1
|
Wu M, Yan C, Peng H, Wan J, Li R, Ye X, Zhang H, Ding S. Adsorption of dibutyl phthalate by multi-walled carbon nanotubes aggravates hemogram, liver and kidney damage in rats. Food Chem Toxicol 2025; 202:115499. [PMID: 40324677 DOI: 10.1016/j.fct.2025.115499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Studies have shown that environmental pollutants, DBP and MWCNTs, are physiologically toxic. To investigate whether MWCNTs with adsorbed DBP would aggravate this toxicity, we exposed rats to this combination via tail vein injection for two weeks. The results showed the numbers of total WBCs, LYMs, MONs, GRAs, RBCs, PLTs were significantly higher, decrease in GSH, and increase in IL-6, TNF-α, and IgE. Tissue sections revealed hepatic steatosis, sinusoidal sludge, and dilated glomerular capillaries filled with erythrocytes. Blood glucose regulation was impaired, and serum TP, ALT, Cr, and BUN levels were elevated. GO and KEGG analyses showed DEPs were mainly enriched in the catabolism of lipids and proteins, hepatobiliary system development, acute inflammatory response, and metabolic pathways. These findings demonstrate that MWCNTs with adsorbed DBP are more toxic, inducing inflammatory factor release and abnormal metabolic protein expression in the liver and kidney through oxidative damage.
Collapse
Affiliation(s)
- Min Wu
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Chao Yan
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Haiyan Peng
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Jian Wan
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Ren Li
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Xin Ye
- Sichuan Province Key Laboratory for Brewing Science and Technology, College of Bioengineering, Sichuan University of Science & Engineering, Yibin, 644000, China
| | - Hongmao Zhang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China.
| | - Shumao Ding
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China.
| |
Collapse
|
2
|
Naigeon M, Roulleaux Dugage M, Danlos FX, Boselli L, Jouniaux JM, de Oliveira C, Ferrara R, Duchemann B, Berthot C, Girard L, Flippot R, Albiges L, Farhane S, Saulnier P, Lacroix L, Griscelli F, Roman G, Hulett T, Marabelle A, Cassard L, Besse B, Chaput N. Human virome profiling identified CMV as the major viral driver of a high accumulation of senescent CD8 + T cells in patients with advanced NSCLC. SCIENCE ADVANCES 2023; 9:eadh0708. [PMID: 37939189 PMCID: PMC10631735 DOI: 10.1126/sciadv.adh0708] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 10/05/2023] [Indexed: 11/10/2023]
Abstract
Circulating senescent CD8+ T (T8sen) cells are characterized by a lack of proliferative capacities but retain cytotoxic activity and have been associated to resistance to immunotherapy in patients with advanced non-small cell lung cancer (aNSCLC). We aimed to better characterize T8sen and to determine which factors were associated with their accumulation in patients with aNSCLC. Circulating T8sen cells were characterized by a higher expression of SA-βgal and the transcription factor T-bet, confirming their senescent status. Using whole virome profiling, cytomegalovirus (CMV) was the only virus associated with T8sen. CMV was necessary but not sufficient to explain high accumulation of T8sen (T8senhigh status). In CMV+ patients, the proportion of T8sen cells increased with cancer progression. Last, CMV-induced T8senhigh phenotype but not CMV seropositivity itself was associated with worse progression-free and overall survival in patients treated with anti-PD-(L)1 therapy but not with chemotherapy. Overall, CMV is the unique viral driver of T8sen-driven resistance to anti-PD-(L)1 antibodies in patients with aNSCLC.
Collapse
Affiliation(s)
- Marie Naigeon
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Faculté de Pharmacie, Université Paris-Saclay, Orsay, France
| | - Matthieu Roulleaux Dugage
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
- Service d’Oncologie Médicale, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
| | - François-Xavier Danlos
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015 and Centre d’Investigation Clinique BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France
| | - Lisa Boselli
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
| | - Jean-Mehdi Jouniaux
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
| | - Caroline de Oliveira
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
| | - Roberto Ferrara
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Boris Duchemann
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
- Département d’oncologie thoracique et médicale, Hôpitaux Universitaires Paris Seine-Saint-Denis, Hôpital Avicenne, AP-HP, Bobigny, France
| | - Caroline Berthot
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
| | - Lou Girard
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
- Faculté de Pharmacie, Université Paris-Saclay, Orsay, France
| | - Ronan Flippot
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Laurence Albiges
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Siham Farhane
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015 and Centre d’Investigation Clinique BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France
| | | | - Ludovic Lacroix
- AMMICa, UAR 3655/US23, Gustave Roussy, Villejuif, France
- Département de Biologie Médicale et Pathologie Médicales, Gustave Roussy, Villejuif, France
| | - Frank Griscelli
- Département de Biologie Médicale et Pathologie Médicales, Gustave Roussy, Villejuif, France
| | - Gabriel Roman
- CDI Laboratories Inc., 1 N. Haven Street, Suite B001, Baltimore, MD 21224, USA
| | - Tyler Hulett
- CDI Laboratories Inc., 1 N. Haven Street, Suite B001, Baltimore, MD 21224, USA
| | - Aurélien Marabelle
- Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015 and Centre d’Investigation Clinique BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France
| | - Lydie Cassard
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Département de Médecine Oncologique, Gustave Roussy, Villejuif, France
| | - Nathalie Chaput
- Laboratoire d'Immunomonitoring en Oncologie, INSERM US23, CNRS UMS 3655, Gustave Roussy, Villejuif, France
- Faculté de Pharmacie, Université Paris-Saclay, Orsay, France
| |
Collapse
|
3
|
Denimal D, Monier S, Bouillet B, Vergès B, Duvillard L. High-Density Lipoprotein Alterations in Type 2 Diabetes and Obesity. Metabolites 2023; 13:metabo13020253. [PMID: 36837872 PMCID: PMC9967905 DOI: 10.3390/metabo13020253] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Alterations affecting high-density lipoproteins (HDLs) are one of the various abnormalities observed in dyslipidemia in type 2 diabetes mellitus (T2DM) and obesity. Kinetic studies have demonstrated that the catabolism of HDL particles is accelerated. Both the size and the lipidome and proteome of HDL particles are significantly modified, which likely contributes to some of the functional defects of HDLs. Studies on cholesterol efflux capacity have yielded heterogeneous results, ranging from a defect to an improvement. Several studies indicate that HDLs are less able to inhibit the nuclear factor kappa-B (NF-κB) proinflammatory pathway, and subsequently, the adhesion of monocytes on endothelium and their recruitment into the subendothelial space. In addition, the antioxidative function of HDL particles is diminished, thus facilitating the deleterious effects of oxidized low-density lipoproteins on vasculature. Lastly, the HDL-induced activation of endothelial nitric oxide synthase is less effective in T2DM and metabolic syndrome, contributing to several HDL functional defects, such as an impaired capacity to promote vasodilatation and endothelium repair, and difficulty counteracting the production of reactive oxygen species and inflammation.
Collapse
Affiliation(s)
- Damien Denimal
- INSERM, UMR1231, University of Burgundy, 21000 Dijon, France
- Department of Biochemistry, CHU Dijon Bourgogne, 21000 Dijon, France
- Correspondence:
| | - Serge Monier
- INSERM, UMR1231, University of Burgundy, 21000 Dijon, France
| | - Benjamin Bouillet
- INSERM, UMR1231, University of Burgundy, 21000 Dijon, France
- Department of Endocrinology and Diabetology, CHU Dijon Bourgogne, 21000 Dijon, France
| | - Bruno Vergès
- INSERM, UMR1231, University of Burgundy, 21000 Dijon, France
- Department of Endocrinology and Diabetology, CHU Dijon Bourgogne, 21000 Dijon, France
| | - Laurence Duvillard
- INSERM, UMR1231, University of Burgundy, 21000 Dijon, France
- Department of Biochemistry, CHU Dijon Bourgogne, 21000 Dijon, France
| |
Collapse
|
4
|
Princová J, Salat-Canela C, Daněk P, Marešová A, de Cubas L, Bähler J, Ayté J, Hidalgo E, Převorovský M. Perturbed fatty-acid metabolism is linked to localized chromatin hyperacetylation, increased stress-response gene expression and resistance to oxidative stress. PLoS Genet 2023; 19:e1010582. [PMID: 36626368 PMCID: PMC9870116 DOI: 10.1371/journal.pgen.1010582] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/23/2023] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Oxidative stress is associated with cardiovascular and neurodegenerative diseases, diabetes, cancer, psychiatric disorders and aging. In order to counteract, eliminate and/or adapt to the sources of stress, cells possess elaborate stress-response mechanisms, which also operate at the level of regulating transcription. Interestingly, it is becoming apparent that the metabolic state of the cell and certain metabolites can directly control the epigenetic information and gene expression. In the fission yeast Schizosaccharomyces pombe, the conserved Sty1 stress-activated protein kinase cascade is the main pathway responding to most types of stresses, and regulates the transcription of hundreds of genes via the Atf1 transcription factor. Here we report that fission yeast cells defective in fatty acid synthesis (cbf11, mga2 and ACC/cut6 mutants; FAS inhibition) show increased expression of a subset of stress-response genes. This altered gene expression depends on Sty1-Atf1, the Pap1 transcription factor, and the Gcn5 and Mst1 histone acetyltransferases, is associated with increased acetylation of histone H3 at lysine 9 in the corresponding gene promoters, and results in increased cellular resistance to oxidative stress. We propose that changes in lipid metabolism can regulate the chromatin and transcription of specific stress-response genes, which in turn might help cells to maintain redox homeostasis.
Collapse
Affiliation(s)
- Jarmila Princová
- Laboratory of Microbial Genomics, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Clàudia Salat-Canela
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, C/Dr. Aiguader, Barcelona, Spain
| | - Petr Daněk
- Laboratory of Microbial Genomics, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Anna Marešová
- Laboratory of Microbial Genomics, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Laura de Cubas
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, C/Dr. Aiguader, Barcelona, Spain
| | - Jürg Bähler
- Institute of Healthy Ageing and Department of Genetics, Evolution & Environment, University College London, London, United Kingdom
| | - José Ayté
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, C/Dr. Aiguader, Barcelona, Spain
| | - Elena Hidalgo
- Oxidative Stress and Cell Cycle Group, Universitat Pompeu Fabra, C/Dr. Aiguader, Barcelona, Spain
| | - Martin Převorovský
- Laboratory of Microbial Genomics, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
5
|
Wallace SR, Pagano PJ, Kračun D. MicroRNAs in the Regulation of NADPH Oxidases in Vascular Diabetic and Ischemic Pathologies: A Case for Alternate Inhibitory Strategies? Antioxidants (Basel) 2022; 12:70. [PMID: 36670932 PMCID: PMC9854786 DOI: 10.3390/antiox12010070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/24/2022] [Accepted: 12/27/2022] [Indexed: 12/30/2022] Open
Abstract
Since their discovery in the vasculature, different NADPH oxidase (NOX) isoforms have been associated with numerous complex vascular processes such as endothelial dysfunction, vascular inflammation, arterial remodeling, and dyslipidemia. In turn, these often underlie cardiovascular and metabolic pathologies including diabetes mellitus type II, cardiomyopathy, systemic and pulmonary hypertension and atherosclerosis. Increasing attention has been directed toward miRNA involvement in type II diabetes mellitus and its cardiovascular and metabolic co-morbidities in the search for predictive and stratifying biomarkers and therapeutic targets. Owing to the challenges of generating isoform-selective NOX inhibitors (NOXi), the development of specific NOXis suitable for therapeutic purposes has been hindered. In that vein, differential regulation of specific NOX isoforms by a particular miRNA or combina-tion thereof could at some point become a reasonable approach for therapeutic targeting under some circumstances. Whereas administration of miRNAs chronically, or even acutely, to patients poses its own set of difficulties, miRNA-mediated regulation of NOXs in the vasculature is worth surveying. In this review, a distinct focus on the role of miRNAs in the regulation of NOXs was made in the context of type II diabetes mellitus and ischemic injury models.
