 8, Gongti South Road, Chaoyang District, Beijing 100020, China.
| |
Collapse
|
143
|
Eguchi Y, Kitajima Y, Hyogo H, Takahashi H, Kojima M, Ono M, Araki N, Tanaka K, Yamaguchi M, Matsuda Y, Ide Y, Otsuka T, Ozaki I, Ono N, Eguchi T, Anzai K. Pilot study of liraglutide effects in non-alcoholic steatohepatitis and non-alcoholic fatty liver disease with glucose intolerance in Japanese patients (LEAN-J). Hepatol Res 2015; 45:269-78. [PMID: 24796231 DOI: 10.1111/hepr.12351] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 04/14/2014] [Accepted: 04/28/2014] [Indexed: 12/14/2022]
Abstract
AIM Non-alcoholic fatty liver disease (NAFLD), a hepatic manifestation of metabolic syndrome, is associated with an increased risk of developing lifestyle-related diseases including type 2 diabetes, cardiovascular disease and cerebral vessel disease. No current drug therapy provides the ideal effects of decreasing hepatic inflammation while simultaneously improving liver fibrosis. Liraglutide is a glucagon-like peptide-1 receptor agonist that affects the histological findings in patients with non-alcoholic steatohepatitis (NASH). This study was conducted to evaluate the effect and action of liraglutide for biopsy-proven NASH. METHODS After lifestyle modification intervention for 24 weeks, subjects whose hemoglobin A1c levels failed to improve to less than 6.0% and/or whose alanine aminotransferase levels were not lower than baseline, received liraglutide at 0.9 mg/body per day for 24 weeks. RESULTS Of 27 subjects, 26 completed the lifestyle modification intervention. Nineteen subjects received liraglutide therapy for 24 weeks. Body mass index, visceral fat accumulation, aminotransferases and glucose abnormalities improved significantly. Repeated liver biopsy was performed in 10 subjects who continued liraglutide therapy for 96 weeks. Six subjects showed decreased histological inflammation as determined by NASH activity score and stage determined by Brunt classification. We saw no significant adverse events during therapy with liraglutide. CONCLUSION Our pilot study demonstrated that treatment with liraglutide had a good safety profile and significantly improved liver function and histological features in NASH patients with glucose intolerance.
Collapse
|
144
|
McCarty MF, DiNicolantonio JJ. Acarbose, lente carbohydrate, and prebiotics promote metabolic health and longevity by stimulating intestinal production of GLP-1. Open Heart 2015; 2:e000205. [PMID: 25685364 PMCID: PMC4316590 DOI: 10.1136/openhrt-2014-000205] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/01/2014] [Accepted: 01/12/2015] [Indexed: 12/25/2022] Open
Abstract
The α-glucosidase inhibitor acarbose, which slows carbohydrate digestion and blunts postprandial rises in plasma glucose, has long been used to treat patients with type 2 diabetes or glucose intolerance. Like metformin, acarbose tends to aid weight control, postpone onset of diabetes and decrease risk for cardiovascular events. Acarbose treatment can favourably affect blood pressure, serum lipids, platelet aggregation, progression of carotid intima-media thickness and postprandial endothelial dysfunction. In mice, lifetime acarbose feeding can increase median and maximal lifespan-an effect associated with increased plasma levels of fibroblast growth factor 21 (FGF21) and decreased levels of insulin-like growth factor-I (IGF-I). There is growing reason to suspect that an upregulation of fasting and postprandial production of glucagon-like peptide-1 (GLP-1)-stemming from increased delivery of carbohydrate to L cells in the distal intestinal tract-is largely responsible for the versatile health protection conferred by acarbose. Indeed, GLP-1 exerts protective effects on vascular endothelium, the liver, the heart, pancreatic β cells, and the brain which can rationalise many of the benefits reported with acarbose. And GLP-1 may act on the liver to modulate its production of FGF21 and IGF-I, thereby promoting longevity. The benefits of acarbose are likely mimicked by diets featuring slowly-digested 'lente' carbohydrate, and by certain nutraceuticals which can slow carbohydrate absorption. Prebiotics that promote colonic generation of short-chain fatty acids represent an alternative strategy for boosting intestinal GLP-1 production. The health benefits of all these measures presumably would be potentiated by concurrent use of dipeptidyl peptidase 4 inhibitors, which slow the proteolysis of GLP-1 in the blood.
