1
|
Chege BM, Mwangi PW, Githinji CG, Bukachi F. Dietary regimens appear to possess significant effects on the development of combined antiretroviral therapy (cART)-associated metabolic syndrome. PLoS One 2024; 19:e0298752. [PMID: 38416754 PMCID: PMC10901320 DOI: 10.1371/journal.pone.0298752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/29/2024] [Indexed: 03/01/2024] Open
Abstract
INTRODUCTION This study investigated the interactions between a low protein high calorie (LPHC) diet and an integrase inhibitor-containing antiretroviral drug regimen (INI-CR)in light of evidence suggesting that the initiation of cART in patients with poor nutritional status is a predictor of mortality independent of immune status. METHODS Freshly weaned Sprague Dawley rats (120) were randomized into the standard, LPHC and normal protein high calorie (NPHC) diet groups (n = 40/group) initially for 15 weeks. Thereafter, experimental animals in each diet group were further randomized into four treatment sub-groups (n = 10/group) Control (normal saline), group 1(TDF+3TC+DTG and Tesamorelin), group 2 (TDF+3TC+DTG), and Positive control (AZT+3TC+ATV/r) with treatment and diets combined for 9 weeks. Weekly body weights, fasting blood glucose (FBG), oral glucose tolerance test (OGTT); lipid profiles, liver weights, hepatic triglycerides and adiposity were assessed at week 24. RESULTS At week 15, body weights increased between the diet group in phase 1(standard 146 ± 1.64 vs. 273.1 ± 1.56 g), (NPHC, 143.5 ± 2.40 vs. 390.2 ± 4.94 g) and (LPHC, 145.5 ± 2.28 g vs. 398.3 ± 4.89 g) (p< 0.0001). A similar increase was noted in the FBG and OGTT (p< 0.0001). In phase 2, there was an increase in FBG, OGTT, body weights, lipid profile, liver weights, hepatic triglycerides, adiposity and insulin levels in group 2 and positive control in both NPHC and LPHC diet groups (p<0.0001). Growth hormone levels were decreased in Tesamorelin-free group 2 and positive control in both NPHC and LPHC (p< 0.0001). CONCLUSIONS The obesogenic activities of the LPHC diet exceeded that of the NPHC diet and interacted with both integrase-containing and classical cART drug regimens to reproduce cART associated metabolic dysregulation. The effects were however reversed by co-administration with tesamorelin, a synthetic growth hormone releasing hormone analogue.
Collapse
Affiliation(s)
- Boniface M Chege
- School of Health Sciences, Dedan Kimathi University of Technology, Nyeri, Kenya
- Department of Human Anatomy and Medical Physiology, University of Nairobi, Nairobi, Kenya
| | - Peter W Mwangi
- Department of Human Anatomy and Medical Physiology, University of Nairobi, Nairobi, Kenya
| | - Charles G Githinji
- Department of Human Anatomy and Medical Physiology, University of Nairobi, Nairobi, Kenya
| | - Frederick Bukachi
- Department of Human Anatomy and Medical Physiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
2
|
Swain J, Jadhao P, Sravya SL, Teli B, Lavanya K, Singh J, Sahoo A, Das S. Mitochondrial Dysfunction and Imeglimin: A New Ray of Hope for the Treatment of Type-2 Diabetes Mellitus. Mini Rev Med Chem 2024; 24:1575-1589. [PMID: 37861052 DOI: 10.2174/0113895575260225230921062013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/03/2023] [Accepted: 07/26/2023] [Indexed: 10/21/2023]
Abstract
Diabetes is a rapidly growing health challenge and epidemic in many developing countries, including India. India, being the diabetes capital of the world, has the dubious dual distinction of being the leading nations for both undernutrition and overnutrition. Diabetes prevalence has increased in both rural and urban areas, affected the younger population and increased the risk of complications and economic burden. These alarming statistics ring an alarm bell to achieve glycemic targets in the affected population in order to decrease diabetes-related morbidity and mortality. In the recent years, diabetes pathophysiology has been extended from an ominous triad through octet and dirty dozen etc. There is a new scope to target multiple pathways at the molecular level to achieve a better glycemic target and further prevent micro- and macrovascular complications. Mitochondrial dysfunction has a pivotal role in both β-cell failure and insulin resistance. Hence, targeting this molecular pathway may help with both insulin secretion and peripheral tissue sensitization to insulin. Imeglimin is the latest addition to our anti-diabetic armamentarium. As imeglimin targets, this root cause of defective energy metabolism and insulin resistance makes it a new add-on therapy in different diabetic regimes to achieve the proper glycemic targets. Its good tolerability and efficacy profiles in recent studies shows a new ray of hope in the journey to curtail diabetes-related morbidity.
Collapse
Affiliation(s)
- Jayshree Swain
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Pooja Jadhao
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - S L Sravya
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Brij Teli
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Kasukurti Lavanya
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Jaspreet Singh
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Abhay Sahoo
- Department of Endocrinology, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, India
| | - Srijit Das
- Department of Human & Clinical Anatomy, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat 123, Sultanate of Oman
| |
Collapse
|
3
|
Garg R, Agarwal A, Katekar R, Goand UK, Singh N, Yadav S, Rathaur S, Verma S, Maity D, Vishwakarma S, Gayen JR. Pancreastatin inhibitor PSTi8 ameliorates insulin resistance by decreasing fat accumulation and oxidative stress in high-fat diet-fed mice. Amino Acids 2023; 55:1587-1600. [PMID: 37716928 DOI: 10.1007/s00726-023-03332-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 09/01/2023] [Indexed: 09/18/2023]
Abstract
Abnormal fat accumulation, enhanced free fatty acids (FFA) release, and their metabolites cause insulin resistance (IR) in major glucose-lipid metabolic organs such as skeletal muscle and adipose tissue. However, excessive lipolysis and FFA release from adipose tissue elevate plasma FFA levels leading to oxidative stress and skeletal muscle IR. Indeed, in obese individuals, there is enhanced pro-inflammatory secretion from adipose tissue influencing insulin signaling in skeletal muscles. Here, we investigated the effect of PSTi8 on FFA-induced IR in both in vitro and in vivo models. Palmitate (Pal)-treated 3T3-L1 cells increased lipid accumulation as well as lipolysis, which reduced the insulin-stimulated glucose uptake. PSTi8 treatment significantly prevented Pal-induced lipid accumulation, and release and enhanced insulin-stimulated glucose uptake. It further reduced the release of pro-inflammatory cytokines from Pal-treated 3T3-L1 cells as well as from adipose tissue explants. In addition, PSTi8 treatment decreases M1 surface markers in Pal-treated bone marrow-derived monocytes (BMDM). PSTi8 treatment also significantly enhanced the Pal-mediated reduced skeletal muscle glucose disposal and reduced intracellular oxidative stress. In vitro effect of PSTi8 was consistent with in vivo HFD-fed mice IR model. PSTi8 treatment in HFD-fed mice significantly improved glucose metabolism and enhanced skeletal muscle insulin sensitivity with reduced adiposity and pro-inflammatory cytokines. Taken together, our results support that PSTi8 treatment can protect both adipose and skeletal muscles from FFA-induced IR.
Collapse
Affiliation(s)
- Richa Garg
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Arun Agarwal
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Roshan Katekar
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Umesh Kumar Goand
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Naveen Singh
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
| | - Shubhi Yadav
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shivam Rathaur
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
| | - Saurabh Verma
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Debalina Maity
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
| | - Sachin Vishwakarma
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India
| | - Jiaur R Gayen
- Pharmaceutics and Pharmacokinetics, Pharmacology Division, CSIR-Central Drug Research Institute, Jankipuram Extension, Lucknow, 226031, India.
- Pharmacology Division, CSIR-Central Drug Research Institute, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
4
|
Schön M, Zaharia OP, Strassburger K, Kupriyanova Y, Bódis K, Heilmann G, Strom A, Bönhof GJ, Michelotti F, Yurchenko I, Möser C, Huttasch M, Bombrich M, Kelm M, Burkart V, Schrauwen-Hinderling VB, Wagner R, Roden M. Intramyocellular Triglyceride Content During the Early Course of Type 1 and Type 2 Diabetes. Diabetes 2023; 72:1483-1492. [PMID: 37478166 PMCID: PMC10545555 DOI: 10.2337/db23-0353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/10/2023] [Indexed: 07/23/2023]
Abstract
Intramyocellular lipid content (IMCL) is elevated in insulin-resistant humans, but it changes over time, and relationships with comorbidities remain unclear. We examined IMCL during the initial course of diabetes and its associations with complications. Participants of the German Diabetes Study (GDS) with recent-onset type 1 (n = 132) or type 2 diabetes (n = 139) and glucose-tolerant control subjects (n = 128) underwent 1H-MRS to measure IMCL and muscle volume, whole-body insulin sensitivity (hyperinsulinemic-euglycemic clamps; M-value), and cycling spiroergometry (VO2max). Subgroups underwent the same measurements after 5 years. At baseline, IMCL was ∼30% higher in type 2 diabetes than in other groups independently of age, sex, BMI, and muscle volume. In type 2 diabetes, the M-value was ∼36% and ∼62% lower compared with type 1 diabetes and control subjects, respectively. After 5 years, the M-value decreased by ∼29% in type 1 and ∼13% in type 2 diabetes, whereas IMCL remained unchanged. The correlation between IMCL and M-value in type 2 diabetes at baseline was modulated by VO2max. IMCL also associated with microalbuminuria, the Framingham risk score for cardiovascular disease, and cardiac autonomic neuropathy. Changes in IMCL within 5 years after diagnosis do not mirror the progression of insulin resistance in type 2 diabetes but associate with early diabetes-related complications. ARTICLE HIGHLIGHTS Intramyocellular lipid content (IMCL) can be elevated in insulin-resistant humans, but its dynamics and association with comorbidities remain unclear. Independently of age, sex, body mass, and skeletal muscle volume, IMCL is higher in recent-onset type 2, but not type 1 diabetes, and remains unchanged within 5 years, despite worsening insulin resistance. A degree of physical fitness modulates the association between IMCL and insulin sensitivity in type 2 diabetes. Whereas higher IMCL associates with lower insulin sensitivity in people with lower physical fitness, there is no association between IMCL and insulin sensitivity in those with higher degree of physical fitness. IMCL associates with progression of microalbuminuria, cardiovascular disease risk, and cardiac autonomic neuropathy.
Collapse
Affiliation(s)
- Martin Schön
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Oana P. Zaharia
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Yuliya Kupriyanova
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Kálmán Bódis
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Geronimo Heilmann
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Gidon J. Bönhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Filippo Michelotti
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Iryna Yurchenko
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Clara Möser
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Maximilian Huttasch
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Maria Bombrich
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Malte Kelm
- Department of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Volker Burkart
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Vera B. Schrauwen-Hinderling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Robert Wagner
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
5
|
Yan T, Wang M, Yang S, Wang Y, Zhou X, Zhu X, Ma W, Tang S, Li J. Prevalence of insulin resistance in Chinese solar greenhouse and field workers: evidence from a solar greenhouse and field workers study. Front Public Health 2023; 11:1257183. [PMID: 37693717 PMCID: PMC10485250 DOI: 10.3389/fpubh.2023.1257183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Evidence suggests that agricultural workers are at higher risk of insulin resistance (IR), but few studies have investigated IR in solar greenhouse workers, who are exposed to higher concentrations of agricultural risk factors than traditional agricultural workers. A prevalence study was conducted in a greenhouse vegetable farm in China. In total, 948 participants were enrolled in this study. Among them, 721 participants were allocated to the greenhouse worker group (G group), and 227 participants were assigned to the field worker group (F group). The TyG index, which is an indicator to evaluate prediabetes (IR), was calculated by the formula: TyG index = ln [fasting triglycerides (mg/dL) × fasting plasma glucose (mg/dL)/2]. To evaluate the associations of TyG index alternation with solar greenhouse and field work, multiple linear regression (MLR) and logistic regression models were performed. The TyG index in the G group (8.53 ± 0.56) was higher than that in the F group (8.44 ± 0.59) (p < 0.05). Solar greenhouse work was positively associated with an increased TyG index in both the multiple linear regression model [β = 0.207, (0.006, 0.408)] and the logistic regression model [OR = 1.469, (1.070, 2.016)]. IR was associated with the solar greenhouse work. However, the determination of agricultural hazard factors needs to be further strengthened to improve exposure assessment. Graphical Abstract.