Collapse
Affiliation(s)
- Sean R. Wallace
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Patrick J. Pagano
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Damir Kračun
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
6
|
Wang W, Liu L, Shan R, Wang C. Associations between dietary copper intake, general obesity and abdominal obesity risk: A nationwide cohort study in China. Front Nutr 2022; 9:1009721. [PMID: 36466427 PMCID: PMC9716269 DOI: 10.3389/fnut.2022.1009721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/07/2022] [Indexed: 01/03/2024] Open
Abstract
OBJECTIVE Copper plays a crucial role in redox reactions. The aims of this research are to examine the effects of copper consumption on general obesity and abdominal obesity risk. METHODS Overall, data of 13,282 participants were obtained from the China Health and Nutrition Survey (1997-2011). A combination of individual 24-h recall and household survey was used to assess dietary intake. Time-dependent mixed effect Cox regression model treating family as a random effect were used to assess the associations between quintiles of copper intake, general obesity and abdominal obesity risk. Obesity was defined by BMI ≥ 28 kg/m2, and abdominal obesity was defined as waist circumference ≥85 cm in men and ≥80 cm in women. RESULTS During follow-up, 1,073 and 4,583 incident cases of general obesity and abdominal obesity occurred respectively. There were U-shaped associations of dietary copper intakes with general obesity and abdominal obesity (P for nonlinearity <0.001). In the general obesity track, compared with quintile 3 (reference category), participants in the top quintile and bottom quintile showed higher general obesity risk (HR, 2.00; 95%CI: 1.63, 2.45 for the top quintile, HR, 1.34; 95%CI: 1.08, 1.68 for the bottom quintile). In the abdominal obesity track, compared with quintile 3, the top quintile and bottom quintile were also associated with a significantly increased risk of abdominal obesity (HR, 1.68; 95%CI: 1.52, 1.87 for the top quintile, HR, 1.36; 95%CI: 1.22, 1.52 for the bottom quintile). CONCLUSIONS We demonstrated U-shaped associations between dietary copper, general and abdominal obesity risk in Chinese and emphasized the importance of maintaining appropriate copper intake level for the prevention of obesity.
Collapse
Affiliation(s)
- Weiqi Wang
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Lin Liu
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Ruiqi Shan
- Department of Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Changhong Wang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
7
|
A multi-marker integrative analysis reveals benefits and risks of bariatric surgery. Sci Rep 2022; 12:18877. [PMID: 36344536 PMCID: PMC9640526 DOI: 10.1038/s41598-022-23241-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
Bariatric surgery (BS) is an effective intervention for severe obesity and associated comorbidities. Although several studies have addressed the clinical and metabolic effects of BS, an integrative analysis of the complex body response to surgery is still lacking. We conducted a longitudinal data study with 36 patients with severe obesity who were tested before, 6 and 12 months after restrictive BS for more than one hundred blood biomarkers, including clinical, oxidative stress and metabolic markers, peptide mediators and red blood cell membrane lipids. By using a synthetic data-driven modeling based on principal component and correlation analyses, we provided evidence that, besides the early, well-known glucose metabolism- and weight loss-associated beneficial effects of BS, a tardive, weight-independent increase of the hepatic cholesterol metabolism occurs that is associated with potentially detrimental inflammatory and metabolic effects. Canonical correlation analysis indicated that oxidative stress is the most predictive feature of the BS-induced changes of both glucose and lipids metabolism. Our results show the power of multi-level correlation analysis to uncover the network of biological pathways affected by BS. This approach highlighted potential health risks of restrictive BS that are disregarded with the current practice to use weight loss as surrogate of BS success.
Collapse
|
8
|
Heras-Molina A, Núñez Y, Benítez R, Pesántez-Pacheco JL, García-Contreras C, Vázquez-Gómez M, Astiz S, Isabel B, González-Bulnes A, Óvilo C. Hypothalamic transcriptome analysis reveals male-specific differences in molecular pathways related to oxidative phosphorylation between Iberian pig genotypes. PLoS One 2022; 17:e0272775. [PMID: 35972914 PMCID: PMC9380940 DOI: 10.1371/journal.pone.0272775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
The hypothalamus is implicated in controlling feeding and adiposity, besides many other physiological functions, and thus can be of great importance in explaining productive differences between lean and fatty pig breeds. The present study aimed to evaluate the hypothalamic transcriptome of pure Iberian (IBxIB) and Large White x Iberian crossbreds (IBxLW) at 60 days-old, produced in a single maternal environment. Results showed the implication of gender and genotype in the hypothalamic transcriptome, with 51 differentially expressed genes (DEGs) between genotypes and 10 DEGs between genders. Fourteen genotype by sex interactions were found, due to a higher genotype effect on transcriptome found in males. In fact, just 31 DEGs were identified when using only females but 158 using only males. A higher expression of genes related to mitochondrial activity in IBxIB male animals (ND3, ND4, ND5, UQCRC2 and ATP6) was found, which was related to a higher oxidative phosphorylation and greater reactive oxygen species and nitric oxide production. IBxLW male animals showed higher expression of SIRT3 regulator, also related to mitochondrial function. When females were analysed, such differences were not found, since only some differences in genes related to the tricarboxylic acid cycle. Thus, the results indicate a significant effect and interaction of the breed and the sex on the hypothalamic transcriptome at this early age.
Collapse
Affiliation(s)
- Ana Heras-Molina
- Department of Animal Breeding, INIA-CSIC, Madrid, Spain
- Department of Animal Production, Veterinary Faculty, UCM, Madrid, Spain
- * E-mail:
| | - Yolanda Núñez
- Department of Animal Breeding, INIA-CSIC, Madrid, Spain
| | - Rita Benítez
- Department of Animal Breeding, INIA-CSIC, Madrid, Spain
| | - José Luis Pesántez-Pacheco
- Department of Animal Reproduction, INIA-CSIC, Madrid, Spain
- School of Veterinary Medicine and Zootechnics, Faculty of Agricultural Sciences, UC, Cuenca, Ecuador
| | | | - Marta Vázquez-Gómez
- Department of Animal Production, Veterinary Faculty, UCM, Madrid, Spain
- Nutrition and Obesities: Systemic Approaches Research Unit (NutriOmics), INSERM, Sorbonne Université, Paris, France
| | - Susana Astiz
- Department of Animal Reproduction, INIA-CSIC, Madrid, Spain
| | - Beatriz Isabel
- Department of Animal Production, Veterinary Faculty, UCM, Madrid, Spain
| | - Antonio González-Bulnes
- Department of Animal Reproduction, INIA-CSIC, Madrid, Spain
- Department of Animal Production, Veterinary Faculty, UCH-CEU, Valencia, Spain
| | | |
Collapse
|
9
|
Wickramaratne AC, Li L, Hopkins JB, Joachimiak LA, Green CB. The Disordered Amino Terminus of the Circadian Enzyme Nocturnin Modulates Its NADP(H) Phosphatase Activity by Changing Protein Dynamics. Biochemistry 2022; 61:10.1021/acs.biochem.2c00072. [PMID: 35535990 PMCID: PMC9646931 DOI: 10.1021/acs.biochem.2c00072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Endogenous circadian clocks control the rhythmicity of a broad range of behavioral and physiological processes, and this is entrained by the daily fluctuations in light and dark. Nocturnin (Noct) is a rhythmically expressed gene regulated by the circadian clock that belongs to the CCR4 family of endonuclease-exonuclease-phosphatase (EEP) enzymes, and the NOCT protein exhibits phosphatase activity, catalyzing the removal of the 2'-phosphate from NADP(H). In addition to its daily nighttime peak of expression, it is also induced by acute stimuli. Loss of Nocturnin (Noct-/-) in mice results in resistance to high-fat diet-induced obesity, and loss of Noct in HEK293T cells confers a protective effect to oxidative stress. Modeling of the full-length Nocturnin protein reveals a partially structured amino terminus that is disparate from its CCR4 family members. The high sequence conservation of a leucine zipper-like (LZ-like) motif, the only structural element in the amino terminus, highlights the potential importance of this domain in modulating phosphatase activity. In vitro biochemical and biophysical techniques demonstrate that the LZ-like domain within the flexible N-terminus is necessary for preserving the active site cleft in an optimal conformation to promote the efficient turnover of the substrate. This modulation occurs in cis and is pivotal in maintaining the stability and conformational integrity of the enzyme. These new findings suggest an additional layer of modulating the activity of Nocturnin in addition to its rhythmicity to provide fine-tuned control over cellular levels of NADPH.
Collapse
Affiliation(s)
- Anushka C. Wickramaratne
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Li Li
- Center for Alzheimer’s and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jesse B. Hopkins
- Biophysics Collaborative Access Team, Illinois Institute of Technology, Advanced Photon Source, Argonne National Laboratory, Lemont, IL 60439, USA
| | - Lukasz A. Joachimiak
- Center for Alzheimer’s and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carla B. Green
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
10
|
Sanders OD, Rajagopal L, Rajagopal JA. The oxidatively damaged DNA and amyloid-β oligomer hypothesis of Alzheimer's disease. Free Radic Biol Med 2022; 179:403-412. [PMID: 34506904 DOI: 10.1016/j.freeradbiomed.2021.08.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 01/13/2023]
Abstract
The amyloid-β (Aβ) oligomer hypothesis of Alzheimer's disease (AD) still dominates the field, yet the clinical trial evidence does not robustly support it. A falsifiable prediction of the hypothesis is that Aβ oligomer levels should be elevated in the brain regions and at the disease stages where and when neuron death and synaptic protein loss begin and are the most severe, but we review previous evidence to demonstrate that this is not consistently the case. To rescue the Aβ oligomer hypothesis from falsification, we propose the novel ad-hoc hypothesis that the exceptionally vulnerable hippocampus may normally produce Aβ peptides even in healthily aging individuals, and hippocampal oxidatively damaged DNA, pathogen DNA, and metal ions such as zinc may initiate and drive Aβ peptide aggregation into oligomers and spreading, neuron death, synaptic dysfunction, and other aspects of AD neurodegeneration. We highlight additional evidence consistent with the underwhelming efficacy of Aβ oligomer-lowering agents, such as aducanumab, and of antioxidants, such as vitamin E, versus the so far isolated case report that DNase-I treatment for 2 months resulted in a severe AD patient's Mini-Mental State Exam score increasing from 3 to 18, reversing his diagnosis to moderate AD, according to the Mini-Mental State Exam.