Collapse
Affiliation(s)
| | - James J DiNicolantonio
- Mid America Heart Institute, St. Luke's Hospital , Kansas City, Missouri , USA ; Wegmans Pharmacy , Ithaca, New York , USA
| |
Collapse
|
145
|
Jensterle M, Kocjan T, Kravos NA, Pfeifer M, Janez A. Short-term intervention with liraglutide improved eating behavior in obese women with polycystic ovary syndrome. Endocr Res 2015; 40:133-8. [PMID: 25330463 DOI: 10.3109/07435800.2014.966385] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIM Glucagon-like peptide 1 receptor agonists (GLP-1 RA) stimulate satiety leading to reductions in food intake and body weight. The effects of long- acting GLP-1 RA liraglutide on eating behavior of women with PCOS have not been investigated yet. METHODS Thirty-six obese women with PCOS (mean ± SD, aged 31.2 ± 7.8 years, with BMI 38.7 ± 0.1 kg/m(2)), pretreated with metformin (1000 mg BID) were switched to liraglutide 1.2 mg QD sc for 12 weeks. Adiposity parameters and eating behavior as assessed by Three-Factor Eating Questionnaire (TFEQ-R18) were examined at baseline and after 12 weeks. RESULTS Subjects treated with liraglutide lost on average 3.8 ± 0.1 kg (p < 0.001). Significant reductions of waist circumference and visceral adipose tissue (VAT) mass, volume and area were demonstrated from liraglutide induction to the end of the study. TFEQ-R18 scores were significantly different pre- and post-liraglutide intervention. After treatment with liraglutide the uncontrolled eating (UE) score decreased from 36.8 ± 24.5 to 19.6 ± 18.4 (p < 0.001) and emotional eating (EE) score decreased from 49.9 ± 33.3 to 28.5 ± 26.9 (p < 0.001). Scores for cognitive restraint (CR) were not changed. CONCLUSIONS Short-term liraglutide treatment was associated with weight loss and significantly improved eating behavior in obese women with PCOS.
Collapse
Affiliation(s)
- Mojca Jensterle
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana , Ljubljana , Slovenia
| | | | | | | | | |
Collapse
|
146
|
Junker AE, Gluud LL, Pedersen J, Langhoff JL, Holst JJ, Knop FK, Vilsbøll T. A 25-year-old woman with type 2 diabetes and liver disease. Case Rep Gastroenterol 2014; 8:398-403. [PMID: 25606030 PMCID: PMC4296242 DOI: 10.1159/000369968] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A 25-year-old female nurse was referred to our diabetes outpatient clinic with poorly controlled type 2 diabetes, obesity and elevated liver function tests (LFTs). Following a liver biopsy she was diagnosed with non-alcoholic steatohepatitis (NASH) and liver fibrosis. Treatment with subcutaneous injections of the glucagon-like peptide-1 receptor (GLP-1R) agonist liraglutide was initiated. After 46 weeks of treatment the patient had lost 16 kg, glycemic control was excellent and LFTs had normalized. Repeat liver biopsy and ultrasound showed reduction in hepatic fat content and inflammatory cells. The biopsy no longer fulfilled the criteria for NASH. The liver biopsies did not express hepatic GLP-1Rs using quantitative polymerase chain reaction. Our case suggests that liraglutide may benefit patients with NASH.