Collapse
Affiliation(s)
- Tenglong Yan
- Beijing Institute of Occupational Disease Prevention and Treatment, Beijing, China
| | - Minghui Wang
- Department of Neurology, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Siwen Yang
- National Center for Occupational Safety and Health, National Health Commission of the People’s Republic of China, Beijing, China
| | - Yuqian Wang
- Beijing Key Laboratory of Occupational Safety and Health, Institute of Urban Safety and Environmental Science, Beijing Academy of Science and Technology, Beijing, China
| | - Xingfan Zhou
- Beijing Key Laboratory of Occupational Safety and Health, Institute of Urban Safety and Environmental Science, Beijing Academy of Science and Technology, Beijing, China
| | - Xiaojun Zhu
- National Center for Occupational Safety and Health, National Health Commission of the People’s Republic of China, Beijing, China
| | - Wenjun Ma
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Shichuan Tang
- Beijing Key Laboratory of Occupational Safety and Health, Institute of Urban Safety and Environmental Science, Beijing Academy of Science and Technology, Beijing, China
| | - Jue Li
- Beijing Institute of Occupational Disease Prevention and Treatment, Beijing, China
| |
Collapse
|
6
|
Moreira MLM, de Araújo IM, Fukada SY, Venturini LGR, Guidorizzi NR, Garrido CE, Rosen CJ, de Paula FJA. Refining Evaluation of Bone Mass and Adipose Distribution in Dunnigan Syndrome. Int J Mol Sci 2023; 24:13118. [PMID: 37685926 PMCID: PMC10488191 DOI: 10.3390/ijms241713118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Familial partial lipodystrophies (FPLD) are rare diseases characterized by selective loss of subcutaneous adipose tissue at different sites. This cross-sectional observational study aimed to estimate adipose tissue in the bone marrow (BMAT), intra (IMCL) and extra-myocyte lipids (EMCL), and define the bone phenotype in the context of FPLD2/Dunnigan syndrome (DS). The subjects comprised 23 controls (C) and 18 DS patients, matched by age, weight and height. Blood samples, dual-energy X-ray absorptiometry for bone mineral density (BMD) and trabecular bone score (TBS) and 1H-spectroscopy using magnetic resonance to estimate BMAT in the lumbar spine, IMCL, EMCL and osteoclastogenesis were assessed. The prevalence of diabetes mellitus was 78% in DS patients. Glucose, HbA1c, triglycerides, insulin and HOMA-IR levels were elevated in DS, whereas HDLc, 25(OH)D, PTH and osteocalcin levels were reduced. BMD was similar between groups at all sites, except 1/3 radius, which was lower in DS group. TBS was reduced in DS. DS presented increased osteoclastogenesis and elevated BMAT, with greater saturation levels and higher IMCL than the C group. HOMA-IR and EMCL were negatively associated with TBS; osteocalcin and EMCL were correlated negatively with BMD. This study contributes to refining the estimation of adipose tissue in DS by showing increased adiposity in the lumbar spine and muscle tissue. DXA detected lower TBS and BMD in the 1/3 radius, suggesting impairment in bone quality and that bone mass is mainly affected in the cortical bone.
Collapse
Affiliation(s)
- Mariana Lima Mascarenhas Moreira
- Department of Internal Medicine, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, SP, Brazil; (M.L.M.M.); (I.M.d.A.); (N.R.G.)
| | - Iana Mizumukai de Araújo
- Department of Internal Medicine, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, SP, Brazil; (M.L.M.M.); (I.M.d.A.); (N.R.G.)
| | - Sandra Yasuyo Fukada
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto 14040-900, SP, Brazil; (S.Y.F.); (L.G.R.V.)
| | - Lucas Gabriel R. Venturini
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto 14040-900, SP, Brazil; (S.Y.F.); (L.G.R.V.)
| | - Natalia Rossin Guidorizzi
- Department of Internal Medicine, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, SP, Brazil; (M.L.M.M.); (I.M.d.A.); (N.R.G.)
| | - Carlos Ernesto Garrido
- Department of Physics, Faculty of Philosophy, Sciences and Letters of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto 14049-900, SP, Brazil;
| | | | - Francisco José Albuquerque de Paula
- Department of Internal Medicine, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirao Preto 14049-900, SP, Brazil; (M.L.M.M.); (I.M.d.A.); (N.R.G.)
| |
Collapse
|
7
|
Aneis YM, El Refaye GE, Taha MM, Aldhahi MI, Elsisi HF. Concurrent Aerobic and Strength Training with Caloric Restriction Reduces Insulin Resistance in Obese Premenopausal Women: A Randomized Controlled Trial. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1193. [PMID: 37512005 PMCID: PMC10384259 DOI: 10.3390/medicina59071193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023]
Abstract
Background and Objectives: Obese premenopausal women are at high risk of developing insulin resistance (IR). Concurrent aerobic and strength training (CAST) has been shown to provide remarkable advantages, yet its effects, along with caloric restriction in such a high-risk population, are not yet established. This study aimed to investigate the impact of concurrent aerobic and strength training with caloric restriction (CAST-CR) on IR in obese premenopausal women. Materials and Methods: Forty-two obese premenopausal women with reported IR, aged 40-50 years, were randomly allocated to either the (CAST-CR) intervention group, who underwent CAST with caloric restriction, or the (AT-CR) control group, who received aerobic training in addition to caloric restriction. Both groups completed 12 weeks of controlled training with equivalent training time. Aerobic training began at 60% and gradually progressed to achieve 75% of the maximum heart rate, while strength training was executed at 50% to 70% of the one-repetition maximum (1RM). Anthropometric measures, abdominal adiposity, metabolic parameters, and homeostasis model assessment-estimated insulin resistance (HOMA-IR) were evaluated prior to and following the intervention. Results: Both groups experienced a substantial enhancement in the selected parameters compared to the baseline (p < 0.001), with higher improvement within the CAST-CR group. The changes in HOMA-IR were -1.24 (95%CI, -1.37 to -1.12) in the CAST-CR group vs. -1.07 (95%CI, -1.19 to -0.94) in the AT-CR group. Conclusions: While AT-CR improved insulin sensitivity in premenopausal women who were obese and hyperinsulinemic, CAST with calorie restriction improved insulin sensitivity more significantly, suggesting it as a preferable alternative.
Collapse
Affiliation(s)
- Yasser M Aneis
- Department of Basic Sciences, Faculty of Physical Therapy, Cairo University, Giza 11432, Egypt
- Department of Basic Sciences, Faculty of Physical Therapy, Delta University for Science and Technology, Gamasa 11152, Egypt
| | - Ghada E El Refaye
- Department of Physical Therapy for Women's Health, Faculty of Physical Therapy, Cairo University, Giza 11432, Egypt
- Department of Physical Therapy for Women's Health, Faculty of Physical Therapy, Pharos University, Alexandria 21311, Egypt
| | - Mona Mohamed Taha
- Department of Rehabilitation Sciences, College of Health and Rehabilitation Sciences, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Monira I Aldhahi
- Department of Rehabilitation Sciences, College of Health and Rehabilitation Sciences, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Hany F Elsisi
- Department of Physical Therapy for Cardiovascular/Respiratory Disorders and Geriatrics, Faculty of Physical Therapy, Cairo University, Giza 11432, Egypt
- Department of Respiratory Therapy, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Epidemiological and mechanistic studies have reported relationships between blood lipids, mostly measured by traditional method in clinical settings, and gestational diabetes mellitus (GDM). Recent advances of high-throughput lipidomics techniques have made available more comprehensive lipid profiling in biological samples. This review aims to summarize evidence from prospective studies in assessing relations between blood lipids and GDM, and discuss potential underlying mechanisms. RECENT FINDINGS Mass spectrometry and nuclear magnetic resonance spectroscopy-based analytical platforms are extensively used in lipidomics research. Epidemiological studies have identified multiple novel lipidomic biomarkers that are associated with risk of GDM, such as certain types of fatty acids, glycerolipids, glycerophospholipids, sphingolipids, cholesterol, and lipoproteins. However, the findings are inconclusive mainly due to the heterogeneities in study populations, sample sizes, and analytical platforms. Mechanistic evidence indicates that abnormal lipid metabolism may be involved in the pathogenesis of GDM by impairing pancreatic β-cells and inducing insulin resistance through several etiologic pathways, such as inflammation and oxidative stress. SUMMARY Lipidomics is a powerful tool to study pathogenesis and biomarkers for GDM. Lipidomic biomarkers and pathways could help to identify women at high risk for GDM and could be potential targets for early prevention and intervention of GDM.
Collapse
Affiliation(s)
- Yi Wang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiong-Fei Pan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University
- Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu, China
| | - An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| |
Collapse
|
9
|
Abstract
Growth hormone (GH) and insulin-like growth factor 1 (IGF-1) are essential to normal growth, metabolism, and body composition, but in acromegaly, excesses of these hormones strikingly alter them. In recent years, the use of modern methodologies to assess body composition in patients with acromegaly has revealed novel aspects of the acromegaly phenotype. In particular, acromegaly presents a unique pattern of body composition changes in the setting of insulin resistance that we propose herein to be considered an acromegaly-specific lipodystrophy. The lipodystrophy, initiated by a distinctive GH-driven adipose tissue dysregulation, features insulin resistance in the setting of reduced visceral adipose tissue (VAT) mass and intra-hepatic lipid (IHL) but with lipid redistribution, resulting in ectopic lipid deposition in muscle. With recovery of the lipodystrophy, adipose tissue mass, especially that of VAT and IHL, rises, but insulin resistance is lessened. Abnormalities of adipose tissue adipokines may play a role in the disordered adipose tissue metabolism and insulin resistance of the lipodystrophy. The orexigenic hormone ghrelin and peptide Agouti-related peptide may also be affected by active acromegaly as well as variably by acromegaly therapies, which may contribute to the lipodystrophy. Understanding the pathophysiology of the lipodystrophy and how acromegaly therapies differentially reverse its features may be important to optimizing the long-term outcome for patients with this disease. This perspective describes evidence in support of this acromegaly lipodystrophy model and its relevance to acromegaly pathophysiology and the treatment of patients with acromegaly.
Collapse
Affiliation(s)
- Pamela U. Freda
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, United States
| |
Collapse
|
10
|
Adamowski M, Wołodko K, Oliveira J, Castillo-Fernandez J, Murta D, Kelsey G, Galvão AM. Leptin Signaling in the Ovary of Diet-Induced Obese Mice Regulates Activation of NOD-Like Receptor Protein 3 Inflammasome. Front Cell Dev Biol 2021; 9:738731. [PMID: 34805147 PMCID: PMC8595835 DOI: 10.3389/fcell.2021.738731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/04/2021] [Indexed: 12/21/2022] Open
Abstract
Obesity leads to ovarian dysfunction and the establishment of local leptin resistance. The aim of our study was to characterize the levels of NOD-like receptor protein 3 (NLRP3) inflammasome activation in ovaries and liver of mice during obesity progression. Furthermore, we tested the putative role of leptin on NLRP3 regulation in those organs. C57BL/6J female mice were treated with equine chorionic gonadotropin (eCG) or human chorionic gonadotropin (hCG) for estrous cycle synchronization and ovary collection. In diet-induced obesity (DIO) protocol, mice were fed chow diet (CD) or high-fat diet (HFD) for 4 or 16 weeks, whereas in the hyperleptinemic model (LEPT), mice were injected with leptin for 16 days (16 L) or saline (16 C). Finally, the genetic obese leptin-deficient ob/ob (+/? and −/−) mice were fed CD for 4 week. Either ovaries and liver were collected, as well as cumulus cells (CCs) after superovulation from DIO and LEPT. The estrus cycle synchronization protocol showed increased protein levels of NLRP3 and interleukin (IL)-18 in diestrus, with this stage used for further sample collections. In DIO, protein expression of NLRP3 inflammasome components was increased in 4 week HFD, but decreased in 16 week HFD. Moreover, NLRP3 and IL-1β were upregulated in 16 L and downregulated in ob/ob. Transcriptome analysis of CC showed common genes between LEPT and 4 week HFD modulating NLRP3 inflammasome. Liver analysis showed NLRP3 protein upregulation after 16 week HFD in DIO, but also its downregulation in ob/ob−/−. We showed the link between leptin signaling and NLRP3 inflammasome activation in the ovary throughout obesity progression in mice, elucidating the molecular mechanisms underpinning ovarian failure in maternal obesity.
Collapse
Affiliation(s)
- Marek Adamowski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Karolina Wołodko
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Joana Oliveira
- Centro de Investigação em Ciências Veterinárias, Lusófona University, Lisbon, Portugal
| | | | - Daniel Murta
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Escola Superior de Saúde Egas Moniz, Campus Universitário, Monte de Caparica, Portugal.,Centro de Investigação Interdisciplinar em Sanidade Animal (C.I.I.S.A.), Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom.,Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - António M Galvão
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland.,Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom.,Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Liu D, Yu H, Gu Y, Pang Q. Effect of rare earth element lanthanum on lipid deposition and Wnt10b signaling in the liver of male zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 240:105994. [PMID: 34656894 DOI: 10.1016/j.aquatox.2021.105994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 10/02/2021] [Accepted: 10/06/2021] [Indexed: 06/13/2023]
Abstract
This paper investigates the effect of lanthanum (La) on lipid deposition and Wnt10b signaling in the liver of male zebrafish with exposure of 0, 10, 20, and 30 μmol/L La. It suggests that La can be accumulated in liver, and its treatments decrease the activities and gene expression of enzymes related to fatty acid synthesis. The levels of total cholesterol (TC), triglyceride (TG), and nonesterified fatty acids (NEFA) as well as the size of lipid droplets are decreased by La treatments. Moreover, La treatments affect the composition of fatty acids and the content of nutrient elements. Meanwhile, they also induce the gene expression of wnt10b, β-catenin, pparα, and pparγ, but inhibit gsk-3β gene expression in liver. Further study on the result of wnt10b gene interference shows that Wnt10b/β-catenin signaling plays a crucial role in the regulatory process of hepatic lipid deposition. Taken together, our observations suggest that La accumulation affects lipid deposition in the liver of male zebrafish, and Wnt10b signaling pathway may be involved in this process.