Collapse
Affiliation(s)
| | - Lekshmy Rajagopal
- Seven Hills Hospital, Marol Maroshi Rd, Shivaji Nagar JJC, Marol, Andheri East, Mumbai, Maharashtra, 400059, India
| | | |
Collapse
|
11
|
Pathological Role of Reactive Oxygen Species on Female Reproduction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1391:201-220. [PMID: 36472824 DOI: 10.1007/978-3-031-12966-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxidative stress (OS), a clinical predicament characterized by a shift in homeostatic imbalance among prooxidant molecules embracing reactive oxygen species (ROS) and reactive nitrogen species (RNS), along with antioxidant defenses, has been established to play an indispensable part in the pathophysiology of subfertility in both human males and females. ROS are highly reactive oxidizing by-products generated during critical oxygen-consuming processes or aerobic metabolism. A healthy body system has its own course of action to maintain the equilibrium between prooxidants and antioxidants with an efficient defense system to fight against ROS. But when ROS production crosses its threshold, the disturbance in homeostatic balance results in OS. Besides their noxious effects, literature studies have depicted that controlled and adequate ROS concentrations exert physiologic functions, especially that gynecologic OS is an important mediator of conception in females. Yet the impact of ROS on oocytes and reproductive functions still needs a strong attestation for further analysis because the disruption in prooxidant and antioxidant balance leads to abrupt ROS generation initiating multiple reproductive diseases such as polycystic ovary syndrome (PCOS), endometriosis, and unexplained infertility in addition to other impediments in pregnancy such as recurrent pregnancy loss, spontaneous abortion, and preeclampsia. The current article elucidates the skeptical state of affairs created by ROS that influences female fertility.
Collapse
|
12
|
Hurtado-Carneiro V, Dongil P, Pérez-García A, Álvarez E, Sanz C. Preventing Oxidative Stress in the Liver: An Opportunity for GLP-1 and/or PASK. Antioxidants (Basel) 2021; 10:antiox10122028. [PMID: 34943132 PMCID: PMC8698360 DOI: 10.3390/antiox10122028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023] Open
Abstract
The liver’s high metabolic activity and detoxification functions generate reactive oxygen species, mainly through oxidative phosphorylation in the mitochondria of hepatocytes. In contrast, it also has a potent antioxidant mechanism for counterbalancing the oxidant’s effect and relieving oxidative stress. PAS kinase (PASK) is a serine/threonine kinase containing an N-terminal Per-Arnt-Sim (PAS) domain, able to detect redox state. During fasting/feeding changes, PASK regulates the expression and activation of critical liver proteins involved in carbohydrate and lipid metabolism and mitochondrial biogenesis. Interestingly, the functional inactivation of PASK prevents the development of a high-fat diet (HFD)-induced obesity and diabetes. In addition, PASK deficiency alters the activity of other nutrient sensors, such as the AMP-activated protein kinase (AMPK) and the mammalian target of rapamycin (mTOR). In addition to the expression and subcellular localization of nicotinamide-dependent histone deacetylases (SIRTs). This review focuses on the relationship between oxidative stress, PASK, and other nutrient sensors, updating the limited knowledge on the role of PASK in the antioxidant response. We also comment on glucagon-like peptide 1 (GLP-1) and its collaboration with PASK in preventing the damage associated with hepatic oxidative stress. The current knowledge would suggest that PASK inhibition and/or exendin-4 treatment, especially under fasting conditions, could ameliorate disorders associated with excess oxidative stress.
Collapse
Affiliation(s)
- Verónica Hurtado-Carneiro
- Department of Physiology, Faculty of Medicine, Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Complutense University of Madrid, Ciudad Universitaria, 28040 Madrid, Spain
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Complutense University of Madrid, Ciudad Universitaria, 28040 Madrid, Spain; (P.D.); (A.P.-G.); (E.Á.)
- Correspondence:
| | - Pilar Dongil
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Complutense University of Madrid, Ciudad Universitaria, 28040 Madrid, Spain; (P.D.); (A.P.-G.); (E.Á.)
- Department of Cell Biology, Faculty of Medicine, Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Complutense University of Madrid, Ciudad Universitaria, 28040 Madrid, Spain;
| | - Ana Pérez-García
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Complutense University of Madrid, Ciudad Universitaria, 28040 Madrid, Spain; (P.D.); (A.P.-G.); (E.Á.)
- Department of Cell Biology, Faculty of Medicine, Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Complutense University of Madrid, Ciudad Universitaria, 28040 Madrid, Spain;
| | - Elvira Álvarez
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Complutense University of Madrid, Ciudad Universitaria, 28040 Madrid, Spain; (P.D.); (A.P.-G.); (E.Á.)
| | - Carmen Sanz
- Department of Cell Biology, Faculty of Medicine, Institute of Medical Research at the San Carlos Clinic Hospital (IdISSC), Complutense University of Madrid, Ciudad Universitaria, 28040 Madrid, Spain;
| |
Collapse
|
13
|
Gender-Specific Behaviour in Obesity Stages I-II: Imbalance of Aminothiol Status and Adipomyokine Profile in Subjects with Different Insulin Resistance Severity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9713582. [PMID: 34868459 PMCID: PMC8635872 DOI: 10.1155/2021/9713582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/11/2021] [Accepted: 11/11/2021] [Indexed: 12/03/2022]
Abstract
The hyperproduction of oxidative stress and inflammatory biomarkers, which is paralleled by decreased levels of antioxidant and anti-inflammatory mediators, is part of cellular mechanisms that contribute to the disruption of metabolic homeostasis in obesity. Whether gender-specific alterations and gender-restricted associations in these biomarkers underlie the increased cardiometabolic risk in men compared to women is unclear. We enrolled 31 women and 29 men, aged ≥50 and ≤70 years and with body mass index ≥ 30 and <40 kg/m2. We assessed the concentrations of aminothiols (cysteine, homocysteine, and glutathione), expression of oxidant/antioxidant balance, adipomyokines (leptin, adiponectin, myostatin, and interleukin-6), markers of chronic inflammation, and vitamin D, an index of nutritional state, in plasma and serum samples by using HPLC, ELISA, and chemiluminescent immunoassay methods. We measured insulin resistance (IR) by the homeostasis model assessment (HOMA) index. Despite comparable levels of visceral adiposity, IR, and a similar dietary regimen, men showed, with respect to women, higher oxidant concentrations and lower antioxidant levels, which paralleled IR severity. Myostatin levels correlated with prooxidant aminothiols among men only. Gender-specific alterations in aminothiol status and adipomyokine profile and the gender-restricted association between these biomarkers and metabolic derangement are consistent with an increased cardiometabolic risk in men compared to age-matched women with stage I-II obesity. Strict control of redox and inflammatory status, even addressing gender-specific nutritional targets, may be useful to prevent obesity-related metabolic alterations and comorbidities.
Collapse
|
14
|
Kalafati M, Lenz M, Ertaylan G, Arts ICW, Evelo CT, van Greevenbroek MMJ, Blaak EE, Adriaens M, Kutmon M. Assessing the Contribution of Relative Macrophage Frequencies to Subcutaneous Adipose Tissue. Front Nutr 2021; 8:675935. [PMID: 34136521 PMCID: PMC8200404 DOI: 10.3389/fnut.2021.675935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/16/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Macrophages play an important role in regulating adipose tissue function, while their frequencies in adipose tissue vary between individuals. Adipose tissue infiltration by high frequencies of macrophages has been linked to changes in adipokine levels and low-grade inflammation, frequently associated with the progression of obesity. The objective of this project was to assess the contribution of relative macrophage frequencies to the overall subcutaneous adipose tissue gene expression using publicly available datasets. Methods: Seven publicly available microarray gene expression datasets from human subcutaneous adipose tissue biopsies (n = 519) were used together with TissueDecoder to determine the adipose tissue cell-type composition of each sample. We divided the subjects in four groups based on their relative macrophage frequencies. Differential gene expression analysis between the high and low relative macrophage frequencies groups was performed, adjusting for sex and study. Finally, biological processes were identified using pathway enrichment and network analysis. Results: We observed lower frequencies of adipocytes and higher frequencies of adipose stem cells in individuals characterized by high macrophage frequencies. We additionally studied whether, within subcutaneous adipose tissue, interindividual differences in the relative frequencies of macrophages were reflected in transcriptional differences in metabolic and inflammatory pathways. Adipose tissue of individuals with high macrophage frequencies had a higher expression of genes involved in complement activation, chemotaxis, focal adhesion, and oxidative stress. Similarly, we observed a lower expression of genes involved in lipid metabolism, fatty acid synthesis, and oxidation and mitochondrial respiration. Conclusion: We present an approach that combines publicly available subcutaneous adipose tissue gene expression datasets with a deconvolution algorithm to calculate subcutaneous adipose tissue cell-type composition. The results showed the expected increased inflammation gene expression profile accompanied by decreased gene expression in pathways related to lipid metabolism and mitochondrial respiration in subcutaneous adipose tissue in individuals characterized by high macrophage frequencies. This approach demonstrates the hidden strength of reusing publicly available data to gain cell-type-specific insights into adipose tissue function.
Collapse
Affiliation(s)
- Marianthi Kalafati
- Deparment of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Michael Lenz
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Institute of Organismic and Molecular Evolution, Johannes Gutenberg University of Mainz, Mainz, Germany.,Preventive Cardiology and Preventive Medicine-Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gökhan Ertaylan
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Unit Health, Flemish Institute for Technological Research, Antwerp, Belgium
| | - Ilja C W Arts
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Department of Epidemiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Chris T Evelo
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Department of Bioinformatics-BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Marleen M J van Greevenbroek
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | - Ellen E Blaak
- Deparment of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| | - Michiel Adriaens
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - Martina Kutmon
- Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands.,Department of Bioinformatics-BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
15
|
Matsumoto M, Suganuma H, Ozato N, Shimizu S, Katashima M, Katsuragi Y, Mikami T, Itoh K, Nakaji S. Association between Serum Concentration of Carotenoid and Visceral Fat. Nutrients 2021; 13:nu13030912. [PMID: 33799771 PMCID: PMC7999533 DOI: 10.3390/nu13030912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/27/2021] [Accepted: 03/08/2021] [Indexed: 01/17/2023] Open
Abstract
Consumption of fruits and vegetables rich in carotenoids has been widely reported to prevent cardiovascular diseases. However, the relationship between serum carotenoid concentrations and visceral fat area (VFA), which is considered a better predictor of cardiovascular diseases than the body-mass index (BMI) and waist circumference, remains unclear. Therefore, we examined the relationship in healthy individuals in their 20s or older, stratified by sex and age, to compare the relationship between serum carotenoid concentrations and VFA and BMI. The study was conducted on 805 people, the residents in Hirosaki city, Aomori prefecture, who underwent a health checkup. An inverse relationship between serum carotenoid concentrations and VFA and BMI was observed only in women. In addition, the results were independent of the intake of dietary fiber, which is mainly supplied from vegetables as well as carotenoids. This suggests that consumption of a diet rich in carotenoids (especially lutein and beta-carotene) is associated with lower VFA, which is a good predictor of cardiovascular disease, especially in women. This study is the first to comprehensively evaluate the association between serum carotenoid levels and VFA in healthy individuals.