Collapse
Affiliation(s)
- Anders Ellekær Junker
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Copenhagen, Denmark ; NNF Center for Basic Metabolic Research and Department of Biomedical Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Lotte Gluud
- Department of Gastroenterology, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Jens Pedersen
- NNF Center for Basic Metabolic Research and Department of Biomedical Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jill Levin Langhoff
- Department of Pathology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip Krag Knop
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Copenhagen, Denmark
| |
Collapse
|
147
|
Trovato FM, Catalano D, Musumeci G, Trovato GM. 4Ps medicine of the fatty liver: the research model of predictive, preventive, personalized and participatory medicine-recommendations for facing obesity, fatty liver and fibrosis epidemics. EPMA J 2014; 5:21. [PMID: 25937854 PMCID: PMC4417534 DOI: 10.1186/1878-5085-5-21] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 11/24/2014] [Indexed: 12/13/2022]
Abstract
Relationship between adipose tissue and fatty liver, and its possible evolution in fibrosis, is supported by clinical and research experience. Given the multifactorial pathogenesis of non-alcoholic fatty liver disease (NAFLD), treatments for various contributory risk factors have been proposed; however, there is no single validated therapy or drug association recommended for all cases which can stand alone. Mechanisms, diagnostics, prevention and treatment of obesity, fatty liver and insulin resistance are displayed along with recommendations and position points. Evidences and practice can get sustainable and cost-benefit valuable outcomes by participatory interventions. These recommendations can be enhanced by comprehensive research projects, addressed to societal issues and innovation, market appeal and industry development, cultural acceptance and sustainability. The basis of participatory medicine is a greater widespread awareness of a condition which is both a disease and an easy documented and inclusive clue for associated diseases and unhealthy lifestyle. This model is suitable for addressing prevention and useful for monitoring improvement, worsening and adherence with non-invasive imaging tools which allow targeted approaches. The latter include health psychology and nutritional and physical exercise prescription expertise disseminated by continuous medical education but, more important, by concrete curricula for training undergraduate and postgraduate students. It is possible and recommended to do it by early formal teaching of ultrasound imaging procedures and of practical lifestyle intervention strategies, including approaches aimed to healthier fashion suggestions. Guidelines and requirements of research project funding calls should be addressed also to NAFLD and allied conditions and should encompass the goal of training by research and the inclusion of participatory medicine topics. A deeper awareness of ethics of competences in health professionals and the articulation of knowledge, expertise and skills of medical doctors, dieticians, health psychologists and sport and physical exercise graduates are the necessary strategy for detectin a suboptimal health status and achieving realistically beneficial lifestyle changes. “The devil has put a penalty on all things we enjoy in life. Either we suffer in health or we suffer in soul or we get fat” (Albert Einstein); the task of medical research and intervention is to make possible to enjoy life also without things that make sufferance in health and souls and which excessively increase body fat.
Collapse
Affiliation(s)
- Francesca Maria Trovato
- Department of Clinical and Experimental Medicine, Internal Medicine Division, School of Medicine, University of Catania, Via S. Sofia, 78-95123 Catania, Italy
| | - Daniela Catalano
- Department of Clinical and Experimental Medicine, Internal Medicine Division, School of Medicine, University of Catania, Via S. Sofia, 78-95123 Catania, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Via S. Sofia, 87-95123 Catania, Italy
| | - Guglielmo M Trovato
- Department of Clinical and Experimental Medicine, Internal Medicine Division, School of Medicine, University of Catania, Via S. Sofia, 78-95123 Catania, Italy
| |
Collapse
|
148
|
Ectopic lipid storage in non-alcoholic fatty liver disease is not mediated by impaired mitochondrial oxidative capacity in skeletal muscle. Clin Sci (Lond) 2014; 127:655-63. [PMID: 24738611 DOI: 10.1042/cs20130404] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD), characterized by lipid deposition within the liver [intrahepatocellular lipid (IHCL)], is associated with insulin resistance and the metabolic syndrome (MS). It has been suggested that impaired skeletal muscle mitochondrial function may contribute to ectopic lipid deposition, and the associated MS, by altering post-prandial energy storage. To test this hypothesis, we performed a cross-sectional study of 17 patients with NAFLD [mean±S.D.; age, 45±11 years; body mass index (BMI), 31.6±3.4 kg/m2] and 18 age- and BMI-matched healthy controls (age, 44±11 years; BMI, 30.5±5.2 kg/m2). We determined body composition by MRI, IHCL and intramyocellular (soleus and tibialis anterior) lipids (IMCLs) by proton magnetic resonance spectroscopy (1H-MRS) and skeletal muscle mitochondrial function by dynamic phosphorus magnetic resonance spectroscopy (31P-MRS) of quadriceps muscle. Although matched for BMI and total adiposity, after statistical adjustment for gender, patients with NAFLD (defined by IHCL ≥ 5.5%) had higher IHCLs (25±16% compared with 2±2%; P<0.0005) and a higher prevalence of the MS (76% compared with 28%) compared with healthy controls. Despite this, the visceral fat/subcutaneous fat ratio, IMCLs and muscle mitochondrial function were similar between the NAFLD and control groups, with no significant difference in the rate constants of post-exercise phosphocreatine (PCr) recovery (1.55±0.4 compared with 1.51±0.4 min-1), a measure of muscle mitochondrial function. In conclusion, impaired muscle mitochondrial function does not seem to underlie ectopic lipid deposition, or the accompanying features of the MS, in patients with NAFLD.