Collapse
Affiliation(s)
- Dongwu Liu
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255049, China; Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo 255049, China.
| | - Hairui Yu
- College of Biological and Agricultural Engineering, Weifang Bioengineering Technology Research Center, Weifang University, Weifang 261061, China
| | - Yaqi Gu
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo 255049, China
| | - Qiuxiang Pang
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo 255049, China.
| |
Collapse
|
12
|
Zhang Q, Wang Y, Yu N, Ding H, Li D, Zhao X. Metabolic syndrome predicts incident disability and functional decline among Chinese older adults: results from the China Health and Retirement Longitudinal Study. Aging Clin Exp Res 2021; 33:3073-3080. [PMID: 33743170 DOI: 10.1007/s40520-021-01827-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/27/2021] [Indexed: 12/20/2022]
Abstract
AIMS To investigate the longitudinal association of metabolic syndrome (MetS) and its components with disability outcomes. METHODS A total of 5875 participants aged 60 and above completed the 2011 and 2015 waves of the China Health and Retirement Longitudinal Study (CHARLS). MetS at baseline was measured by the National Cholesterol Education Program Adult Treatment Panel III criteria. Logistic regressions were conducted to analyze the associations between baseline MetS and incident disability, measured as the onset of limitations regarding instrumental activities of daily living (IADL) and activities of daily living (ADL) 4 years later. Linear regression was adopted to analyze the longitudinal impact of baseline MetS on the number of IADL and ADL limitations in 2015. A comprehensive list of baseline covariates was adjusted in all regression analyses. RESULTS Baseline MetS was related to increased odds of incident IADL disability (OR = 1.28, 95% CI 1.05-1.55) and incident ADL disability (OR = 1.27, 95% CI 1.05-1.53) among disability-free participants at baseline. Baseline MetS was also associated with an increase in the number of IADL (beta = 0.15, 95% CI 0.07-0.23) and ADL limitations (beta = 0.10, 95% CI 0.01-0.18), while adjusting for baseline functional performance. Significant MetS component predictors of disability outcomes include abdominal obesity, high blood pressure, and a low level of high-density lipoprotein cholesterol. CONCLUSIONS Our findings suggest an increased risk of incident disability and deteriorated functional performance over 4 years, associated with the presence of MetS and its components.
Collapse
|
13
|
Xu X, Huang X, Zhang L, Huang X, Qin Z, Hua F. Adiponectin protects obesity-related glomerulopathy by inhibiting ROS/NF-κB/NLRP3 inflammation pathway. BMC Nephrol 2021; 22:218. [PMID: 34107901 PMCID: PMC8191043 DOI: 10.1186/s12882-021-02391-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 05/05/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Adiponectin is an adipocytokine that plays a key regulatory role in glucose and lipid metabolism in obesity. The prevalence of obesity has led to an increase in the incidence of obesity-related glomerulopathy (ORG). This study aimed to identify the protective role of adiponectin in ORG. METHODS Small-interfering RNA (siRNA) against the gene encoding adiponectin was transfected into podocytes. The oxidative stress level was determined using a fluorometric assay. Apoptosis was analyzed by flow cytometry. The expressions of podocyte markers and pyrin domain containing protein 3 (NLRP3) inflammasome-related proteins were measured by qRT-PCR, immunohistochemistry, and Western blot. RESULTS Podocytes treated with palmitic acid (PA) showed downregulated expressions of podocyte markers, increased apoptosis, upregulated levels of NLRP3 inflammasome-related proteins, increased production of inflammatory cytokines (IL-18 and IL-1β), and induced activation of NF-κB as compared to the vehicle-treated controls. Decreased adiponectin expression was observed in the serum samples from high fat diet (HFD)-fed mice. Decreased podocin expression and upregulated NLRP3 expression were observed in the kidney samples from high fat diet (HFD)-fed mice. Treatment with adiponectin or the NLRP3 inflammasome inhibitor, MCC950, protected cultured podocytes against podocyte apoptosis and inflammation. Treatment with adiponectin protected mouse kidney tissues against decreased podocin expression and upregulated NLRP3 expression. The knockout of adiponectin gene by siRNA increased ROS production, resulting in the activation of NLRP3 inflammasome and the phosphorylation of NF-κB in podocytes. Pyrrolidine dithiocarbamate, an NF-κB inhibitor, prevented adiponectin from ameliorating FFA-induced podocyte injury and NLRP3 activation. CONCLUSIONS Our study showed that adiponectin ameliorated PA-induced podocyte injury in vitro and HFD-induced injury in vivo via inhibiting the ROS/NF-κB/NLRP3 pathway. These data suggest the potential use of adiponectin for the prevention and treatment of ORG.
Collapse
Affiliation(s)
- Xiaohong Xu
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, No.185 Bureau Front Street, 213003, Changzhou City, China
- Department of Nephrology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian City, China
- Department of Nephrology, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian City, China
| | - Xiaolin Huang
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, No.185 Bureau Front Street, 213003, Changzhou City, China
| | - Liexiang Zhang
- Department of Neurosurgery, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian City, China
- Department of Neurosurgery, Suqian People's Hospital, Nanjing Drum Tower Hospital Group, Suqian City, China
| | - Xiaoli Huang
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, No.185 Bureau Front Street, 213003, Changzhou City, China
| | - Zihan Qin
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, No.185 Bureau Front Street, 213003, Changzhou City, China
| | - Fei Hua
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, No.185 Bureau Front Street, 213003, Changzhou City, China.
| |
Collapse
|
14
|
Ahn SH, Lee JH, Lee JW. Inverse association between triglyceride glucose index and muscle mass in Korean adults: 2008-2011 KNHANES. Lipids Health Dis 2020; 19:243. [PMID: 33222694 PMCID: PMC7682063 DOI: 10.1186/s12944-020-01414-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022] Open
Abstract
Background Since sarcopenia is an important risk factor for falls or cardiovascular disease, early detection and prevention of sarcopenia are being increasingly emphasized. Emerging evidence has indicated relationships between sarcopenia, insulin resistance, and inflammation. The triglyceride glucose (TyG) index, a novel surrogate marker of insulin resistance and systemic inflammation, has not yet been shown to be associated with sarcopenia. This study aimed to examine the relationship between the TyG index and muscle mass in Korean adults. Methods This study included 15,741 non-diabetic adults over 19 years old using data from the 2008–2011 Korea National Health and Nutrition Examination Survey. Participants were divided into three groups according to tertiles of the TyG index. A low skeletal muscle mass index (LSMI) was defined by the Foundation for the National Institutes of Health Sarcopenia Project criteria. A weighted multivariate logistic regression model was used to analyze relationships between TyG index tertiles and LSMI. Results The ORs (95% CIs) for LSMI in the second and third TyG tertiles, compared to the first tertile, were 1.463 (1.131–1.892) and 1.816 (1.394–2.366), respectively, after adjusting for confounding factors. Higher TyG index values were also associated with increased odds of LSMI in adults under 65 years who did not exercise regularly, who consumed less than 30 g of alcohol per day, who did not currently smoke, and who ate less than 1.5 g of protein/kg/day. Conclusion The TyG index was significantly and positively associated with LSMI in Korean adults. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-020-01414-4.
Collapse
Affiliation(s)
- Sung-Ho Ahn
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, 211 Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea
| | - Jun-Hyuk Lee
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Department of Family Medicine, Yonsei University College of Medicine, Yongin Severance Hospital, 363, Dongbaekjukjeon-daero, Giheung-gu, Yongin-si, Gyeonggi-do, 16995, Republic of Korea.
| | - Ji-Won Lee
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Department of Family Medicine, Yonsei University College of Medicine, Gangnam Severance Hospital, 211 Eonju-ro, Gangnam-gu, Seoul, 06273, Republic of Korea.
| |
Collapse
|
15
|
García-García FJ, Monistrol-Mula A, Cardellach F, Garrabou G. Nutrition, Bioenergetics, and Metabolic Syndrome. Nutrients 2020; 12:E2785. [PMID: 32933003 PMCID: PMC7551996 DOI: 10.3390/nu12092785] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
According to the World Health Organization (WHO), the global nutrition report shows that whilst part of the world's population starves, the other part suffers from obesity and associated complications. A balanced diet counterparts these extreme conditions with the proper proportion, composition, quantity, and presence of macronutrients, micronutrients, and bioactive compounds. However, little is known on the way these components exert any influence on our health. These nutrients aiming to feed our bodies, our tissues, and our cells, first need to reach mitochondria, where they are decomposed into CO2 and H2O to obtain energy. Mitochondria are the powerhouse of the cell and mainly responsible for nutrients metabolism, but they are also the main source of oxidative stress and cell death by apoptosis. Unappropriated nutrients may support mitochondrial to become the Trojan horse in the cell. This review aims to provide an approach to the role that some nutrients exert on mitochondria as a major contributor to high prevalent Western conditions including metabolic syndrome (MetS), a constellation of pathologic conditions which promotes type II diabetes and cardiovascular risk. Clinical and experimental data extracted from in vitro animal and cell models further demonstrated in patients, support the idea that a balanced diet, in a healthy lifestyle context, promotes proper bioenergetic and mitochondrial function, becoming the best medicine to prevent the onset and progression of MetS. Any advance in the prevention and management of these prevalent complications help to face these challenging global health problems, by ameliorating the quality of life of patients and reducing the associated sociosanitary burden.
Collapse
Affiliation(s)
- Francesc Josep García-García
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| | - Anna Monistrol-Mula
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| | - Francesc Cardellach
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| | - Glòria Garrabou
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| |
Collapse
|
16
|
Wu L, Pan Y. Reactive oxygen species mediate TNF-α-induced inflammatory response in bone marrow mesenchymal cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2020; 22:1296-1301. [PMID: 32128094 PMCID: PMC7038432 DOI: 10.22038/ijbms.2019.37893.9006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objectives It is generally believed that the inflammatory response in bone marrow mesenchymal stem cells (BMSCs) transplantation leads to poor survival and unsatisfactory effects, and is mainly mediated by cytokines, including interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α). In this study, we explored the mechanisms underlying the TNF-α-induced inflammatory response in BMSCs. Materials and Methods We treated BMSCs with TNF-α (1 and 10 ng/ml) for 5 days. The expression levels of key inflammatory mediators were evaluated by Real-time PCR. Intracellular ROS level was measured by using a 2, 7-dichlorofluorescein diacetate (DCF-DA). Results We found that TNF-α treatment dramatically increased the expression levels of some key inflammatory mediators, including IL-6, IL-1β, IFN-γ and transforming growth factor β (TGF-β). Moreover, TNF-α induced intracellular oxidative stress by elevating intracellular reactive oxygen species (ROS) level, which is due to the increase of lipid peroxidation, the reduction of antioxidant Glutathione (GSH) levels and the inhibition of many antioxidant enzyme activities in BMSCs. Interestingly, 5 µM curcumin, a ROS scavenger, dramatically lowered the TNF-α-induced inflammatory response in BMSCs. In addition, TNF-α induced the activation of extracellular-signal-regulated kinases 1/2 (ERK1/2), c-Jun N-terminal kinases (JNK), p38 and their down-stream transcription factors nuclear factor kappa B (NF-κB) pathway. Conclusion ROS mediated the TNF-α-induced inflammatory response via MAPK and NF-κB pathway, and may provide a novel strategy to prevent the inflammatory-dependent impairments in BMSCs.
Collapse
Affiliation(s)
- Liuzhong Wu
- Department of Periodontics, School of Stomatology, China Medical University, Liaoning Province, China.,Department of Periodontics, Shenyang Stomatological Hospital, Shenyang, Liaoning Province, China
| | - Yaping Pan
- Department of Periodontics, School of Stomatology, China Medical University, Liaoning Province, China
| |
Collapse
|
17
|
Obesity Does Not Modulate the Glycometabolic Benefit of Insoluble Cereal Fibre in Subjects with Prediabetes-A Stratified Post Hoc Analysis of the Optimal Fibre Trial (OptiFiT). Nutrients 2019; 11:nu11112726. [PMID: 31717901 PMCID: PMC6893443 DOI: 10.3390/nu11112726] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/18/2019] [Accepted: 10/22/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity does not modulate the glycometabolic benefit of insoluble cereal fibre in subjects with prediabetes—a stratified post hoc analysis of the Optimal Fibre Trial (OptiFiT). Background: OptiFiT demonstrated the beneficial effect of insoluble oat fibres on dysglycemia in prediabetes. Recent analyses of OptiFiT and other randomised controlled trials (RCTs) indicated that this effect might be specific for the subgroup of patients with impaired fasting glucose (IFG). As subjects with IFG are more often obese, there is a need to clarify if the effect modulation is actually driven by glycemic state or body mass index (BMI). Aim: We conducted a stratified post hoc analysis of OptiFiT based on the presence or absence of obesity. Methods: 180 Caucasian participants with impaired glucose tolerance (IGT) were randomised in a double-blinded fashion to either twice-a-day fibre or placebo supplementation for 2 years (n = 89 and 91, respectively). Once a year, they underwent fasting blood sampling, an oral glucose tolerance test (oGTT) and full anthropometry. At baseline, out of 136 subjects who completed the first year of intervention, 87 (62%) were classified as OBESE (BMI >30) and 49 subjects were NONOBESE. We performed a stratified per-protocol analysis of the primary glycemic and secondary metabolic effects attributable to dietary fibre supplementation after 1 year of intervention. Results: Neither the NONOBESE nor the OBESE subgroup showed significant differences between the respective fibre and placebo groups in metabolic, anthropometric or inflammatory outcomes. None of the four subgroups showed a significant improvement in either fasting glucose or glycated haemoglobin (HbA1c) after 1 year of intervention and only OBESE fibre subjects improved 2 h glucose. Within the NONOBESE stratum, there were no significant differences in the change of primary or secondary metabolic parameters between the fibre and placebo arms. We found a significant interaction effect for leukocyte count (time × supplement × obesity status). Within the OBESE stratum, leukocyte count and gamma-glutamyl transferase (GGT) levels decreased more in the fibre group compared with placebo (adjusted for change in body weight). Comparison of both fibre groups revealed that OBESE subjects had a significantly stronger benefit with respect to leukocyte count and fasting C-peptide levels than NONOBESE participants. Only the effect on leukocyte count survived correction for multiple comparisons. In contrast, under placebo conditions, NONOBESE subjects managed to decrease their body fat content significantly more than OBESE ones. Intention-to-treat (ITT) analysis resulted in similar outcomes. Conclusions: The state of obesity does not relevantly modulate the beneficial effect of cereal fibre on major glycometabolic parameters by fibre supplementation, but leukocyte levels may be affected. Hence, BMI is not a suitable parameter to stratify this cohort with respect to diabetes risk or responsiveness to cereal fibre, but obesity needs to be accounted for when assessing anti-inflammatory effects of fibre treatments. Targeted diabetes prevention should focus on the actual metabolic state rather than on mere obesity.