Collapse
Affiliation(s)
- Mai Matsumoto
- Innovation Division, KAGOME CO. LTD., 17 Nishitomiyama, Nasushiobara, Tochigi 329-2762, Japan; (H.S.); (S.S.)
- Correspondence: ; Tel.: +81-80-1581-1874
| | - Hiroyuki Suganuma
- Innovation Division, KAGOME CO. LTD., 17 Nishitomiyama, Nasushiobara, Tochigi 329-2762, Japan; (H.S.); (S.S.)
| | - Naoki Ozato
- Department of Active Life Promotion Sciences, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; (N.O.); (M.K.); (Y.K.)
- Health & Wellness Products Research Laboratories, Kao Corporation, Tokyo 131-8501, Japan
| | - Sunao Shimizu
- Innovation Division, KAGOME CO. LTD., 17 Nishitomiyama, Nasushiobara, Tochigi 329-2762, Japan; (H.S.); (S.S.)
- Department of Vegetable Life Science, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan;
- Innovation Center for Health Promotion, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; (T.M.); (S.N.)
| | - Mitsuhiro Katashima
- Department of Active Life Promotion Sciences, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; (N.O.); (M.K.); (Y.K.)
- Health & Wellness Products Research Laboratories, Kao Corporation, Tokyo 131-8501, Japan
| | - Yoshihisa Katsuragi
- Department of Active Life Promotion Sciences, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; (N.O.); (M.K.); (Y.K.)
- Health & Wellness Products Research Laboratories, Kao Corporation, Tokyo 131-8501, Japan
| | - Tatsuya Mikami
- Innovation Center for Health Promotion, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; (T.M.); (S.N.)
| | - Ken Itoh
- Department of Vegetable Life Science, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan;
- Department of Stress Response Science, Center for Advanced Medical Research, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| | - Shigeyuki Nakaji
- Innovation Center for Health Promotion, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan; (T.M.); (S.N.)
- Department of Social Medicine, Graduate School of Medicine, Hirosaki University, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan
| |
Collapse
|
16
|
Xiao L, Miwa N. Hydrogen Nano-Bubble Water Suppresses ROS Generation, Adipogenesis, and Interleukin-6 Secretion in Hydrogen-Peroxide- or PMA-Stimulated Adipocytes and Three-Dimensional Subcutaneous Adipose Equivalents. Cells 2021; 10:cells10030626. [PMID: 33799840 PMCID: PMC7998368 DOI: 10.3390/cells10030626] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/07/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS)-induced oxidative stress in adipose tissue is associated with inflammation and the development of obesity-related metabolic disorders. The aim of this study is to investigate the effects of hydrogen nano-bubble water (HW) on ROS generation, adipogenesis, and interleukin-6 (IL-6) secretion in hydrogen peroxide (H2O2) or phorbol 12-myristate 13-acetate (PMA)-stimulated OP9 adipocytes, and three-dimensional (3D) subcutaneous adipose equivalents. Nanoparticle tracking analysis showed that fresh HW contains 1.17 × 108/mL of nano-sized hydrogen bubbles. Even after 8 to 13 months of storage, approximately half of the bubbles still remained in the water. CellROX® staining showed that HW could diminish H2O2- or PMA-induced intracellular ROS generation in human keratinocytes HaCaT and OP9 cells. We discovered that PMA could markedly increase lipid accumulation to 180% and IL-6 secretion 2.7-fold in OP9 adipocytes. Similarly, H2O2 (5 µM) also significantly stimulated lipid accumulation in OP9 cells and the 3D adipose equivalents. HW treatment significantly repressed H2O2- or PMA-induced lipid accumulation and IL-6 secretion in OP9 adipocytes and the 3D adipose equivalents. In conclusion, HW showed a possibility of repressing oxidative stress, inflammatory response, and adipogenesis at cellular/tissue levels. It can be used for preventing the development of metabolic disorders amongst obese people.
Collapse
Affiliation(s)
- Li Xiao
- Department of Pharmacology, School of Life Dentistry at Tokyo, Nippon Dental University, Tokyo 102-8159, Japan
- Correspondence: ; Tel.: +81-3-3261-8772
| | - Nobuhiko Miwa
- Faculty of Life Sciences, Prefectural University of Hiroshima, Hiroshima 727-0023, Japan;
| |
Collapse
|
17
|
Loss of α7 nicotinic acetylcholine receptors in GABAergic neurons causes sex-dependent decreases in radial glia-like cell quantity and impairments in cognitive and social behavior. Brain Struct Funct 2021; 226:365-379. [PMID: 33398432 DOI: 10.1007/s00429-020-02179-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022]
Abstract
The dentate gyrus (DG) is a unique brain structure in that neurons can be generated postnatally and integrated within existing circuitry throughout life. The maturation process of these newly generated neurons (granule cells) is modulated by nicotinic acetylcholine receptors (nAChRs) through a variety of mechanisms such as neural stem pool proliferation, cell survival, signal modulation, and dendritic integration. Disrupted nAChR signaling has been implicated in neuropsychiatric and neurodegenerative disorders, potentially via alterations in DG neurogenesis. GABAergic interneurons are known to express nAChRs, predominantly the α7 subtype, and have been shown to shape development, integration, and circuit reorganization of DG granule cells. Therefore, we examined histological and behavioral effects of knocking out α7 nAChRs in GABAergic neurons. Deletion of α7 nAChRs resulted in a reduction of radial glia-like cells within the subgranular zone of the DG and a concomitant trend towards decreased immature neurons, specifically in male mice, as well as sex-dependent changes in several behaviors, including social recognition and spatial learning. Overall, these findings suggest α7 nAChRs expressed in GABAergic neurons play an important role in regulating the adult neural stem cell pool and behavior in a sex-dependent manner. This provides important insight into the mechanisms by which cholinergic dysfunction contributes to the cognitive and behavioral changes associated with neurodevelopmental and neurodegenerative disorders.
Collapse
|
18
|
Long-term effects of early overfeeding and food restriction during puberty on cardiac remodeling in adult rats. J Dev Orig Health Dis 2020; 11:492-498. [PMID: 32524941 DOI: 10.1017/s2040174420000513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nutritional disorders during the perinatal period cause cardiometabolic dysfunction, which is observable in the early overfeeding (EO) experimental model. Therefore, severe caloric restriction has the potential of affecting homeostasis through the same epigenetic mechanisms, and its effects need elucidation. This work aims to determine the impact of food restriction (FR) during puberty in early overfed obese and non-obese animals in adult life. Three days after delivery (PN3), Wistar rats were separated into two groups: normal litter (NL; 9 pups) and small litter (SL; 3 pups). At PN30, some offspring were subjected to FR (50%) until PN60, or maintained with free access to standard chow. NL and SL animals submitted to food restriction (NLFR and SLFR groups) were kept in recovery with free access to standard chow from PN60 until PN120. Body weight and food intake were monitored throughout the experimental period. At PN120 cardiovascular parameters were analyzed and the animals were euthanized for sample collection. SLNF and SLFR offspring were overweight and had increased adiposity. Differences in blood pressure were observed only between obese and non-obese animals. Obese and FR animals have cardiac remodeling showing cardiomyocyte hypertrophy and the presence of interstitial and perivascular fibrosis. FR animals also show increased expression of AT1 and AT2 receptors and of total ERK and p-ERK. The present study showed that EO leads to the obese phenotype and cardiovascular disruptions. Interestingly, we demonstrated that severe FR during puberty leads to cardiac remodeling.
Collapse
|
19
|
McElwain C, McCarthy CM. Investigating mitochondrial dysfunction in gestational diabetes mellitus and elucidating if BMI is a causative mediator. Eur J Obstet Gynecol Reprod Biol 2020; 251:60-65. [PMID: 32480181 DOI: 10.1016/j.ejogrb.2020.04.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/07/2020] [Accepted: 04/15/2020] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Gestational diabetes mellitus (GDM) is defined as any degree of glucose intolerance which is diagnosed during pregnancy and poses considerable health risks for mother and child. Maternal body mass index (BMI) correlates with GDM diagnosis and the pathophysiology of this link may be explained through oxidative stress and mitochondrial dysfunction. In this study we investigate if mitochondrial dysfunction is evident in GDM by measuring cell free mitochondrial DNA concentration and determine if a potential relationship exists between maternal mitochondrial function and GDM diagnosis. STUDY DESIGN Plasma samples were taken at 20 weeks' gestation from women who subsequently developed GDM (n = 44) and matched with women with uncomplicated pregnancies (n = 85) as controls. Control group 1 was matched by maternal age and BMI (n = 41) to GDM cases, while control group 2 was matched by maternal age alone (n = 44). Prediction potential was determined by binary regression analysis. Statistical analysis was performed on SPSS Statistics v25. RESULTS Binary regression analysis showed a statistically significant association between mtDNA concentration and GDM diagnosis (p = 0.032) in GDM cases versus control group 2, indicating that GDM patients have higher circulating mtDNA concentrations relative to healthy control patients. The lack of statistical significance in control group 1 suggests that BMI may be linked to mitochondrial function in GDM patients. CONCLUSION These results demonstrate a potential pathogenic role for mitochondrial dysfunction in GDM, with BMI presenting as a likely physiological mediator.
Collapse
Affiliation(s)
- Colm McElwain
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.
| | - Cathal M McCarthy
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland.
| |
Collapse
|
20
|
Tam BT, Morais JA, Santosa S. Obesity and ageing: Two sides of the same coin. Obes Rev 2020; 21:e12991. [PMID: 32020741 DOI: 10.1111/obr.12991] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/30/2019] [Indexed: 02/06/2023]
Abstract
Conditions and comorbidities of obesity mirror those of ageing and age-related diseases. Obesity and ageing share a similar spectrum of phenotypes such as compromised genomic integrity, impaired mitochondrial function, accumulation of intracellular macromolecules, weakened immunity, shifts in tissue and body composition, and enhanced systemic inflammation. Moreover, it has been shown that obesity reduces life expectancy by 5.8 years in men and 7.1 years in women after the age of 40. Shorter life expectancy could be because obesity holistically accelerates ageing at multiple levels. Besides jeopardizing nuclear DNA and mitochondrial DNA integrity, obesity modifies the DNA methylation pattern, which is associated with epigenetic ageing in different tissues. Additionally, other signs of ageing are seen in individuals with obesity including telomere shortening, systemic inflammation, and functional declines. This review aims to show how obesity and ageing are "two sides of the same coin" through discussing how obesity predisposes an individual to age-related conditions, illness, and disease. We will further demonstrate how the mechanisms that perpetuate the early-onset of chronic diseases in obesity parallel those of ageing.