Collapse
|
149
|
Xu F, Li Z, Zheng X, Liu H, Liang H, Xu H, Chen Z, Zeng K, Weng J. SIRT1 mediates the effect of GLP-1 receptor agonist exenatide on ameliorating hepatic steatosis. Diabetes 2014; 63:3637-46. [PMID: 24947350 DOI: 10.2337/db14-0263] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
GLP-1 and incretin mimetics, such as exenatide, have been shown to attenuate hepatocyte steatosis in vivo and in vitro, but the specific underlying mechanism is unclear. SIRT1, an NAD(+)-dependent protein deacetylase, has been considered as a crucial regulator in hepatic lipid homeostasis by accumulated studies. Here, we speculate that SIRT1 might mediate the effect of the GLP-1 receptor agonist exenatide (exendin-4) on ameliorating hepatic steatosis. After 8 weeks of exenatide treatment in male SIRT1(+/-) mice challenged with a high-fat diet and their wild-type (WT) littermates, we found that lipid deposition and inflammation in the liver, which were improved dramatically in the WT group, diminished in SIRT1(+/-) mice. In addition, the protein expression of SIRT1 and phosphorylated AMPK was upregulated, whereas lipogenic-related protein, including SREBP-1c and PNPLA3, was downregulated in the WT group after exenatide treatment. However, none of these changes were observed in SIRT1(+/-) mice. In HepG2 cells, exendin-4-reversed lipid deposition induced by palmitate was hampered when SIRT1 was silenced by SIRT1 RNA interference. Our data demonstrate that SIRT1 mediates the effect of exenatide on ameliorating hepatic steatosis, suggesting the GLP-1 receptor agonist could serve as a potential drug for nonalcoholic fatty liver disease (NAFLD), especially in type 2 diabetes combined with NAFLD, and SIRT1 could be a therapeutic target of NAFLD.
Collapse
Affiliation(s)
- Fen Xu
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Zhuo Li
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Xiaobin Zheng
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Hongxia Liu
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Hua Liang
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Haixia Xu
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Zonglan Chen
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Kejing Zeng
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Jianping Weng
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| |
Collapse
|
150
|
Wang XC, Gusdon AM, Liu H, Qu S. Effects of glucagon-like peptide-1 receptor agonists on non-alcoholic fatty liver disease and inflammation. World J Gastroenterol 2014; 20:14821-14830. [PMID: 25356042 PMCID: PMC4209545 DOI: 10.3748/wjg.v20.i40.14821] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/14/2014] [Accepted: 07/16/2014] [Indexed: 02/06/2023] Open
Abstract
Glucagon-like peptide1 (GLP-1) is secreted from Langerhans cells in response to oral nutrient intake. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are a new class of incretin-based anti-diabetic drugs. They function to stimulate insulin secretion while suppressing glucagon secretion. GLP-1-based therapies are now well established in the management of type 2 diabetes mellitus (T2DM), and recent literature has suggested potential applications of these drugs in the treatment of obesity and for protection against cardiovascular and neurological diseases. As we know, along with change in lifestyles, the prevalence of non-alcoholic fatty liver disease (NAFLD) in China is rising more than that of viral hepatitis and alcoholic fatty liver disease, and NAFLD has become the most common chronic liver disease in recent years. Recent studies further suggest that GLP-1RAs can reduce transaminase levels to improve NAFLD by improving blood lipid levels, cutting down the fat content to promote fat redistribution, directly decreasing fatty degeneration of the liver, reducing the degree of liver fibrosis and improving inflammation. This review shows the NAFLD-associated effects of GLP-1RAs in animal models and in patients with T2DM or obesity who are participants in clinical trials.
Collapse
|