Collapse
|
18
|
Kabisch S, Meyer NMT, Honsek C, Gerbracht C, Dambeck U, Kemper M, Osterhoff MA, Birkenfeld AL, Arafat AM, Hjorth MF, Weickert MO, Pfeiffer AFH. Fasting Glucose State Determines Metabolic Response to Supplementation with Insoluble Cereal Fibre: A Secondary Analysis of the Optimal Fibre Trial (OptiFiT). Nutrients 2019; 11:nu11102385. [PMID: 31590438 PMCID: PMC6835423 DOI: 10.3390/nu11102385] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 09/28/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023] Open
Abstract
Background: High intake of cereal fibre is associated with reduced risk for type 2 diabetes and long-term complications. Within the first long-term randomized controlled trial specifically targeting cereal fibre, the Optimal Fibre Trial (OptiFiT), intake of insoluble oat fibre was shown to significantly reduce glycaemia. Previous studies suggested that this effect might be limited to subjects with impaired fasting glucose (IFG). Aim: We stratified the OptiFiT cohort for normal and impaired fasting glucose (NFG, IFG) and conducted a secondary analysis comparing the effects of fibre supplementation between these subgroups. Methods: 180 Caucasian participants with impaired glucose tolerance (IGT) were randomized to twice-a-day fibre or placebo supplementation for 2 years (n = 89 and 91, respectively), while assuring double-blinded intervention. Fasting blood sampling, oral glucose tolerance test and full anthropometry were assessed annually. At baseline, out of 136 subjects completing the first year of intervention, 72 (54%) showed IFG and IGT, while 64 subjects had IGT only (labelled “NFG”). Based on these two groups, we performed a stratified per-protocol analysis of glycometabolic and secondary effects during the first year of intervention. Results: The NFG group did not show significant differences between fibre and placebo group concerning anthropometric, glycometabolic, or other biochemical parameters. Within the IFG stratum, 2-h glucose, HbA1c, and gamma-glutamyl transferase levels decreased more in the fibre group, with a significant supplement x IFG interaction effect for HbA1c. Compared to NFG subjects, IFG subjects had larger benefits from fibre supplementation with respect to fasting glucose levels. Results were robust against adjustment for weight change and sex. An ITT analysis did not reveal any differences from the per-protocol analysis. Conclusions: Although stratification resulted in relatively small subgroups, we were able to pinpoint our previous findings from the entire cohort to the IFG subgroup. Cereal fibre can beneficially affect glycemic metabolism, with most pronounced or even isolated effectiveness in subjects with impaired fasting glucose.
Collapse
Affiliation(s)
- Stefan Kabisch
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| | - Nina M T Meyer
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| | - Caroline Honsek
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
| | - Christiana Gerbracht
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
| | - Ulrike Dambeck
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
| | - Margrit Kemper
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
| | - Martin A Osterhoff
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
- Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany.
| | - Andreas L Birkenfeld
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
- Section of Metabolic Vascular Medicine, Medical Clinic III and Paul Langerhans Institute Dresden of the Helmholtz Center Munich at University Hospital and Faculty of Medicine, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany.
- Section of Diabetes and Nutritional Sciences, Rayne Institute, Denmark Hill Campus, King's College London, SE5 9NT London, UK.
| | - Ayman M Arafat
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
- Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany.
| | - Mads F Hjorth
- University of Copenhagen, Faculty of Science, Department of Nutrition, Exercise, and Sports, 2200 Copenhagen, Denmark.
| | - Martin O Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism; The ARDEN NET Centre, ENETS CoE; University Hospitals Coventry and Warwickshire NHS Trust, CV2 2DX Coventry, UK.
- Centre of Applied Biological & Exercise Sciences (ABES), Faculty of Health & Life Sciences, Coventry University, CV1 5FB Coventry, UK.
- Translational & Experimental Medicine, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, CV4 7AL Coventry, UK.
| | - Andreas F H Pfeiffer
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbrücke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
- Deutsches Zentrum für Diabetesforschung e.V., Geschäftsstelle am Helmholtz-Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany.
- Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, Charité University Medicine, Hindenburgdamm 30, 12203 Berlin, Germany.
| |
Collapse
|
19
|
Marisol MM, Celeste TM, Laura MM, Fernando EG, José PC, Alejandro Z, Omar MC, Francisco AA, Julio César AP, Erika CN, Angélica SC, Gladis F, Enrique JF, Gabriela R. Effect of Cucumis sativus on Dysfunctional 3T3-L1 Adipocytes. Sci Rep 2019; 9:13372. [PMID: 31527805 PMCID: PMC6746747 DOI: 10.1038/s41598-019-49458-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/23/2019] [Indexed: 12/13/2022] Open
Abstract
Obesity is caused by lipid accumulation in adipose tissues inducing adipocyte dysfunction, characterized by insulin resistance, increased lipolysis, oxidative stress, and inflammation, leading to increased levels of adipokines. Herein the capacity of the subfractions (SFs) SF1, SF2, and SF3 from the Cucumis sativus aqueous fraction and their combinations (M) to control adipocyte dysfunction in vitro, in 3T3-L1 adipocytes was studied. Adipocytes, previously treated with dexamethasone or IL-1 to induce dysfunction, were incubated with different concentrations of the subfractions for 24 h. 2-deoxyglucose consumption and glycerol release were evaluated, and a surface model was constructed to determine the most effective SF concentrations to improve both parameters. Effective SF combinations were assessed in their capacity to control metabolic, pro-oxidative, and pro-inflammatory conditions. SF1, SF2 (40 μg/ml each) and SF3 (20 μg/ml) improved 2-deoxyglucose consumption by 87%, 57%, and 87%, respectively. SF1 and SF2 (5 μg/ml each) and SF3 (40 μg/mL) increased glycerol secretion by 10.6%, 18.9%, and 11.8%, respectively. Among five combinations tested, only M4 (SF1 40 μg/ml:SF2 60 μg/ml:SF3 30 μg/ml) and M5 (SF1 40 μg/ml:SF2 60 μg/mL:SF3 10 μg/ml) controlled effectively the metabolic, pro-oxidative, and proinflammatory conditions studied. Glycine, asparagine, and arginine were the main components in these SFs.
Collapse
Affiliation(s)
- Méndez-Martínez Marisol
- Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, CP, 62209, Mexico
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, CP, 62350, Mexico
| | - Trejo-Moreno Celeste
- Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, CP, 62209, Mexico
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, CP, 62350, Mexico
| | - Maldonado-Mejía Laura
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, CP, 62350, Mexico
| | | | - Pedraza-Chaverri José
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, CP, 04510, Mexico
| | - Zamilpa Alejandro
- Laboratorio de Farmacología, Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec, Morelo, CP, 62790s, Mexico
| | - Medina-Campos Omar
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Coyoacán, Ciudad de México, CP, 04510, Mexico
| | - Alarcón-Aguilar Francisco
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana de Iztapalapa, CDMX, C.P, 09640, Mexico
| | - Almanza-Pérez Julio César
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana de Iztapalapa, CDMX, C.P, 09640, Mexico
| | - Contreras-Nuñez Erika
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana de Iztapalapa, CDMX, C.P, 09640, Mexico
| | - Santana-Calderón Angélica
- Centro de Investigación en Dinámica Celular (IICBA), Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, CP, 62209, Mexico
| | - Fragoso Gladis
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, CP, 04510, Mexico
| | - Jiménez-Ferrer Enrique
- Laboratorio de Farmacología, Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec, Morelo, CP, 62790s, Mexico.
| | - Rosas Gabriela
- Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, CP, 62209, Mexico.
| |
Collapse
|
20
|
Tamarai K, Bhatti JS, Reddy PH. Molecular and cellular bases of diabetes: Focus on type 2 diabetes mouse model-TallyHo. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2276-2284. [PMID: 31082469 DOI: 10.1016/j.bbadis.2019.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/17/2022]
Abstract
Diabetes is a chronic lifestyle disorder that affects millions of people worldwide. Diabetes is a condition where the body does not produce sufficient insulin or does not use it efficiently. Insulin resistance in diabetes or obesity causes the pancreatic β-cells to increase the insulin output. Diabetes occurs in multiple forms, including type 1, type 2, type 3 and gestational. Type 2 diabetes accounts for ∼90-95% of total affected population and is associated with both impaired insulin production by the β-cells of the pancreas and impaired insulin release in response to high blood glucose levels. Diabetes is tightly linked with genetic mutations and genetic and lifestyle activities, including diet and exercise. Recent epidemiological studies established a close link between the diabetes and progression to Alzheimer's disease. This article summarizes various molecular mechanisms involved in the developments of diabetes, including biochemical characteristics, genetic and molecular links with Alzheimer's disease, β-cell function, and factors associated with diabetes. This will help us in the development of novel therapeutic strategies targeting AD in future.
Collapse
Affiliation(s)
- Kavya Tamarai
- Internal Medicine Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, Lubbock, TX 79430, United States
| | - Jasvinder Singh Bhatti
- Internal Medicine Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, Lubbock, TX 79430, United States; Department of Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India
| | - P Hemachandra Reddy
- Internal Medicine Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, Lubbock, TX 79430, United States; Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, Lubbock, TX 79430, United States; Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Neurology Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Speech, Language and Hearing Sciences Department, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Department of Public Health, Graduate School of Biomedical Sciences, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States.
| |
Collapse
|
21
|
Pinti MV, Fink GK, Hathaway QA, Durr AJ, Kunovac A, Hollander JM. Mitochondrial dysfunction in type 2 diabetes mellitus: an organ-based analysis. Am J Physiol Endocrinol Metab 2019; 316:E268-E285. [PMID: 30601700 PMCID: PMC6397358 DOI: 10.1152/ajpendo.00314.2018] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/27/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a systemic disease characterized by hyperglycemia, hyperlipidemia, and organismic insulin resistance. This pathological shift in both circulating fuel levels and energy substrate utilization by central and peripheral tissues contributes to mitochondrial dysfunction across organ systems. The mitochondrion lies at the intersection of critical cellular pathways such as energy substrate metabolism, reactive oxygen species (ROS) generation, and apoptosis. It is the disequilibrium of these processes in T2DM that results in downstream deficits in vital functions, including hepatocyte metabolism, cardiac output, skeletal muscle contraction, β-cell insulin production, and neuronal health. Although mitochondria are known to be susceptible to a variety of genetic and environmental insults, the accumulation of mitochondrial DNA (mtDNA) mutations and mtDNA copy number depletion is helping to explain the prevalence of mitochondrial-related diseases such as T2DM. Recent work has uncovered novel mitochondrial biology implicated in disease progressions such as mtDNA heteroplasmy, noncoding RNA (ncRNA), epigenetic modification of the mitochondrial genome, and epitranscriptomic regulation of the mtDNA-encoded mitochondrial transcriptome. The goal of this review is to highlight mitochondrial dysfunction observed throughout major organ systems in the context of T2DM and to present new ideas for future research directions based on novel experimental and technological innovations in mitochondrial biology. Finally, the field of mitochondria-targeted therapeutics is discussed, with an emphasis on novel therapeutic strategies to restore mitochondrial homeostasis in the setting of T2DM.