Collapse
Affiliation(s)
- Bjorn T Tam
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Quebec, Montreal, Canada.,Metabolism, Obesity, and Nutrition Lab, PERFORM Centre, Concordia University, Quebec, Montreal, Canada
| | - Jose A Morais
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Quebec, Montreal, Canada.,Division of Geriatric Medicine and Research Institute, McGill University Health Centre, Quebec, Montreal, Canada
| | - Sylvia Santosa
- Department of Health, Kinesiology, and Applied Physiology, Concordia University, Quebec, Montreal, Canada.,Metabolism, Obesity, and Nutrition Lab, PERFORM Centre, Concordia University, Quebec, Montreal, Canada.,Research Centre, Centre intégré universitarie de santé et de services sociaux du Nord-de-I'Île-de-Montréal, Hôpital du Sacré-Cœur de Monréal (CIUSS-NIM, HSCM), Quebec, Montreal, Canada
| |
Collapse
|
21
|
Anusruti A, Jansen EHJM, Gào X, Xuan Y, Brenner H, Schöttker B. Longitudinal Associations of Body Mass Index, Waist Circumference, and Waist-to-Hip Ratio with Biomarkers of Oxidative Stress in Older Adults: Results of a Large Cohort Study. Obes Facts 2020; 13:66-76. [PMID: 31986512 PMCID: PMC7098284 DOI: 10.1159/000504711] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/10/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND In the literature, obesity is discussed as a determinant of high oxidative stress (OS). Hence, prevention or reduction of obesity could prevent high OS and subsequently serve as a target for "healthy aging." METHODS Diacron's reactive oxygen metabolites test (D-ROM) and total thiol levels (TTL), a marker of antioxidant defense capacity, were measured in 1,734 participants of a population-based cohort study of older adults (age range: 57-83 years) at 2 time points 3 years apart. The longitudinal associations of body mass index, waist-to-hip ratio, and waist circumference with D-ROM and TTL were assessed with multivariable adjusted generalized linear models. Dose-response analyses were conducted with restricted cubic splines. RESULTS D-ROM was not significantly associated with any of the weight measures. On the contrary, TTL showed statistically significant, inverse linear associations with all weight measures. CONCLUSION A healthy body weight seems to be highly relevant for the antioxidative defense capacity of human beings. In contrast, D-ROM levels were independent of the study participant's weight. Clinical trials are needed to corroborate if loss of weight by obese individuals can effectively increase TTL and subsequently also life expectancy.
Collapse
Affiliation(s)
- Ankita Anusruti
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Network Aging Research, Heidelberg University, Heidelberg, Germany
| | - Eugène H J M Jansen
- Center for Health Protection, National Institute of Public Health and the Environment, Bilthoven, The Netherlands
| | - Xīn Gào
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Network Aging Research, Heidelberg University, Heidelberg, Germany
| | - Yang Xuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Network Aging Research, Heidelberg University, Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Network Aging Research, Heidelberg University, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Ben Schöttker
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany,
- Network Aging Research, Heidelberg University, Heidelberg, Germany,
| |
Collapse
|
22
|
McElwain CJ, Tuboly E, McCarthy FP, McCarthy CM. Mechanisms of Endothelial Dysfunction in Pre-eclampsia and Gestational Diabetes Mellitus: Windows Into Future Cardiometabolic Health? Front Endocrinol (Lausanne) 2020; 11:655. [PMID: 33042016 PMCID: PMC7516342 DOI: 10.3389/fendo.2020.00655] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Placental insufficiency and adipose tissue dysregulation are postulated to play key roles in the pathophysiology of both pre-eclampsia (PE) and gestational diabetes mellitus (GDM). A dysfunctional release of deleterious signaling motifs can offset an increase in circulating oxidative stressors, pro-inflammatory factors and various cytokines. It has been previously postulated that endothelial dysfunction, instigated by signaling from endocrine organs such as the placenta and adipose tissue, may be a key mediator of the vasculopathy that is evident in both adverse obstetric complications. These signaling pathways also have significant effects on long term maternal cardiometabolic health outcomes, specifically cardiovascular disease, hypertension, and type II diabetes. Recent studies have noted that both PE and GDM are strongly associated with lower maternal flow-mediated dilation, however the exact pathways which link endothelial dysfunction to clinical outcomes in these complications remains in question. The current diagnostic regimen for both PE and GDM lacks specificity and consistency in relation to clinical guidelines. Furthermore, current therapeutic options rely largely on clinical symptom control such as antihypertensives and insulin therapy, rather than that of early intervention or prophylaxis. A better understanding of the pathogenic origin of these obstetric complications will allow for more targeted therapeutic interventions. In this review we will explore the complex signaling relationship between the placenta and adipose tissue in PE and GDM and investigate how these intricate pathways affect maternal endothelial function and, hence, play a role in acute pathophysiology and the development of future chronic maternal health outcomes.
Collapse
Affiliation(s)
- Colm J. McElwain
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
- *Correspondence: Colm J. McElwain
| | - Eszter Tuboly
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, Cork, Ireland
| | - Cathal M. McCarthy
- Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| |
Collapse
|
23
|
DeVallance E, Li Y, Jurczak MJ, Cifuentes-Pagano E, Pagano PJ. The Role of NADPH Oxidases in the Etiology of Obesity and Metabolic Syndrome: Contribution of Individual Isoforms and Cell Biology. Antioxid Redox Signal 2019; 31:687-709. [PMID: 31250671 PMCID: PMC6909742 DOI: 10.1089/ars.2018.7674] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Highly prevalent in Western cultures, obesity, metabolic syndrome, and diabetes increase the risk of cardiovascular morbidity and mortality and cost health care systems billions of dollars annually. At the cellular level, obesity, metabolic syndrome, and diabetes are associated with increased production of reactive oxygen species (ROS). Increased levels of ROS production in key organ systems such as adipose tissue, skeletal muscle, and the vasculature cause disruption of tissue homeostasis, leading to increased morbidity and risk of mortality. More specifically, growing evidence implicates the nicotinamide adenine dinucleotide phosphate oxidase (NOX) enzymes in these pathologies through impairment of insulin signaling, inflammation, and vascular dysfunction. The NOX family of enzymes is a major driver of redox signaling through its production of superoxide anion, hydrogen peroxide, and attendant downstream metabolites acting on redox-sensitive signaling molecules. Recent Advances: The primary goal of this review is to highlight recent advances and survey our present understanding of cell-specific NOX enzyme contributions to metabolic diseases. Critical Issues: However, due to the short half-lives of individual ROS and/or cellular defense systems, radii of ROS diffusion are commonly short, often restricting redox signaling and oxidant stress to localized events. Thus, special emphasis should be placed on cell type and subcellular location of NOX enzymes to better understand their role in the pathophysiology of metabolic diseases. Future Directions: We discuss the targeting of NOX enzymes as potential therapy and bring to light potential emerging areas of NOX research, microparticles and epigenetics, in the context of metabolic disease.
Collapse
Affiliation(s)
- Evan DeVallance
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Pittsburgh Heart, Lung and Blood, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yao Li
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Pittsburgh Heart, Lung and Blood, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Pittsburgh Heart, Lung and Blood, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Patrick J Pagano
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Pittsburgh Heart, Lung and Blood, Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
24
|
Wang N, Ma Y, Liu Z, Liu L, Yang K, Wei Y, Liu Y, Chen X, Sun X, Wen D. Hydroxytyrosol prevents PM 2.5-induced adiposity and insulin resistance by restraining oxidative stress related NF-κB pathway and modulation of gut microbiota in a murine model. Free Radic Biol Med 2019; 141:393-407. [PMID: 31279968 DOI: 10.1016/j.freeradbiomed.2019.07.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/25/2019] [Accepted: 07/03/2019] [Indexed: 12/18/2022]
Abstract
Exposure to fine particular matter (≤2.5 μM, PM2.5) contributes to increased risk of obesity and type 2 diabetes. Hydroxytyrosol (HT), a simple polyphenol found in virgin olive oil, is considered to be beneficial for cardiovascular and metabolic disorders. The current study determined whether HT could improve PM2.5-induced adiposity and insulin resistance (IR), and explored the underlying mechanisms. Fifteen adult female C57BL/6j mice on a chow diet were randomly divided into three groups receiving (1) sterile PBS, (2) PM2.5 suspended in sterile PBS (1 mg/mL) and (3) PM2.5+HT (50 mg/kg/day). PM2.5/PBS exposure was administered by oropharynx instillation every other day and HT supplementation was achieved by gavage every day. Four-week PM2.5 exposure did not affect body weight, but significantly increased visceral fat mass. The abdominal adiposity coincided with adipocyte hypertrophy and proliferation in visceral white adipose tissue (WAT), as well as decreased metabolic activity in brown adipose tissue and subcutaneous WAT. PM2.5 enhanced the oxidative stress by diminishing antioxidant enzyme activities in liver and serum, whereas contents of 4-hydroxynonenal (4-HNE), malondialdehyde (MDA) levels in liver and serum were elevated. These changes were accompanied by macrophage infiltration and activation of NF-κB pathway in the liver. Moreover, PM2.5 exposure led to glucose intolerance and insulin insensitivity, impaired hepatic glycogenesis, and decreased insulin-stimulated Akt phosphorylation in peripheral tissues. Importantly, HT treatment prevented PM2.5-induced visceral adipogenesis, oxidative stress, hepatic inflammation and NF-κB activation, systemic and peripheral IR. In vitro, after HepG2 cells were incubated with PM2.5 (0, 5, 25, 50, 100 and 200 μg/mL), reduced glutathione depletion and 4-HNE, 8-hydroxy-2'-deoxyguanosine, MDA increment in a dose-dependent manner were observed; likewise, insulin-stimulated glucose uptake decreased in a dose-dependent manner. Further, with antioxidant NAC and NF-κB inhibitor PDTC, we confirmed that HT attenuated PM2.5-induced IR through restraining NF-κB activation evoked by oxidative stress. In addition, HT could expand gut microbiota richness, reduce pathogenic bacteria and accommodate the microbial architecture in PM2.5-exposed mice, which were correlated with parameters of adiposity, oxidative stress and glycometabolism. HT could effectively correct imbalanced oxidative stress triggered by PM2.5, in turn ameliorated NF-κB pathway and insulin signaling. Gut microbiota may mediate the actions of HT.
Collapse
Affiliation(s)
- Ningning Wang
- Department of Nutrition & Food Hygiene, School of Public Health, Dalian Medical University, Dalian, Liaoning, PR China.
| | - Yanan Ma
- School of Public Health, China Medical University, Shenyang, Liaoning, PR China.
| | - Zhuoqun Liu
- School of Public Health, Dalian Medical University, Dalian, Liaoning, PR China.
| | - Lei Liu
- School of Public Health, Dalian Medical University, Dalian, Liaoning, PR China.
| | - Keming Yang
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA.
| | - Yaguang Wei
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Yang Liu
- The Institute of Health Science, China Medical University, Shenyang, Liaoning, PR China.
| | - Xin Chen
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, Liaoning, PR China.
| | - Xiance Sun
- Department of Occupational and Environmental Health, School of Public Health, Dalian Medical University, Dalian, Liaoning, PR China.
| | - Deliang Wen
- The Institute of Health Science, China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
25
|
Chen CC, Lee TY, Leu YL, Wang SH. Pigment epithelium-derived factor inhibits adipogenesis in 3T3-L1 adipocytes and protects against high-fat diet-induced obesity and metabolic disorders in mice. Transl Res 2019; 210:26-42. [PMID: 31121128 DOI: 10.1016/j.trsl.2019.04.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 03/23/2019] [Accepted: 04/12/2019] [Indexed: 01/10/2023]
Abstract
Obesity is a major cause of metabolic syndrome and type II diabetes, and it presents with metabolic disorders, such as hyperglycemia, hyperlipidemia, and insulin resistance. Pigment epithelium-derived factor (PEDF), a protein isolated from retinal pigment epithelial cells, has multiple functions, including neuronal protection, antineoplastic effects, and anti-inflammatory activity. The aim of this study is to investigate the antiobesity effects of PEDF. The antiobesity effects of PEDF on fat accumulation, inflammation, energy expenditure, insulin resistance, and obesity-related physiological parameters and protein levels were assessed in high-fat diet (HFD)-induced obese mice in vivo and in 3T3-L1 adipocytes, palmitate (PA)-treated HepG2 cells, and C2C12 myotubes in vitro. In an in vivo assay, PEDF effectively decreased body weight gain, white adipose tissue mass, and inflammation and improved insulin resistance, dyslipidemia, and hyperglycemia in HFD-induced mice. In liver tissue, PEDF decreased lipid accumulation and fibrosis. In an in vitro assay, PEDF diminished the differentiation of 3T3-L1 preadipocytes. We also determined that PEDF promoted lipolysis and prolonged cell cycle progression, through the mTOR-S6K pathway and downstream transcription factors, such as peroxisome proliferator-activated receptor gamma, CCAAT/enhancer-binding protein α (CEBP-α), and CEBP-β. In addition, PEDF decreased reactive oxygen species production in PA-induced HepG2 cells and improved glucose uptake ability in PA-induced HepG2 cells and C2C12 myotubes. In the present study, PEDF protected against HFD-induced obesity and metabolic disorders in mice, inhibited adipogenesis, and improved insulin resistance. These results provide a new potential treatment for obesity in the future.
Collapse
Affiliation(s)
- Chin-Chuan Chen
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan; Tissue Bank, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Ting-Yau Lee
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan; Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu-Huei Wang
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
26
|
Mitochondrial Uncoupling: A Key Controller of Biological Processes in Physiology and Diseases. Cells 2019; 8:cells8080795. [PMID: 31366145 PMCID: PMC6721602 DOI: 10.3390/cells8080795] [Citation(s) in RCA: 293] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022] Open
Abstract
Mitochondrial uncoupling can be defined as a dissociation between mitochondrial membrane potential generation and its use for mitochondria-dependent ATP synthesis. Although this process was originally considered a mitochondrial dysfunction, the identification of UCP-1 as an endogenous physiological uncoupling protein suggests that the process could be involved in many other biological processes. In this review, we first compare the mitochondrial uncoupling agents available in term of mechanistic and non-specific effects. Proteins regulating mitochondrial uncoupling, as well as chemical compounds with uncoupling properties are discussed. Second, we summarize the most recent findings linking mitochondrial uncoupling and other cellular or biological processes, such as bulk and specific autophagy, reactive oxygen species production, protein secretion, cell death, physical exercise, metabolic adaptations in adipose tissue, and cell signaling. Finally, we show how mitochondrial uncoupling could be used to treat several human diseases, such as obesity, cardiovascular diseases, or neurological disorders.
Collapse
|
27
|
Li C, Qu L, Farragher C, Vella A, Zhou B. MicroRNA Regulated Macrophage Activation in Obesity. J Transl Int Med 2019; 7:46-52. [PMID: 31380236 PMCID: PMC6661877 DOI: 10.2478/jtim-2019-0011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Chuan Li
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT, USA
| | - Lili Qu
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT, USA
| | - Cullen Farragher
- College of Liberal Arts and Sciences, University of Connecticut, Storrs, CT, USA
| | - Anthony Vella
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT, USA
| | - Beiyan Zhou
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT, USA
- Institute for Systems Genomics, University of Connecticut, Farmington, CT, USA
| |
Collapse
|
28
|
Häusler N, Heinzer R, Haba-Rubio J, Marques-Vidal P. Does sleep affect weight gain? Assessing subjective sleep and polysomnography measures in a population-based cohort study (CoLaus/HypnoLaus). Sleep 2019; 42:5413663. [DOI: 10.1093/sleep/zsz077] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 02/18/2019] [Indexed: 01/16/2023] Open
Affiliation(s)
- Nadine Häusler
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Raphaël Heinzer
- Center for Investigation and Research in Sleep (CIRS), Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Jose Haba-Rubio
- Center for Investigation and Research in Sleep (CIRS), Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
29
|
Stygar D, Skrzep-Poloczek B, Romuk E, Chełmecka E, Poloczek J, Sawczyn T, Maciarz J, Kukla M, Karcz KW, Jochem J. The influence of high-fat, high-sugar diet and bariatric surgery on HSP70 and HSP90 plasma and liver concentrations in diet-induced obese rats. Cell Stress Chaperones 2019; 24:427-439. [PMID: 30840227 PMCID: PMC6439084 DOI: 10.1007/s12192-019-00976-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/22/2019] [Accepted: 01/30/2019] [Indexed: 12/31/2022] Open
Abstract
Metabolic surgery ameliorates insulin resistance and is associated with long-term, effective weight loss, but the mechanisms involved remain unknown. Here, the duodenal-jejunal omega switch (DJOS) surgery in combination with high-fat, high-carbohydrate diet was performed on diet obese rats and joint effects of bariatric surgery and different dietary patterns on heat shock protein 70 (HSP70) and HSP90 plasma and liver concentrations were measured. We found that plasma and liver levels of HSP70 were lower after DJOS surgery in comparison to the control in the groups of animals kept on control diet (CD) and high-fat, high-sugar diet (HFS) but the postoperative change of the diet led to the increase in HSP70 in plasma and liver concentration in DJOS-operated animals. A high-calorie meal, rich in carbohydrates and fats, significantly increased circulating levels of HSP90, reducing the normalising effect of DJOS. The HFS diet applied during all stages of the experiment led to the higher levels of liver HSP90 concentration. The combination of CD and DJOS surgery was the most efficient in the lowering of the HSP90 liver concentration. The normalisation of circulating levels and liver concentrations of HSP70 and HSP90 may be achieved in a combination of DJOS procedure with a proper dietary plan.
Collapse
Affiliation(s)
- Dominika Stygar
- Department of Physiology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Katowice, Poland.
| | - Bronisława Skrzep-Poloczek
- Department of Physiology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Ewa Romuk
- Department of Biochemistry, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Elżbieta Chełmecka
- Department of Statistics, Department of Instrumental Analysis, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Katowice, Poland
| | - Jakub Poloczek
- Department of Rehabilitation, 3rd Specialist Hospital in Rybnik, Rybnik, Poland
| | - Tomasz Sawczyn
- Department of Physiology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Justyna Maciarz
- Department of Physiology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Michał Kukla
- Department of Gastroenterology and Hepatology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Konrad W Karcz
- Clinic of General, Visceral, Transplantation and Vascular Surgery, Hospital of the Ludwig Maximilian University, Munich, Germany
| | - Jerzy Jochem
- Department of Physiology, School of Medicine with the Division of Dentistry in Zabrze, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
30
|
Macrophage phenotype and bioenergetics are controlled by oxidized phospholipids identified in lean and obese adipose tissue. Proc Natl Acad Sci U S A 2018; 115:E6254-E6263. [PMID: 29891687 PMCID: PMC6142199 DOI: 10.1073/pnas.1800544115] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Adipose tissue macrophages (ATMs) maintain adipose tissue homeostasis. However, during obesity ATMs become inflammatory, resulting in impaired adipose tissue function. Oxidative stress increases during obesity, which is thought to contribute to adipose tissue inflammation. To date, the connection between oxidative stress and adipose tissue inflammation remain unclear. In this study, we identify two classes of phospholipid oxidation products in lean and obese adipose tissue, which polarize macrophages to an antioxidant or proinflammatory state, respectively. Furthermore, we show that these phospholipids differently affect macrophage cellular metabolism, reflecting the metabolisms of ATMs found in lean and obese adipose tissue. Identification of pathways controlling ATM metabolism will lead to novel therapies for insulin resistance. Adipose tissue macrophages (ATMs) adapt their metabolic phenotype either to maintain lean tissue homeostasis or drive inflammation and insulin resistance in obesity. However, the factors in the adipose tissue microenvironment that control ATM phenotypic polarization and bioenergetics remain unknown. We have recently shown that oxidized phospholipids (OxPL) uniquely regulate gene expression and cellular metabolism in Mox macrophages, but the presence of the Mox phenotype in adipose tissue has not been reported. Here we show, using extracellular flux analysis, that ATMs isolated from lean mice are metabolically inhibited. We identify a unique population of CX3CR1neg/F4/80low ATMs that resemble the Mox (Txnrd1+HO1+) phenotype to be the predominant ATM phenotype in lean adipose tissue. In contrast, ATMs isolated from obese mice had characteristics typical of the M1/M2 (CD11c+CD206+) phenotype with highly activated bioenergetics. Quantifying individual OxPL species in the stromal vascular fraction of murine adipose tissue, using targeted liquid chromatography-mass spectrometry, revealed that high fat diet-induced adipose tissue expansion led to a disproportional increase in full-length over truncated OxPL species. In vitro studies showed that macrophages respond to truncated OxPL species by suppressing bioenergetics and up-regulating antioxidant programs, mimicking the Mox phenotype of ATMs isolated from lean mice. Conversely, full-length OxPL species induce proinflammatory gene expression and an activated bioenergetic profile that mimics ATMs isolated from obese mice. Together, these data identify a redox-regulatory Mox macrophage phenotype to be predominant in lean adipose tissue and demonstrate that individual OxPL species that accumulate in adipose tissue instruct ATMs to adapt their phenotype and bioenergetic profile to either maintain redox homeostasis or to promote inflammation.