Collapse
Affiliation(s)
- Mark V Pinti
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
- West Virginia University School of Pharmacy , Morgantown, West Virginia
| | - Garrett K Fink
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Quincy A Hathaway
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
- Toxicology Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Andrya J Durr
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| | - Amina Kunovac
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| | - John M Hollander
- Division of Exercise Physiology, West Virginia University School of Medicine , Morgantown, West Virginia
- Mitochondria, Metabolism, and Bioenergetics Working Group, West Virginia University School of Medicine , Morgantown, West Virginia
| |
Collapse
|
22
|
Rheinheimer J, de Souza BM, Cardoso NS, Bauer AC, Crispim D. Current role of the NLRP3 inflammasome on obesity and insulin resistance: A systematic review. Metabolism 2017; 74:1-9. [PMID: 28764843 DOI: 10.1016/j.metabol.2017.06.002] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/17/2017] [Accepted: 06/07/2017] [Indexed: 12/13/2022]
Abstract
NLRP3 inflammasome activation seems to be a culprit behind the chronic inflammation characteristic of obesity and insulin resistance (IR). Nutrient excess generates danger-associated molecules that activate NLRP3 inflammasome-caspase 1, leading to maturation of IL-1β and IL-18, which are proinflammatory cytokines released by immune cells infiltrating the adipose tissue (AT) from obese subjects. Although several studies have reported an association of the NLRP3 inflammasome with obesity and/or IR; contradictory results were also reported by other studies. Therefore, we conducted a systematic review to summarize results of studies that evaluated the association of the NLRP3 with obesity and IR. Nineteen studies were included in the review. These studies focused on NLRP3 expression/polymorphism analyses in AT. Overall, human studies indicate that obesity and IR are associated with increased NLRP3 expression in AT. Studies in obese mice corroborate this association. Moreover, high fat diet (HFD) increases Nlrp3 expression in murine AT while calorie-restricted diet decreases its expression. Hence, Nlrp3 blockade in mice protects against HFD-induced obesity and IR. NLRP3 rs10754558 polymorphism is associated with risk for T2DM in Chinese Han populations. In conclusion, available studies strongly points for an association between NLRP3 inflammasome and obesity/IR.
Collapse
Affiliation(s)
- Jakeline Rheinheimer
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Post-graduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Bianca M de Souza
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Post-graduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Natali S Cardoso
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Andrea C Bauer
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Post-graduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Daisy Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Post-graduation Program in Endocrinology, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
23
|
Bhatti JS, Bhatti GK, Reddy PH. Mitochondrial dysfunction and oxidative stress in metabolic disorders - A step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1066-1077. [PMID: 27836629 PMCID: PMC5423868 DOI: 10.1016/j.bbadis.2016.11.010] [Citation(s) in RCA: 832] [Impact Index Per Article: 118.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/02/2016] [Accepted: 11/03/2016] [Indexed: 01/06/2023]
Abstract
Mitochondria are the powerhouses of the cell and are involved in essential functions of the cell, including ATP production, intracellular Ca2+ regulation, reactive oxygen species production & scavenging, regulation of apoptotic cell death and activation of the caspase family of proteases. Mitochondrial dysfunction and oxidative stress are largely involved in aging, cancer, age-related neurodegenerative and metabolic syndrome. In the last decade, tremendous progress has been made in understanding mitochondrial structure, function and their physiology in metabolic syndromes such as diabetes, obesity, stroke and hypertension, and heart disease. Further, progress has also been made in developing therapeutic strategies, including lifestyle interventions (healthy diet and regular exercise), pharmacological strategies and mitochondria-targeted approaches. These strategies were mainly focused to reduce mitochondrial dysfunction and oxidative stress and to maintain mitochondrial quality in metabolic syndromes. The purpose of our article is to highlight the recent progress on the mitochondrial role in metabolic syndromes and also summarize the progress of mitochondria-targeted molecules as therapeutic targets to treat metabolic syndromes. 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)
- Jasvinder Singh Bhatti
- Department of Biotechnology and Bioinformatics, Sri Guru Gobind Singh College, Sector-26, Chandigarh 160019, India; Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States.
| | - Gurjit Kaur Bhatti
- UGC Centre of Excellence in Nano applications, Panjab University, UIPS building, Chandigarh 160014, India
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neuroscience & Pharmacology Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Neurology Department, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Speech, Language and Hearing Sciences Departments, Texas Tech University Health Sciences Center, 3601 4th Street, MS 9424, Lubbock, TX 79430, United States; Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Suite E, MS 7495, Lubbock, TX 79413, United States
| |
Collapse
|
24
|
Effect of functional sympathetic nervous system impairment of the liver and abdominal visceral adipose tissue on circulating triglyceride-rich lipoproteins. PLoS One 2017; 12:e0173934. [PMID: 28346471 PMCID: PMC5367791 DOI: 10.1371/journal.pone.0173934] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/28/2017] [Indexed: 02/07/2023] Open
Abstract
Background Interruption of sympathetic innervation to the liver and visceral adipose tissue (VAT) in animal models has been reported to reduce VAT lipolysis and hepatic secretion of very low density lipoprotein (VLDL) and concentrations of triglyceride-rich lipoprotein particles. Whether functional impairment of sympathetic nervous system (SNS) innervation to tissues of the abdominal cavity reduce circulating concentrations of triglyceride (TG) and VLDL particles (VLDL-P) was tested in men with spinal cord injury (SCI). Methods One hundred-three non-ambulatory men with SCI [55 subjects with neurologic injury at or proximal to the 4th thoracic vertebrae (↑T4); 48 subjects with SCI at or distal to the 5th thoracic vertebrae (↓T5)] and 53 able-bodied (AB) subjects were studied. Fasting blood samples were obtained for determination of TG, VLDL-P concentration by NMR spectroscopy, serum glucose by autoanalyzer, and plasma insulin by radioimmunoassay. VAT volume was determined by dual energy x-ray absorptiometry imaging with calculation by a validated proprietary software package. Results Significant group main effects for TG and VLDL-P were present; post-hoc tests revealed that serum TG concentrations were significantly higher in ↓T5 group compared to AB and ↑T4 groups [150±9 vs. 101±8 (p<0.01) and 112±8 mg/dl (p<0.05), respectively]. VLDL-P concentration was significantly elevated in ↓T5 group compared to AB and ↑T4 groups [74±4 vs. 58±4 (p<0.05) and 55±4 μmol/l (p<0.05)]. VAT volume was significantly higher in both SCI groups than in the AB group, and HOMA-IR was higher and approached significance in the SCI groups compared to the AB group. A linear relationship between triglyceride rich lipoproteins (i.e., TG or Large VLDL-P) and VAT volume or HOMA-IR was significant only in the ↓T5 group. Conclusions Despite a similar VAT volume and insulin resistance in both SCI groups, the ↓T5 group had significantly higher serum TG and VLDL-P values than that observed in the ↑T4 and the AB control groups. Thus, level of injury is an important determinate of the concentration of circulating triglyceride rich lipoproteins, which may play a role in the genesis of cardiometabolic dysfunction.
Collapse
|
25
|
Bhatti JS, Kumar S, Vijayan M, Bhatti GK, Reddy PH. Therapeutic Strategies for Mitochondrial Dysfunction and Oxidative Stress in Age-Related Metabolic Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 146:13-46. [PMID: 28253984 DOI: 10.1016/bs.pmbts.2016.12.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mitochondria are complex, intercellular organelles present in the cells and are involved in multiple roles including ATP formation, free radicals generation and scavenging, calcium homeostasis, cellular differentiation, and cell death. Many studies depicted the involvement of mitochondrial dysfunction and oxidative damage in aging and pathogenesis of age-related metabolic disorders and neurodegenerative diseases. Remarkable advancements have been made in understanding the structure, function, and physiology of mitochondria in metabolic disorders such as diabetes, obesity, cardiovascular diseases, and stroke. Further, much progress has been done in the improvement of therapeutic strategies, including lifestyle interventions, pharmacological, and mitochondria-targeted therapeutic approaches. These strategies were mainly focused to reduce the mitochondrial dysfunction caused by oxidative stress and to retain the mitochondrial health in various diseases. In this chapter, we have highlighted the involvement of mitochondrial dysfunction in the pathophysiology of various disorders and recent progress in the development of mitochondria-targeted molecules as therapeutic measures for metabolic disorders.
Collapse
Affiliation(s)
- J S Bhatti
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Department of Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India.
| | - S Kumar
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - M Vijayan
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - G K Bhatti
- UGC Centre of Excellence in Nano Applications, Panjab University, Chandigarh, India
| | - P H Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
26
|
Rehman K, Akash MSH. Mechanisms of inflammatory responses and development of insulin resistance: how are they interlinked? J Biomed Sci 2016; 23:87. [PMID: 27912756 PMCID: PMC5135788 DOI: 10.1186/s12929-016-0303-y] [Citation(s) in RCA: 298] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 11/24/2016] [Indexed: 02/06/2023] Open
Abstract
Background Insulin resistance (IR) is one of the major hallmark for pathogenesis and etiology of type 2 diabetes mellitus (T2DM). IR is directly interlinked with various inflammatory responses which play crucial role in the development of IR. Inflammatory responses play a crucial role in the pathogenesis and development of IR which is one of the main causative factor for the etiology of T2DM. Methods A comprehensive online English literature was searched using various electronic search databases. Different search terms for pathogenesis of IR, role of various inflammatory responses were used and an advanced search was conducted by combining all the search fields in abstracts, keywords, and titles. Results We summarized the data from the searched articles and found that inflammatory responses activate the production of various pro-inflammatory mediators notably cytokines, chemokines and adipocytokines through the involvement of various transcriptional mediated molecular pathways, oxidative and metabolic stress. Overnutrition is one of the major causative factor that contributes to induce the state of low-grade inflammation due to which accumulation of elevated levels of glucose and/or lipids in blood stream occur that leads to the activation of various transcriptional mediated molecular and metabolic pathways. This results in the induction of various pro-inflammatory mediators that are decisively involved to provoke the pathogenesis of tissue-specific IR by interfering with insulin signaling pathways. Once IR is developed, it increases oxidative stress in β-cells of pancreatic islets and peripheral tissues which impairs insulin secretion, and insulin sensitivity in β-cells of pancreatic islets and peripheral tissues, respectively. Moreover, we also summarized the data regarding various treatment strategies of inflammatory responses-induced IR. Conclusions In this article, we have briefly described that how pro-inflammatory mediators, oxidative stress, transcriptional mediated molecular and metabolic pathways are involved in the pathogenesis of tissues-specific IR. Moreover, based on recent investigations, we have also described that to counterfeit these inflammatory responses is one of the best treatment strategy to prevent the pathogenesis of IR through ameliorating the incidences of inflammatory responses.
Collapse
Affiliation(s)
- Kanwal Rehman
- Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
27
|
Zhang Q, Sun X, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang Z, Qi C, Wang T, Wang X. Maternal Chromium Restriction Leads to Glucose Metabolism Imbalance in Mice Offspring through Insulin Signaling and Wnt Signaling Pathways. Int J Mol Sci 2016; 17:ijms17101767. [PMID: 27782077 PMCID: PMC5085791 DOI: 10.3390/ijms17101767] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/08/2016] [Accepted: 10/17/2016] [Indexed: 12/13/2022] Open
Abstract
An adverse intrauterine environment, induced by a chromium-restricted diet, is a potential cause of metabolic disease in adult life. Up to now, the relative mechanism has not been clear. C57BL female mice were time-mated and fed either a control diet (CD), or a chromium-restricted diet (CR) throughout pregnancy and the lactation period. After weaning, some offspring continued the diet diagram (CD-CD or CR-CR), while other offspring were transferred to another diet diagram (CD-CR or CR-CD). At 32 weeks of age, glucose metabolism parameters were measured, and the liver from CR-CD group and CD-CD group was analyzed using a gene array. Quantitative real-time polymerase chain reaction (qPCR) and Western blot were used to verify the result of the gene array. A maternal chromium-restricted diet resulted in obesity, hyperglycemia, hyperinsulinemia, increased area under the curve (AUC) of glucose in oral glucose tolerance testing and homeostasis model assessment of insulin resistance (HOMA-IR). There were 463 genes that differed significantly (>1.5-fold change, p < 0.05) between CR-CD offspring (264 up-regulated genes, 199 down-regulated genes) and control offspring. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and STRING (Search Tool for the Retrieval of Interacting Genes/Proteins) analysis revealed that the insulin signaling pathway and Wnt signaling pathway were in the center of the gene network. Our study provides the first evidence that maternal chromium deficiency influences glucose metabolism in pups through the regulation of insulin signaling and Wnt signaling pathways.
Collapse
Affiliation(s)
- Qian Zhang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Xiaofang Sun
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Jia Zheng
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Ming Li
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Miao Yu
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Fan Ping
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Zhixin Wang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Cuijuan Qi
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Tong Wang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Xiaojing Wang
- Key Laboratory of Endocrinology, Translational Medicine Centre, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
28
|
Karamchand S, Leisegang R, Schomaker M, Maartens G, Walters L, Hislop M, Dave JA, Levitt NS, Cohen K. Risk Factors for Incident Diabetes in a Cohort Taking First-Line Nonnucleoside Reverse Transcriptase Inhibitor-Based Antiretroviral Therapy. Medicine (Baltimore) 2016; 95:e2844. [PMID: 26945366 PMCID: PMC4782850 DOI: 10.1097/md.0000000000002844] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Efavirenz is the preferred nonnucleoside reverse transcriptase inhibitor (NNRTI) in first-line antiretroviral therapy (ART) regimens in low- and middle-income countries, where the prevalence of diabetes is increasing. Randomized control trials have shown mild increases in plasma glucose in participants in the efavirenz arms, but no association has been reported with overt diabetes. We explored the association between efavirenz exposure and incident diabetes in a large Southern African cohort commencing NNRTI-based first-line ART. Our cohort included HIV-infected adults starting NNRTI-based ART in a private sector HIV disease management program from January 2002 to December 2011. Incident diabetes was identified by the initiation of diabetes treatment. Patients with prevalent diabetes were excluded. We included 56,298 patients with 113,297 patient-years of follow-up (PYFU) on first-line ART. The crude incidence of diabetes was 13.24 per 1000 PYFU. Treatment with efavirenz rather than nevirapine was associated with increased risk of developing diabetes (hazard ratio 1.27 (95% confidence interval (CI): 1.10-1.46)) in a multivariate analysis adjusting for age, sex, body mass index, baseline CD4 count, viral load, NRTI backbone, and exposure to other diabetogenic medicines. Zidovudine and stavudine exposure were also associated with an increased risk of developing diabetes. We found that treatment with efavirenz, as well as stavudine and zidovudine, increased the risk of incident diabetes. Interventions to detect and prevent diabetes should be implemented in ART programs, and use of antiretrovirals with lower risk of metabolic complications should be encouraged.