Collapse
|
31
|
Goutzourelas N, Orfanou M, Charizanis I, Leon G, Spandidos DA, Kouretas D. GSH levels affect weight loss in individuals with metabolic syndrome and obesity following dietary therapy. Exp Ther Med 2018; 16:635-642. [PMID: 30116319 PMCID: PMC6090313 DOI: 10.3892/etm.2018.6204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/10/2018] [Indexed: 12/19/2022] Open
Abstract
This study examined the effects of redox status markers on metabolic syndrome (MetS) and obesity before and after dietary intervention and exercise for weight loss. A total of 103 adults suffering from MetS and obesity participated in this study and followed a personalized diet plan for 6 months. Body weight, body fat (BF) percentage (BF%), respiratory quotient (RQ) and the redox status markers, reduced glutathione (GSH), thiobarbituric acid reactive substances (TBARS) and protein carbonyls (CARB), were measured twice in each individual, before and after intervention. Dietary intervention resulted in weight loss, a reduction in BF% and a decrease in RQ. The GSH levels were significantly decreased following intervention, while the levels of TBARS and CARB were not affected. Based on the initial GSH levels, the patients were divided into 2 groups as follows: The high GSH group (GSH, >3.5 µmol/g Hb) and the low GSH group (GSH <3.5 µmol/g Hb). Greater weight and BF loss were observed in patients with high GSH levels. It was observed that patients with MetS and obesity with high GSH values responded better to the dietary therapy, exhibiting more significant changes in weight and BF%. This finding underscores the importance of identifying redox status markers, particularly GSH, in obese patients with MetS. Knowing the levels of GSH may aid in developing a better design of an individualized dietary plan for individuals who wish to lose weight.
Collapse
Affiliation(s)
- Nikolaos Goutzourelas
- Department of Biochemistry and Biotechnology, University of Thessaly, 41500 Larissa, Greece.,Eatwalk IKE, 15124 Athens, Greece
| | | | | | | | - Demetrios A Spandidos
- Laboratory of Clinical Virology, University of Crete, Medical School, 71409 Heraklion, Greece
| | - Demetrios Kouretas
- Department of Biochemistry and Biotechnology, University of Thessaly, 41500 Larissa, Greece
| |
Collapse
|
32
|
Campbell JP, Turner JE. Debunking the Myth of Exercise-Induced Immune Suppression: Redefining the Impact of Exercise on Immunological Health Across the Lifespan. Front Immunol 2018; 9:648. [PMID: 29713319 PMCID: PMC5911985 DOI: 10.3389/fimmu.2018.00648] [Citation(s) in RCA: 385] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 03/15/2018] [Indexed: 12/17/2022] Open
Abstract
Epidemiological evidence indicates that regular physical activity and/or frequent structured exercise reduces the incidence of many chronic diseases in older age, including communicable diseases such as viral and bacterial infections, as well as non-communicable diseases such as cancer and chronic inflammatory disorders. Despite the apparent health benefits achieved by leading an active lifestyle, which imply that regular physical activity and frequent exercise enhance immune competency and regulation, the effect of a single bout of exercise on immune function remains a controversial topic. Indeed, to this day, it is perceived by many that a vigorous bout of exercise can temporarily suppress immune function. In the first part of this review, we deconstruct the key pillars which lay the foundation to this theory-referred to as the "open window" hypothesis-and highlight that: (i) limited reliable evidence exists to support the claim that vigorous exercise heightens risk of opportunistic infections; (ii) purported changes to mucosal immunity, namely salivary IgA levels, after exercise do not signpost a period of immune suppression; and (iii) the dramatic reductions to lymphocyte numbers and function 1-2 h after exercise reflects a transient and time-dependent redistribution of immune cells to peripheral tissues, resulting in a heightened state of immune surveillance and immune regulation, as opposed to immune suppression. In the second part of this review, we provide evidence that frequent exercise enhances-rather than suppresses-immune competency, and highlight key findings from human vaccination studies which show heightened responses to bacterial and viral antigens following bouts of exercise. Finally, in the third part of this review, we highlight that regular physical activity and frequent exercise might limit or delay aging of the immune system, providing further evidence that exercise is beneficial for immunological health. In summary, the over-arching aim of this review is to rebalance opinion over the perceived relationships between exercise and immune function. We emphasize that it is a misconception to label any form of acute exercise as immunosuppressive, and, instead, exercise most likely improves immune competency across the lifespan.
Collapse
Affiliation(s)
- John P Campbell
- Department for Health, University of Bath, Bath, United Kingdom
| | - James E Turner
- Department for Health, University of Bath, Bath, United Kingdom
| |
Collapse
|
33
|
Xu R, Chen C, Zhou Y, Zhang X, Wan Y. Fingernail selenium levels in relation to the risk of obesity in Chinese children: A cross-sectional study. Medicine (Baltimore) 2018; 97:e0027. [PMID: 29489649 PMCID: PMC5851749 DOI: 10.1097/md.0000000000010027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Selenium (Se) has been suggested to be beneficial to obesity development. However, limited studies have evaluated the association between Se and childhood obesity and the findings are inconsistent.The aim of this study was to examine the association of Se levels with obesity in children in a cross-sectional study.A total of 62 obese (21 girls) and 65 normal-weight children (27 girls) aged 7 to 13 years were recruited in Shanghai, China. Obesity was defined as body mass index (BMI) ≥ its 95th age- and sex-specific percentile for children. Participant demographic data and parental information were obtained through a self-administered questionnaire. Se concentration in fingernail clippings was quantified using flame atomic absorption spectrophotometry.The average age was 10.9 years (standard deviation = 1.0) and the mean BMI was 21.2 kg/m (standard deviation = 5.0). Fingernail Se levels were relatively higher among normal-weight children as compared with obese participants, though the difference was not statistically significant (P = .79). Se levels were inversely associated with the risk of childhood obesity after adjustment for potential confounders. The multivariable-adjusted odds ratio (95% confidence interval) was 0.24 (0.07-0.84) comparing participants in the highest with those who in the lowest tertile of Se levels (Plinear-trend = .03).Our study supported an inverse association between fingernail Se levels and the risk of obesity in Chinese children. Data generated from the present study are useful for designing future prospective cohort studies and/or randomized clinical trials.
Collapse
Affiliation(s)
- Renying Xu
- Department of Clinical Nutrition, Ren Ji Hospital, School of Medicine
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Cheng Chen
- Department of Epidemiology and Biostatistics, School of Public Health – Bloomington, Indiana University, Bloomington, IN
| | - Yiquan Zhou
- Department of Clinical Nutrition, Ren Ji Hospital, School of Medicine
| | - Xiaomin Zhang
- Department of Clinical Nutrition, Ren Ji Hospital, School of Medicine
| | - Yanping Wan
- Department of Clinical Nutrition, Ren Ji Hospital, School of Medicine
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| |
Collapse
|
34
|
Li C, Xu MM, Wang K, Adler AJ, Vella AT, Zhou B. Macrophage polarization and meta-inflammation. Transl Res 2018; 191:29-44. [PMID: 29154757 PMCID: PMC5776711 DOI: 10.1016/j.trsl.2017.10.004] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022]
Abstract
Chronic overnutrition and obesity induces low-grade inflammation throughout the body. Termed "meta-inflammation," this chronic state of inflammation is mediated by macrophages located within the colon, liver, muscle, and adipose tissue. A sentinel orchestrator of immune activity and homeostasis, macrophages adopt variable states of activation as a function of time and environmental cues. Meta-inflammation phenotypically skews these polarization states and has been linked to numerous metabolic disorders. The past decade has revealed several key regulators of macrophage polarization, including the signal transducer and activator of transcription family, the peroxisome proliferator-activated receptor gamma, the CCAAT-enhancer-binding proteins (C/EBP) family, and the interferon regulatory factors. Recent studies have also suggested that microRNAs and long noncoding RNA influence macrophage polarization. The pathogenic alteration of macrophage polarization in meta-inflammation is regulated by both extracellular and intracellular cues, resulting in distinct secretome profiles. Meta-inflammation-altered macrophage polarization has been linked to insulin insensitivity, atherosclerosis, inflammatory bowel disease, cancer, and autoimmunity. Thus, further mechanistic exploration into the skewing of macrophage polarization promises to have profound impacts on improving global health.
Collapse
Affiliation(s)
- Chuan Li
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Maria M Xu
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Kepeng Wang
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Adam J Adler
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Anthony T Vella
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn.
| | - Beiyan Zhou
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn.
| |
Collapse
|
35
|
Chiu YHM, Hsu HHL, Wilson A, Coull BA, Pendo MP, Baccarelli A, Kloog I, Schwartz J, Wright RO, Taveras EM, Wright RJ. Prenatal particulate air pollution exposure and body composition in urban preschool children: Examining sensitive windows and sex-specific associations. ENVIRONMENTAL RESEARCH 2017; 158:798-805. [PMID: 28759881 PMCID: PMC5570541 DOI: 10.1016/j.envres.2017.07.026] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 06/14/2017] [Accepted: 07/11/2017] [Indexed: 05/20/2023]
Abstract
BACKGROUND Evolving animal studies and limited epidemiological data show that prenatal air pollution exposure is associated with childhood obesity. Timing of exposure and child sex may play an important role in these associations. We applied an innovative method to examine sex-specific sensitive prenatal windows of exposure to PM2.5 on anthropometric measures in preschool-aged children. METHODS Analyses included 239 children born ≥ 37 weeks gestation in an ethnically-mixed lower-income urban birth cohort. Prenatal daily PM2.5 exposure was estimated using a validated satellite-based spatio-temporal model. Body mass index z-score (BMI-z), fat mass, % body fat, subscapular and triceps skinfold thickness, waist and hip circumferences and waist-to-hip ratio (WHR) were assessed at age 4.0 ± 0.7 years. Using Bayesian distributed lag interaction models (BDLIMs), we examined sex differences in sensitive windows of weekly averaged PM2.5 levels on these measures, adjusting for child age, maternal age, education, race/ethnicity, and pre-pregnancy BMI. RESULTS Mothers were primarily Hispanic (55%) or Black (26%), had ≤ 12 years of education (66%) and never smoked (80%). Increased PM2.5 exposure 8-17 and 15-22 weeks gestation was significantly associated with increased BMI z-scores and fat mass in boys, but not in girls. Higher PM2.5 exposure 10-29 weeks gestation was significantly associated with increased WHR in girls, but not in boys. Prenatal PM2.5 was not significantly associated with other measures of body composition. Estimated cumulative effects across pregnancy, accounting for sensitive windows and within-window effects, were 0.21 (95%CI = 0.01-0.37) for BMI-z and 0.36 (95%CI = 0.12-0.68) for fat mass (kg) in boys, and 0.02 (95%CI = 0.01-0.03) for WHR in girls, all per µg/m3 increase in PM2.5. CONCLUSIONS Increased prenatal PM2.5 exposure was more strongly associated with indices of increased whole body size in boys and with an indicator of body shape in girls. Methods to better characterize vulnerable windows may provide insight into underlying mechanisms contributing to sex-specific associations.