Collapse
Affiliation(s)
- Sumanth Karamchand
- From the Division of Clinical Pharmacology (SK, RL, GM, KC), Division of Endocrinology, Department of Medicine (JAD, NSL), Center for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town (MS), Aid for AIDS Management (Pty) Limited (MH), Health Intelligence Unit, Medscheme (Pty) Limited (LW), Chronic Disease Initiative for Africa, Cape Town (JAD, NSL), South Africa
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Liu Z, Gan L, Chen Y, Luo D, Zhang Z, Cao W, Zhou Z, Lin X, Sun C. Mark4 promotes oxidative stress and inflammation via binding to PPARγ and activating NF-κB pathway in mice adipocytes. Sci Rep 2016; 6:21382. [PMID: 26888669 PMCID: PMC4766853 DOI: 10.1038/srep21382] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 01/22/2016] [Indexed: 12/25/2022] Open
Abstract
MAP/Microtubule affinity-regulating kinase 4 (Mark4) plays an important role in the regulation of microtubule organization, adipogenesis and apoptosis. However, the role of Mark4 plays in oxidative stress and inflammation are poorly understood. In this study, we found Mark4 was induced by high fat diet (HFD) while PPARγ was elevated significantly in mice adipocytes. Further analyses revealed Mark4 impaired mitochondrial oxidative respiration and increased reactive oxygen species (ROS) production. At same time, the activities of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx) were greatly reduced. By treating cells with H2O2 and vitamin E (VE), Mark4 accentuated oxidative stress along with increased mRNA level of inflammatory factor interleukin-6 (IL-6) and decreased leptin mRNA. Furthermore, we found PPARγ bind to Mark4 promoter region and inhibited Mark4 expression. We showed PPARγ interacted with Mark4 and inhibited the stimulating effect of Mark4 on oxidative stress and inflammation. Finally, we demonstrated that the IKKα/NF-κB signal pathway was involved in Mark4 induced oxidative stress and inflammation, while PTDC, a special inhibitor of NF-κB signal pathway, reduced oxidative stress and inflammation. Thus, our study indicated that Mark4 was a potential drug target for treating metabolic diseases.
Collapse
Affiliation(s)
- Zhenjiang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Lu Gan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yizhe Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Dan Luo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhenzhen Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Weina Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhongjie Zhou
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xueting Lin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
30
|
Huth C, Beuerle S, Zierer A, Heier M, Herder C, Kaiser T, Koenig W, Kronenberg F, Oexle K, Rathmann W, Roden M, Schwab S, Seissler J, Stöckl D, Meisinger C, Peters A, Thorand B. Biomarkers of iron metabolism are independently associated with impaired glucose metabolism and type 2 diabetes: the KORA F4 study. Eur J Endocrinol 2015; 173:643-53. [PMID: 26294793 DOI: 10.1530/eje-15-0631] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/20/2015] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Iron has been suggested to play a role in the etiology of type 2 diabetes mellitus (T2DM). Except for ferritin, evidence is sparse for other markers of iron metabolism that are regulated differently and might act through independent pathways. We therefore investigated the associations of serum ferritin, transferrin, soluble transferrin receptor (sTfR), transferrin saturation (TSAT), sTfR-to-log10ferritin (sTfR-F) index, and iron with impaired glucose metabolism (IGM/'prediabetes'), T2DM, and four continuous glucose and insulin traits. DESIGN AND METHODS Data from 2893 participants of the population-based Cooperative Health Research in the Region of Augsburg (KORA) F4 study (Germany) was investigated through regression analysis. The results were adjusted for socio-demographic, life-style, and obesity measures as well as metabolic, inflammatory, and other iron biomarkers following a step-wise approach. Non-linearity was tested by adding a non-linear spline component to the model. RESULTS Ferritin and transferrin were positively associated with IGM (fourth vs first sex-specific quartile: ferritin odds ratio (OR)=2.08 (95% CI 1.43-3.04) and transferrin OR=1.89 (95% CI 1.32-2.70)), T2DM (ferritin OR=1.98 (95% CI 1.22-3.22) and transferrin OR=2.42 (95% CI 1.54-3.81)), and fasting as well as 2-h glucose. TSAT (OR=0.55 (95% CI 0.34-0.88)) and iron (OR=0.61 (95% CI 0.38-0.97)) were inversely associated with T2DM, sTfR-F-index was inversely associated with IGM (OR=0.67 (95% CI 0.48-0.95)). There was no strong evidence for non-linear relationships. CONCLUSIONS The observed associations of several markers of iron metabolism with hyperglycemia and insulin resistance suggest that iron stores as well as iron-related metabolic pathways contribute to the pathogenesis of IGM and T2DM. Moreover, TSAT levels are decreased in T2DM patients.
Collapse
Affiliation(s)
- Cornelia Huth
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Simon Beuerle
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Astrid Zierer
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Margit Heier
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Christian Herder
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Thorsten Kaiser
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Wolfgang Koenig
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Florian Kronenberg
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Konrad Oexle
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Wolfgang Rathmann
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Michael Roden
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Sigrid Schwab
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Jochen Seissler
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Doris Stöckl
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Christa Meisinger
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Annette Peters
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Barbara Thorand
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| |
Collapse
|
31
|
Weber KS, Nowotny B, Strassburger K, Pacini G, Müssig K, Szendroedi J, Herder C, Roden M. The Role of Markers of Low-Grade Inflammation for the Early Time Course of Glycemic Control, Glucose Disappearance Rate, and β-Cell Function in Recently Diagnosed Type 1 and Type 2 Diabetes. Diabetes Care 2015; 38:1758-67. [PMID: 26153272 DOI: 10.2337/dc15-0169] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/27/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Inflammatory processes are involved in the progression of insulin resistance and β-cell dysfunction in individuals with prediabetes and contribute to the development of diabetes. We hypothesized that higher levels of biomarkers of low-grade inflammation are associated with the early progression of recently diagnosed diabetes. RESEARCH DESIGN AND METHODS Within the prospective German Diabetes Study, patients with recently diagnosed type 1 (n = 42) and type 2 (n = 94) diabetes underwent detailed metabolic characterization within the first year after diagnosis and 2 years thereafter. Associations between changes in markers of low-grade inflammation with changes in glycemic control, β-cell function, and glucose disappearance rate were assessed using multivariable linear regression analysis. Associations were adjusted for age, sex, BMI, smoking status, and 2-year changes in BMI, smoking status, and glucose-lowering medication. RESULTS Patients with type 1 and type 2 diabetes exhibited good glucometabolic control at baseline (mean HbA1c 7.08 ± 1.58% [54 ± 17 mmol/mol] and 6.43 ± 0.98% [47 ± 11 mmol/mol], respectively) and 2 years thereafter (mean HbA1c 7.03 ± 1.20% [53 ± 13 mmol/mol] and 6.62 ± 1.14% [49 ± 13], respectively). Two-year increases of high-sensitivity C-reactive protein, soluble E-selectin (sE-selectin), and soluble intercellular adhesion molecule-1 in type 2 diabetes and of IL-18 in type 1 diabetes were associated with 2-year increases of HbA1c. Additionally, 2-year increases of sE-selectin were associated with 2-year decreases of prehepatic β-cell function in type 2 diabetes (all P < 0.05). CONCLUSIONS These data indicate that with the clinical onset of diabetes, low-grade inflammation relates to worsening of glycemia and that endothelial activation may contribute to decreasing β-cell function.
Collapse
Affiliation(s)
- Katharina S Weber
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Institute for Diabetes Research, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Bettina Nowotny
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Institute for Diabetes Research, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Klaus Strassburger
- German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany Institute for Biometrics and Epidemiology, German Diabetes Center at Heinrich Heine University, Leibniz Institute for Diabetes Research, Düsseldorf, Germany
| | - Giovanni Pacini
- Metabolic Unit, Institute of Biomedical Engineering, National Research Council, Padua, Italy
| | - Karsten Müssig
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Institute for Diabetes Research, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Julia Szendroedi
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Institute for Diabetes Research, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Institute for Diabetes Research, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany
| | - Michael Roden
- Institute for Clinical Diabetology, German Diabetes Center at Heinrich Heine University, Leibniz Institute for Diabetes Research, Düsseldorf, Germany German Center for Diabetes Research, Partner Düsseldorf, Düsseldorf, Germany Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | | |
Collapse
|
32
|
Shi F, Kouadir M, Yang Y. NALP3 inflammasome activation in protein misfolding diseases. Life Sci 2015; 135:9-14. [DOI: 10.1016/j.lfs.2015.05.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 04/09/2015] [Accepted: 05/03/2015] [Indexed: 01/26/2023]
|
33
|
Duodenal-jejunal exclusion improves insulin resistance in type 2 diabetic rats by upregulating the hepatic insulin signaling pathway. Nutrition 2015; 31:733-9. [DOI: 10.1016/j.nut.2014.10.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 09/14/2014] [Accepted: 10/22/2014] [Indexed: 12/19/2022]
|
34
|
Jiang N, Zhang S, Zhu J, Shang J, Gao X. Hypoglycemic, Hypolipidemic and Antioxidant Effects of Peptides from Red Deer Antlers in Streptozotocin-Induced Diabetic Mice. TOHOKU J EXP MED 2015; 236:71-9. [DOI: 10.1620/tjem.236.71] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Ning Jiang
- Department of Biological Science and Technology, Hubei University for Nationalities
- School of Life Science and Technology, China Pharmaceutical University
| | - Shuangjian Zhang
- School of Life Science and Technology, China Pharmaceutical University
| | - Jing Zhu
- School of Life Science and Technology, China Pharmaceutical University
| | - Jing Shang
- New Drug Screening Center, China Pharmaceutical University
| | - Xiangdong Gao
- School of Life Science and Technology, China Pharmaceutical University
| |
Collapse
|
35
|
Ageing, adipose tissue, fatty acids and inflammation. Biogerontology 2014; 16:235-48. [PMID: 25367746 DOI: 10.1007/s10522-014-9536-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 10/20/2014] [Indexed: 12/22/2022]
Abstract
A common feature of ageing is the alteration in tissue distribution and composition, with a shift in fat away from lower body and subcutaneous depots to visceral and ectopic sites. Redistribution of adipose tissue towards an ectopic site can have dramatic effects on metabolic function. In skeletal muscle, increased ectopic adiposity is linked to insulin resistance through lipid mediators such as ceramide or DAG, inhibiting the insulin receptor signalling pathway. Additionally, the risk of developing cardiovascular disease is increased with elevated visceral adipose distribution. In ageing, adipose tissue becomes dysfunctional, with the pathway of differentiation of preadipocytes to mature adipocytes becoming impaired; this results in dysfunctional adipocytes less able to store fat and subsequent fat redistribution to ectopic sites. Low grade systemic inflammation is commonly observed in ageing, and may drive the adipose tissue dysfunction, as proinflammatory cytokines are capable of inhibiting adipocyte differentiation. Beyond increased ectopic adiposity, the effect of impaired adipose tissue function is an elevation in systemic free fatty acids (FFA), a common feature of many metabolic disorders. Saturated fatty acids can be regarded as the most detrimental of FFA, being capable of inducing insulin resistance and inflammation through lipid mediators such as ceramide, which can increase risk of developing atherosclerosis. Elevated FFA, in particular saturated fatty acids, maybe a driving factor for both the increased insulin resistance, cardiovascular disease risk and inflammation in older adults.
Collapse
|
36
|
Lasram MM, Dhouib IB, Annabi A, El Fazaa S, Gharbi N. A review on the molecular mechanisms involved in insulin resistance induced by organophosphorus pesticides. Toxicology 2014; 322:1-13. [DOI: 10.1016/j.tox.2014.04.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 04/23/2014] [Accepted: 04/24/2014] [Indexed: 02/06/2023]
|
37
|
Gao D, Madi M, Ding C, Fok M, Steele T, Ford C, Hunter L, Bing C. Interleukin-1β mediates macrophage-induced impairment of insulin signaling in human primary adipocytes. Am J Physiol Endocrinol Metab 2014; 307:E289-304. [PMID: 24918199 PMCID: PMC4121578 DOI: 10.1152/ajpendo.00430.2013] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 05/20/2014] [Indexed: 01/21/2023]
Abstract
Adipose tissue expansion during obesity is associated with increased macrophage infiltration. Macrophage-derived factors significantly alter adipocyte function, inducing inflammatory responses and decreasing insulin sensitivity. Identification of the major factors that mediate detrimental effects of macrophages on adipocytes may offer potential therapeutic targets. IL-1β, a proinflammatory cytokine, is suggested to be involved in the development of insulin resistance. This study investigated the role of IL-1β in macrophage-adipocyte cross-talk, which affects insulin signaling in human adipocytes. Using macrophage-conditioned (MC) medium and human primary adipocytes, we examined the effect of IL-1β antagonism on the insulin signaling pathway. Gene expression profile and protein abundance of insulin signaling molecules were determined, as was the production of proinflammatory cytokine/chemokines. We also examined whether IL-1β mediates MC medium-induced alteration in adipocyte lipid storage. MC medium and IL-1β significantly reduced gene expression and protein abundance of insulin signaling molecules, including insulin receptor substrate-1, phosphoinositide 3-kinase p85α, and glucose transporter 4 and phosphorylation of Akt. In contrast, the expression and release of the proinflammatory markers, including IL-6, IL-8, monocyte chemotactic protein-1, and chemokine (C-C motif) ligand 5 by adipocytes were markedly increased. These changes were significantly reduced by blocking IL-1β activity, its receptor binding, or its production by macrophages. MC medium-inhibited expression of the adipogenic factors and -stimulated lipolysis was also blunted with IL-1β neutralization. We conclude that IL-1β mediates, at least in part, the effect of macrophages on insulin signaling and proinflammatory response in human adipocytes. Blocking IL-1β could be beneficial for preventing obesity-associated insulin resistance and inflammation in human adipose tissue.
Collapse
MESH Headings
- Adipocytes, White/cytology
- Adipocytes, White/drug effects
- Adipocytes, White/immunology
- Adipocytes, White/metabolism
- Antibodies, Neutralizing/pharmacology
- Caspase 1/chemistry
- Caspase 1/metabolism
- Caspase Inhibitors/pharmacology
- Cell Communication
- Cell Line
- Cells, Cultured
- Culture Media, Conditioned/chemistry
- Culture Media, Conditioned/metabolism
- Gene Expression Regulation/drug effects
- Humans
- Hypoglycemic Agents/pharmacology
- Insulin/pharmacology
- Insulin Resistance
- Interleukin 1 Receptor Antagonist Protein/genetics
- Interleukin 1 Receptor Antagonist Protein/metabolism
- Interleukin-1beta/antagonists & inhibitors
- Interleukin-1beta/metabolism
- Lipolysis/drug effects
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Receptors, Interleukin-1/agonists
- Receptors, Interleukin-1/antagonists & inhibitors
- Receptors, Interleukin-1/metabolism
- Recombinant Proteins/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Dan Gao
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Mohamed Madi
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Cherlyn Ding
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Matthew Fok
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Thomas Steele
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Ford
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Leif Hunter
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Chen Bing
- Department of Obesity and Endocrinology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
38
|
Lasram MM, Bouzid K, Douib IB, Annabi A, El Elj N, El Fazaa S, Abdelmoula J, Gharbi N. Lipid metabolism disturbances contribute to insulin resistance and decrease insulin sensitivity by malathion exposure in Wistar rat. Drug Chem Toxicol 2014; 38:227-34. [DOI: 10.3109/01480545.2014.933348] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
39
|
Kamel MA, Helmy MH, Hanafi MY, Mahmoud SA, Abo Elfetooh H. Impaired peripheral glucose sensing in F1 offspring of diabetic pregnancy. J Physiol Biochem 2014; 70:685-99. [PMID: 24895245 DOI: 10.1007/s13105-014-0338-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 05/19/2014] [Indexed: 01/15/2023]
Abstract
Maternal diabetes can induce permanent changes in glucose homeostasis that can occur pre- and post-natal and leads to type 2 diabetes in adulthood. This study aimed to investigate the effect of maternal diabetes on the F1 offspring peripheral glucose sensing and mitochondrial biogenesis in an attempt to clarify the mechanism of diabetogenic programming. Two groups of female Wistar rats were used (diabetic and control); diabetes was neonatally induced by STZ injection to 5-day old rats. After the pregnancy and delivery, the offspring were weaned to control diet or high-caloric (HCD) diet and followed up for 30 weeks. Every 5 weeks, OGTT was constructed, and serum and tissues were obtained for the assessment of mTFA, mtDNA, UCP2, insulin receptor (IR), phospho-insulin receptor (phospho-IR), and GLUT4. The result indicated impaired glucose tolerance (IGT) and insulin resistance in the offspring under control diet at the 15th week of age and thereafter while those offspring under HCD showed IGT at 10th week, and diabetes was evidenced at the 25th week of age. This defect in glucose metabolism was preceded by impairment in the phosphorylation of IR and decreased IR and Glut4 that cause impaired glucose sensing together with inhibited mitochondrial biogenesis in muscle and adipose tissues. This study indicated that maternal diabetes caused impaired glucose sensing and insulin resistance in the peripheral tissues and caused change in the expression of genes involved in mitochondrial biogenesis and function. Post-natal feeding with HCD may accelerate these changes. Male F1 offspring appears to be more sensitive than females for fetal programming of T2D.
Collapse
MESH Headings
- Adipose Tissue/metabolism
- Animals
- Animals, Newborn
- DNA, Mitochondrial/genetics
- DNA, Mitochondrial/metabolism
- Diabetes Mellitus, Experimental/etiology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes, Gestational/metabolism
- Diet
- Energy Intake
- Female
- Gene Expression
- Glucose/metabolism
- Glucose Tolerance Test
- Glucose Transporter Type 4/metabolism
- Insulin Resistance
- Ion Channels/genetics
- Male
- Mitochondria/metabolism
- Mitochondrial Proteins/genetics
- Muscle, Skeletal/metabolism
- Pregnancy
- Pregnancy in Diabetics/metabolism
- Prenatal Exposure Delayed Effects/etiology
- Prenatal Exposure Delayed Effects/genetics
- Prenatal Exposure Delayed Effects/metabolism
- Rats
- Rats, Wistar
- Receptor, Insulin/metabolism
- Uncoupling Protein 2
Collapse
Affiliation(s)
- Maher A Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, 165 El-Horreya Street, Alexandria, Egypt,
| | | | | | | | | |
Collapse
|
40
|
Targeting mitochondria as therapeutic strategy for metabolic disorders. ScientificWorldJournal 2014; 2014:604685. [PMID: 24757426 PMCID: PMC3976884 DOI: 10.1155/2014/604685] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 02/12/2014] [Indexed: 12/25/2022] Open
Abstract
Mitochondria are critical regulator of cell metabolism; thus, mitochondrial dysfunction is associated with many metabolic disorders. Defects in oxidative phosphorylation, ROS production, or mtDNA mutations are the main causes of mitochondrial dysfunction in many pathological conditions such as IR/diabetes, metabolic syndrome, cardiovascular diseases, and cancer. Thus, targeting mitochondria has been proposed as therapeutic approach for these conditions, leading to the development of small molecules to be tested in the clinical scenario. Here we discuss therapeutic interventions to treat mitochondrial dysfunction associated with two major metabolic disorders, metabolic syndrome, and cancer. Finally, novel mechanisms of regulation of mitochondrial function are discussed, which open new scenarios for mitochondria targeting.
Collapse
|
41
|
Lim HH, Yang SJ, Kim Y, Lee M, Lim Y. Combined treatment of mulberry leaf and fruit extract ameliorates obesity-related inflammation and oxidative stress in high fat diet-induced obese mice. J Med Food 2014; 16:673-80. [PMID: 23957352 DOI: 10.1089/jmf.2012.2582] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The aim of this study was to investigate whether a combined treatment of mulberry leaf extract (MLE) and mulberry fruit extract (MFE) was effective for improving obesity and obesity-related inflammation and oxidative stress in high fat (HF) diet-induced obese mice. After obesity was induced by HF diet for 9 weeks, the mice were divided into eight groups: (1) lean control, (2) HF diet-induced obese control, (3) 1:1 ratio of MLE and MFE at doses of 200 (L1:1), (4) 500 (M1:1), and (5) 1000 (H1:1) mg/kg per day, and (6) 2:1 ratio of MLE and MFE at doses of 200 (L2:1), (7) 500 (M2:1), and (8) 1000 (H2:1) mg/kg per day. All six combined treatments significantly lowered body weight gain, plasma triglycerides, and lipid peroxidation levels after the 12-week treatment period. Additionally, all combined treatments suppressed hepatic fat accumulation and reduced epididymal adipocyte size. These improvements were accompanied by decreases in protein levels of proinflammatory markers (tumor necrosis factor-alpha, C-reactive protein, interleukin-1, inducible nitric oxide synthase, and phospho-nuclear factor-kappa B inhibitor alpha) and oxidative stress markers (heme oxygenase-1 and manganese superoxide dismutase). M2:1 was the most effective ratio and dose for the improvements in obesity, inflammation, and oxidative stress. These results demonstrate that a combined MLE and MFE treatment ameliorated obesity and obesity-related metabolic stressors and suggest that it can be used as a means to prevent and/or treat obesity.
Collapse
Affiliation(s)
- Hyun Hwa Lim
- Department of Food and Nutrition, Kyung Hee University, Seoul, Korea
| | | | | | | | | |
Collapse
|
42
|
Nicholas LM, Rattanatray L, MacLaughlin SM, Ozanne SE, Kleemann DO, Walker SK, Morrison JL, Zhang S, Muhlhäusler BS, Martin-Gronert MS, McMillen IC. Differential effects of maternal obesity and weight loss in the periconceptional period on the epigenetic regulation of hepatic insulin-signaling pathways in the offspring. FASEB J 2013; 27:3786-96. [PMID: 23729590 DOI: 10.1096/fj.13-227918] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Our aim was to determine the effect of exposure to maternal obesity or to maternal weight loss around conception on the programming of hepatic insulin signaling in the offspring. We used an embryo transfer model in sheep to investigate the effects of exposure to either maternal obesity or to weight loss in normal and obese mothers preceding and for 1 wk after conception on the expression of hepatic insulin-signaling and gluconeogenic factors and key miRNAs involved in insulin signaling in the offspring. We found that exposure to maternal obesity resulted in increased hepatic miR-29b (P<0.05), miR-103 (P<0.01), and miR-107 (P<0.05) expression, a decrease in IR (P<0.05), phopsho-Akt (P<0.01), and phospho-FoxO1 (P<0.01) abundance, and a paradoxical decrease in 11βHSD1 (P<0.05), PEPCK-C (P<0.01), and PEPCK-M (P<0.05) expression in lambs. These changes were ablated by a period of moderate dietary restriction imposed during the periconceptional period. Maternal dietary restriction alone also resulted in decreased abundance of a separate subset of hepatic insulin-signaling molecules, namely, IRS1 (P<0.05), PDK1 (P<0.01), phospho-PDK1 (P<0.05), and aPKCζ (P<0.05) and in decreased PEPCK-C (P<0.01) and G6Pase (P<0.01) expression in the lamb. Our findings highlight the sensitivity of the epigenome to maternal nutrition around conception and the need for dietary interventions that maximize metabolic benefits and minimize metabolic costs for the next generation.
Collapse
Affiliation(s)
- Lisa M Nicholas
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Obanda DN, Cefalu WT. Modulation of cellular insulin signaling and PTP1B effects by lipid metabolites in skeletal muscle cells. J Nutr Biochem 2013; 24:1529-37. [PMID: 23481236 DOI: 10.1016/j.jnutbio.2012.12.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 11/07/2012] [Accepted: 12/21/2012] [Indexed: 12/22/2022]
Abstract
Normal glucose regulation is achieved by having adequate insulin secretion and effective glucose uptake/disposal. Excess lipids in peripheral tissues - skeletal muscle, liver and adipose tissue - may attenuate insulin signaling through the protein kinase B (AKt) pathway and up-regulate protein tyrosine phosphatase 1B (PTP1B), a negative regulator of insulin signaling. We studied accumulation of lipid metabolites [triglycerides (TAGs), diglycerides (DAGs)] and ceramides in relation to insulin signaling and expression and phosphorylation of PTP1B by preincubating rat skeletal muscle cells (L6 myotubes) with three saturated and three unsaturated free fatty acids (FFAs) (200 μM). Cells were also evaluated in the presence of wortmannin, an inhibitor of phosphatidylinositol 3-kinases and thus AKt (0-100 nM). Unsaturated FFAs increased DAGs, TAGs and PTP1B expression significantly, but cells remained insulin sensitive as assessed by robust AKt and PTP1B phosphorylation at serine (Ser) 50, Ser 398 and tyrosine 152. Saturated palmitic and stearic acids increased ceramides, up-regulated PTP1B, and had AKt and PTP1B phosphorylation at Ser 50 impaired. We show a significant correlation between phosphorylation levels of AKt and of PTP1B at Ser 50 (R(2)=0.84, P<.05). The same was observed with increasing wortmannin dose (R(2)=0.73, P<.05). Only FFAs that increased ceramides caused impairment of AKt and PTP1B phosphorylation at Ser 50. PTP1B overexpression in the presence of excess lipids may not directly cause insulin resistance unless it is accompanied by decreased PTP1B phosphorylation. A clear relationship between PTP1B phosphorylation levels at Ser 50 and its negative effect on insulin signaling is shown.
Collapse
Affiliation(s)
- Diana N Obanda
- Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA
| | | |
Collapse
|
44
|
Tack CJ, Stienstra R, Joosten LAB, Netea MG. Inflammation links excess fat to insulin resistance: the role of the interleukin-1 family. Immunol Rev 2013; 249:239-52. [PMID: 22889226 DOI: 10.1111/j.1600-065x.2012.01145.x] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A growing body of evidence suggests that cytokines of the interleukin-1 (IL-1) family, particularly IL-1β but also IL-1Ra and IL-18, are involved in obesity-associated inflammation. IL-1β is produced via cleavage of pro-IL-1β by caspase-1, which in turn is activated by a multiprotein complex called the inflammasome. The components of the NLRP3 inflammasome are involved in sensing obesity-associated danger signals, both in mice and in human (obese) subjects, with caspase-1 seemingly the most crucial regulator. Autophagy is upregulated in obesity and may function as a mechanism to control IL-1β gene expression in adipose tissue to mitigate chronic inflammation. All these mechanisms are operative in human adipose tissue and appear to be more pronounced in human visceral compared to subcutaneous tissue. In animal studies, blocking caspase-1 activity results in decreased weight gain, decreased inflammation, and improved insulin sensitivity. Human intervention studies with IL-1Ra (anakinra) have reported beneficial effects in patients with diabetes, yet without significant changes in insulin sensitivity. Clearly, the IL-1 family of cytokines, especially IL-1β, plays an important role in obesity-associated inflammation and insulin resistance and may represent a therapeutic target to reverse the detrimental metabolic consequences of obesity.
Collapse
Affiliation(s)
- Cees J Tack
- Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|
45
|
Alpha-lipoic acid induces adipose triglyceride lipase expression and decreases intracellular lipid accumulation in HepG2 cells. Eur J Pharmacol 2012; 692:10-8. [DOI: 10.1016/j.ejphar.2012.07.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 07/05/2012] [Accepted: 07/07/2012] [Indexed: 01/14/2023]
|
46
|
Winhofer Y, Krššák M, Janković D, Anderwald CH, Reiter G, Hofer A, Trattnig S, Luger A, Krebs M. Short-term hyperinsulinemia and hyperglycemia increase myocardial lipid content in normal subjects. Diabetes 2012; 61:1210-6. [PMID: 22396203 PMCID: PMC3331780 DOI: 10.2337/db11-1275] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Increased myocardial lipid content (MYCL) recently has been linked to the development of cardiomyopathy in diabetes. In contrast to steatosis in skeletal muscle and liver, previous investigations could not confirm a link between MYCL and insulin resistance. Thus, we hypothesized that cardiac steatosis might develop against the background of the metabolic environment typical for prediabetes and early type 2 diabetes: combined hyperglycemia and hyperinsulinemia. Therefore, we aimed to prove the principle that acute hyperglycemia (during a 6-h clamp) affects MYCL and function (assessed by (1)H magnetic resonance spectroscopy and imaging) in healthy subjects (female subjects: n = 8, male subjects: n = 10; aged 28 ± 5 years; BMI 22.4 ± 2.6 kg/m(2)). Combined hyperglycemia (202.0 ± 10.6 mg/dL) and hyperinsulinemia (110.6 ± 59.0 μU/mL) were, despite insulin-mediated suppression of free fatty acids, associated with a 34.4% increase in MYCL (baseline: 0.20 ± 0.17%, clamp: 0.26 ± 0.22% of water signal; P = 0.0009), which was positively correlated with the area under the curve of insulin (R = 0.59, P = 0.009) and C-peptide (R = 0.81, P < 0.0001) during the clamp. Furthermore, an increase in ejection fraction (P < 0.0001) and a decrease in end-systolic volume (P = 0.0002) were observed, which also were correlated with hyperinsulinemia. Based on our findings, we conclude that combined hyperglycemia and hyperinsulinemia induce short-term myocardial lipid accumulation and alterations in myocardial function in normal subjects, indicating that these alterations might be directly responsible for cardiac steatosis in metabolic diseases.
Collapse
Affiliation(s)
- Yvonne Winhofer
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Martin Krššák
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Draženka Janković
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christian-Heinz Anderwald
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Metabolic Unit, Institute of Biomedical Engineering-National Research Council, Padova, Italy
- Medical Direction, Agathenhof, Specialized Hospital for Metabolic Diseases, Micheldorf, Austria
| | | | - Astrid Hofer
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Siegfried Trattnig
- Department of Radiodiagnostics, Centre of Excellence High-Field Magnetic Resonance, Medical University of Vienna, Vienna, Austria
| | - Anton Luger
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Krebs
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Corresponding author: Michael Krebs,
| |
Collapse
|
47
|
Zhang HM, Dang H, Kamat A, Yeh CK, Zhang BX. Geldanamycin derivative ameliorates high fat diet-induced renal failure in diabetes. PLoS One 2012; 7:e32746. [PMID: 22412919 PMCID: PMC3295767 DOI: 10.1371/journal.pone.0032746] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Accepted: 02/01/2012] [Indexed: 11/25/2022] Open
Abstract
Diabetic nephropathy is a serious complication of longstanding diabetes and its pathogenesis remains unclear. Oxidative stress may play a critical role in the pathogenesis and progression of diabetic nephropathy. Our previous studies have demonstrated that polyunsaturated fatty acids (PUFA) induce peroxynitrite generation in primary human kidney mesangial cells and heat shock protein 90β1 (hsp90β1) is indispensable for the PUFA action. Here we investigated the effects of high fat diet (HFD) on kidney function and structure of db/db mice, a widely used rodent model of type 2 diabetes. Our results indicated that HFD dramatically increased the 24 h-urine output and worsened albuminuria in db/db mice. Discontinuation of HFD reversed the exacerbated albuminuria but not the increased urine output. Prolonged HFD feeding resulted in early death of db/db mice, which was associated with oliguria and anuria. Treatment with the geldanamycin derivative, 17-(dimethylaminoehtylamino)-17-demethoxygeldanamycin (17-DMAG), an hsp90 inhibitor, preserved kidney function, and ameliorated glomerular and tubular damage by HFD. 17-DMAG also significantly extended survival of the animals and protected them from the high mortality associated with renal failure. The benefit effect of 17-DMAG on renal function and structure was associated with a decreased level of kidney nitrotyrosine and a diminished kidney mitochondrial Ca2+ efflux in HFD-fed db/db mice. These results suggest that hsp90β1 is a potential target for the treatment of nephropathy and renal failure in diabetes.
Collapse
Affiliation(s)
- Hong-Mei Zhang
- Department of Clinical Oncology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- Department of Medicine, Health Science Center, University of Texas, San Antonio, Texas, United States of America
| | - Howard Dang
- Department of Comprehensive Dentistry, Health Science Center, University of Texas, San Antonio, Texas, United States of America
| | - Amrita Kamat
- Department of Medicine, Health Science Center, University of Texas, San Antonio, Texas, United States of America
- Audie L. Murphy Division, Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
| | - Chih-Ko Yeh
- Department of Comprehensive Dentistry, Health Science Center, University of Texas, San Antonio, Texas, United States of America
- Audie L. Murphy Division, Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
| | - Bin-Xian Zhang
- Department of Medicine, Health Science Center, University of Texas, San Antonio, Texas, United States of America
- Department of Comprehensive Dentistry, Health Science Center, University of Texas, San Antonio, Texas, United States of America
- Audie L. Murphy Division, Geriatric Research, Education and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
48
|
Martins AR, Nachbar RT, Gorjao R, Vinolo MA, Festuccia WT, Lambertucci RH, Cury-Boaventura MF, Silveira LR, Curi R, Hirabara SM. Mechanisms underlying skeletal muscle insulin resistance induced by fatty acids: importance of the mitochondrial function. Lipids Health Dis 2012; 11:30. [PMID: 22360800 PMCID: PMC3312873 DOI: 10.1186/1476-511x-11-30] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 02/23/2012] [Indexed: 01/06/2023] Open
Abstract
Insulin resistance condition is associated to the development of several syndromes, such as obesity, type 2 diabetes mellitus and metabolic syndrome. Although the factors linking insulin resistance to these syndromes are not precisely defined yet, evidence suggests that the elevated plasma free fatty acid (FFA) level plays an important role in the development of skeletal muscle insulin resistance. Accordantly, in vivo and in vitro exposure of skeletal muscle and myocytes to physiological concentrations of saturated fatty acids is associated with insulin resistance condition. Several mechanisms have been postulated to account for fatty acids-induced muscle insulin resistance, including Randle cycle, oxidative stress, inflammation and mitochondrial dysfunction. Here we reviewed experimental evidence supporting the involvement of each of these propositions in the development of skeletal muscle insulin resistance induced by saturated fatty acids and propose an integrative model placing mitochondrial dysfunction as an important and common factor to the other mechanisms.
Collapse
Affiliation(s)
- Amanda R Martins
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 1524, Butantã, São Paulo, SP, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Cha HN, Song SE, Kim YW, Kim JY, Won KC, Park SY. Lack of inducible nitric oxide synthase prevents lipid-induced skeletal muscle insulin resistance without attenuating cytokine level. J Pharmacol Sci 2012; 117:77-86. [PMID: 22001626 DOI: 10.1254/jphs.11093fp] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
Abstract
We examined whether deletion of inducible nitric oxide synthase (iNOS) could prevent lipid infusion-induced insulin resistance in iNOS-knockout and wild-type mice with the in vivo euglycemic-hyperinsulinemic clamp technique. Plasma NO metabolites were increased in lipid-infused wild-type mice, while they were not increased in iNOS-knockout mice. Plasma tumor necrosis factor-α levels were increased in both wild-type and iNOS-knockout by lipid-infusion. Lipid infusion reduced glucose infusion rate (GIR) and whole body glucose uptake in wild-type mice, whereas iNOS-knockout mice displayed comparable GIR and whole body glucose uptake compared with the control. In the gastrocnemius, lipid infusion decreased glucose uptake and glycolysis that were accompanied with increased phosphorylation of c-Jun N-terminal kinase and reduced phosphorylation of phosphoinositide 3-kinases and serine/threonine kinase Akt. However, lipid infusion did not affect glucose uptake or phosphorylation of these proteins in iNOS-knockout mice. The mRNA levels of inflammatory cytokines were also increased in the gastrocnemis of wild-type and iNOS-knockout mice by lipid infusion. Nitrotyrosine level in the gastrocnemius was increased in lipid-infused wild-type mice but it was not increased in iNOS-knockout mice. These results suggest that lack of iNOS prevents lipid infusion-induced skeletal muscle insulin resistance without attenuating cytokine levels.
Collapse
Affiliation(s)
- Hye-Na Cha
- Department of Physiology, College of Medicine, Yeungnam University, South Korea
| | | | | | | | | | | |
Collapse
|
50
|
Snogdal LS, Wod M, Grarup N, Vestmar M, Sparsø T, Jørgensen T, Lauritzen T, Beck-Nielsen H, Henriksen JE, Pedersen O, Hansen T, Højlund K. Common variation in oxidative phosphorylation genes is not a major cause of insulin resistance or type 2 diabetes. Diabetologia 2012; 55:340-8. [PMID: 22095239 DOI: 10.1007/s00125-011-2377-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Accepted: 10/25/2011] [Indexed: 10/15/2022]
Abstract
AIMS/HYPOTHESIS There is substantial evidence that mitochondrial dysfunction is linked to insulin resistance and is present in several tissues relevant to the pathogenesis of type 2 diabetes. Here, we examined whether common variation in genes involved in oxidative phosphorylation (OxPhos) contributes to type 2 diabetes susceptibility or influences diabetes-related metabolic traits. METHODS OxPhos gene variants (n = 10) that had been nominally associated (p < 0.01) with type 2 diabetes in a recent genome-wide meta-analysis (n = 10,108) were selected for follow-up in 3,599 type 2 diabetic and 4,956 glucose-tolerant Danish individuals. A meta-analysis of these variants was performed in 11,729 type 2 diabetic patients and 43,943 non-diabetic individuals. The impact on OGTT-derived metabolic traits was evaluated in 5,869 treatment-naive individuals from the Danish Inter99 study. RESULTS The minor alleles of COX10 rs9915302 (p = 0.02) and COX5B rs1466100 (p = 0.005) showed nominal association with type 2 diabetes in our Danish cohort. However, in the meta-analysis, none of the investigated variants showed a robust association with type 2 diabetes after correction for multiple testing. Among the alleles potentially associated with type 2 diabetes, none negatively influenced surrogate markers of insulin sensitivity in non-diabetic participants, while the minor alleles of UQCRC1 rs2228561 and COX10 rs10521253 showed a weak (p < 0.01 to p < 0.05) negative influence on indices of glucose-stimulated insulin secretion. CONCLUSIONS/INTERPRETATION We cannot rule out the possibility that common variants in or near OxPhos genes may influence beta cell function in non-diabetic individuals. However, our quantitative trait studies and a sufficiently large meta-analysis indicate that common variation in proximity to the examined OxPhos genes is not a major cause of insulin resistance or type 2 diabetes.
Collapse
Affiliation(s)
- L S Snogdal
- Diabetes Research Centre, Department of Endocrinology, Odense University Hospital, Kløvervænget 6, 4th Floor, 5000 Odense, Denmark
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|