Collapse
Affiliation(s)
- Yueh-Hsiu Mathilda Chiu
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Kravis Children's Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hsiao-Hsien Leon Hsu
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ander Wilson
- Department of Statistics, Colorado State University, Fort Collins, CO, USA
| | - Brent A Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mathew P Pendo
- Center for Medicine, Health and Society, Vanderbilt University College of Arts and Science, Nashville, TN, USA
| | - Andrea Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Itai Kloog
- Department of Geography and Environmental Development, Ben-Gurion University of the Negev, Israel
| | - Joel Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elsie M Taveras
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Rosalind J Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Kravis Children's Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
36
|
Luna-Vital D, Weiss M, Gonzalez de Mejia E. Anthocyanins from Purple Corn Ameliorated Tumor Necrosis Factor-α-Induced Inflammation and Insulin Resistance in 3T3-L1 Adipocytes via Activation of Insulin Signaling and Enhanced GLUT4 Translocation. Mol Nutr Food Res 2017; 61. [PMID: 28759152 DOI: 10.1002/mnfr.201700362] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/04/2017] [Indexed: 12/19/2022]
Abstract
SCOPE The aim was to compare the effect of an anthocyanin-rich extract from purple corn pericarp (PCW) and pure anthocyanins on adipogenesis, inflammation, and insulin resistance in 3T3-L1 adipocytes on basal and inflammatory conditions. METHODS AND RESULTS Preadipocytes (3T3-L1) were treated during differentiation with or without PCW. Differentiated adipocytes were treated either individually or in combination with tumor necrosis factor α (TNF-α) and PCW, or pure C3G, Pr3G, P3G. PCW reduced preadipocyte differentiation (IC50 = 0.4 mg/mL). PCW and pure anthocyanins including C3G reduced fatty acid synthase enzymatic activity. PCW reduced TNF-α-dependent inflammatory status increasing adiponectin (39%), and decreasing leptin (-79%). PCW and C3G increased glucose uptake and reduced reactive oxygen species generation in insulin resistant adipocytes. An increase in phosphorylation was observed in AKT, IKK, and MEK, and a decrease in IRS and mTOR activating the insulin receptor-associated pathway. PCW (7.5-fold) and C3G (6.3-fold) enhanced GLUT4 membrane translocation compared to insulin resistant adipocytes. CONCLUSION Anthocyanins from colored corn prevented adipocyte differentiation, lipid accumulation, and reduced PPAR-γ transcriptional activity on adipocytes in basal conditions. Ameliorated TNF-α-induced inflammation and insulin resistance in adipocytes via activation of insulin signaling and enhanced GLUT4 translocation suggesting a reduced hyperglycemia associated with the metabolic syndrome.
Collapse
Affiliation(s)
- Diego Luna-Vital
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Matthew Weiss
- School of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Elvira Gonzalez de Mejia
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
37
|
Rahman MM, Alam MN, Ulla A, Sumi FA, Subhan N, Khan T, Sikder B, Hossain H, Reza HM, Alam MA. Cardamom powder supplementation prevents obesity, improves glucose intolerance, inflammation and oxidative stress in liver of high carbohydrate high fat diet induced obese rats. Lipids Health Dis 2017; 16:151. [PMID: 28806968 PMCID: PMC5557534 DOI: 10.1186/s12944-017-0539-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 08/02/2017] [Indexed: 12/20/2022] Open
Abstract
Background Cardamom is a well-known spice in Indian subcontinent, used in culinary and traditional medicine practices since ancient times. The current investigation was untaken to evaluate the potential benefit of cardamom powder supplementation in high carbohydrate high fat (HCHF) diet induced obese rats. Method Male Wistar rats (28 rats) were divided into four different groups such as Control, Control + cardamom, HCHF, HCHF + cardamom. High carbohydrate and high fat (HCHF) diet was prepared in our laboratory. Oral glucose tolerance test, organs wet weight measurements and oxidative stress parameters analysis as well as liver marker enzymes such as alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) activities were assayed on the tissues collected from the rats. Plasma lipids profiles were also measured in all groups of animals. Moreover, histological staining was also performed to evaluate inflammatory cells infiltration and fibrosis in liver. Results The current investigation showed that, HCHF diet feeding in rats developed glucose intolerance and increased peritoneal fat deposition compared to control rats. Cardamom powder supplementation improved the glucose intolerance significantly (p > 0.05) and prevented the abdominal fat deposition in HCHF diet fed rats. HCHF diet feeding in rats also developed dyslipidemia, increased fat deposition and inflammation in liver compared to control rats. Cardamom powder supplementation significantly prevented the rise of lipid parameters (p > 0.05) in HCHF diet fed rats. Histological assessments confirmed that HCHF diet increased the fat deposition and inflammatory cells infiltration in liver which was normalized by cardamom powder supplementation in HCHF diet fed rats. Furthermore, HCHF diet increased lipid peroxidation, decreased antioxidant enzymes activities and increased advanced protein oxidation product level significantly (p > 0.05) both in plasma and liver tissue which were modulated by cardamom powder supplementation in HCHF diet fed rats. HCHF diet feeding in rats also increased the ALT, AST and ALP enzyme activities in plasma which were also normalized by cardamom powder supplementation in HCHF diet fed rats. Moreover, cardamom powder supplementation ameliorated the fibrosis in liver of HCHF diet fed rats. Conclusion This study suggests that, cardamom powder supplementation can prevent dyslipidemia, oxidative stress and hepatic damage in HCHF diet fed rats.
Collapse
Affiliation(s)
- Md Mizanur Rahman
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Mohammad Nazmul Alam
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Anayt Ulla
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Farzana Akther Sumi
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Nusrat Subhan
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Trisha Khan
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Bishwajit Sikder
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Hemayet Hossain
- BCSIR Laboratories, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Hasan Mahmud Reza
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Md Ashraful Alam
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh.
| |
Collapse
|
38
|
Billeter AT, Vittas S, Israel B, Scheurlen KM, Hidmark A, Fleming TH, Kopf S, Büchler MW, Müller-Stich BP. Gastric bypass simultaneously improves adipose tissue function and insulin-dependent type 2 diabetes mellitus. Langenbecks Arch Surg 2017; 402:901-910. [DOI: 10.1007/s00423-017-1601-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/29/2017] [Indexed: 01/06/2023]
|
39
|
Does Regular Exercise Counter T Cell Immunosenescence Reducing the Risk of Developing Cancer and Promoting Successful Treatment of Malignancies? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4234765. [PMID: 28751932 PMCID: PMC5511671 DOI: 10.1155/2017/4234765] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 05/11/2017] [Accepted: 06/01/2017] [Indexed: 12/15/2022]
Abstract
Moderate intensity aerobic exercise training or regular physical activity is beneficial for immune function. For example, some evidence shows that individuals with an active lifestyle exhibit stronger immune responses to vaccination compared to those who are inactive. Encouragingly, poor vaccine responses, which are characteristic of an ageing immune system, can be improved by single or repeated bouts of exercise. In addition, exercise-induced lymphocytosis, and the subsequent lymphocytopenia, is thought to facilitate immune surveillance, whereby lymphocytes search tissues for antigens derived from viruses, bacteria, or malignant transformation. Aerobic exercise training is anti-inflammatory and is linked to lower morbidity and mortality from diseases with infectious, immunological, and inflammatory aetiologies, including cancer. These observations have led to the view that aerobic exercise training might counter the age-associated decline in immune function, referred to as immunosenescence. This article summarises the aspects of immune function that are sensitive to exercise-induced change, highlighting the observations which have stimulated the idea that aerobic exercise training could prevent, limit, or delay immunosenescence, perhaps even restoring aged immune profiles. These potential exercise-induced anti-immunosenescence effects might contribute to the mechanisms by which active lifestyles reduce the risk of developing cancer and perhaps benefit patients undergoing cancer therapy.
Collapse
|
40
|
Peleli M, Carlstrom M. Adenosine signaling in diabetes mellitus and associated cardiovascular and renal complications. Mol Aspects Med 2017; 55:62-74. [DOI: 10.1016/j.mam.2016.12.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/18/2016] [Accepted: 12/21/2016] [Indexed: 12/21/2022]
|
41
|
Samson R, Qi A, Jaiswal A, Le Jemtel TH, Oparil S. Obesity-Associated Hypertension: the Upcoming Phenotype in African-American Women. Curr Hypertens Rep 2017; 19:41. [DOI: 10.1007/s11906-017-0738-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
42
|
Justice AE, Winkler TW, Feitosa MF, Graff M, Fisher VA, Young K, Barata L, Deng X, Czajkowski J, Hadley D, Ngwa JS, Ahluwalia TS, Chu AY, Heard-Costa NL, Lim E, Perez J, Eicher JD, Kutalik Z, Xue L, Mahajan A, Renström F, Wu J, Qi Q, Ahmad S, Alfred T, Amin N, Bielak LF, Bonnefond A, Bragg J, Cadby G, Chittani M, Coggeshall S, Corre T, Direk N, Eriksson J, Fischer K, Gorski M, Neergaard Harder M, Horikoshi M, Huang T, Huffman JE, Jackson AU, Justesen JM, Kanoni S, Kinnunen L, Kleber ME, Komulainen P, Kumari M, Lim U, Luan J, Lyytikäinen LP, Mangino M, Manichaikul A, Marten J, Middelberg RPS, Müller-Nurasyid M, Navarro P, Pérusse L, Pervjakova N, Sarti C, Smith AV, Smith JA, Stančáková A, Strawbridge RJ, Stringham HM, Sung YJ, Tanaka T, Teumer A, Trompet S, van der Laan SW, van der Most PJ, Van Vliet-Ostaptchouk JV, Vedantam SL, Verweij N, Vink JM, Vitart V, Wu Y, Yengo L, Zhang W, Hua Zhao J, Zimmermann ME, Zubair N, Abecasis GR, Adair LS, Afaq S, Afzal U, Bakker SJL, Bartz TM, Beilby J, Bergman RN, Bergmann S, Biffar R, Blangero J, Boerwinkle E, Bonnycastle LL, Bottinger E, Braga D, Buckley BM, Buyske S, Campbell H, et alJustice AE, Winkler TW, Feitosa MF, Graff M, Fisher VA, Young K, Barata L, Deng X, Czajkowski J, Hadley D, Ngwa JS, Ahluwalia TS, Chu AY, Heard-Costa NL, Lim E, Perez J, Eicher JD, Kutalik Z, Xue L, Mahajan A, Renström F, Wu J, Qi Q, Ahmad S, Alfred T, Amin N, Bielak LF, Bonnefond A, Bragg J, Cadby G, Chittani M, Coggeshall S, Corre T, Direk N, Eriksson J, Fischer K, Gorski M, Neergaard Harder M, Horikoshi M, Huang T, Huffman JE, Jackson AU, Justesen JM, Kanoni S, Kinnunen L, Kleber ME, Komulainen P, Kumari M, Lim U, Luan J, Lyytikäinen LP, Mangino M, Manichaikul A, Marten J, Middelberg RPS, Müller-Nurasyid M, Navarro P, Pérusse L, Pervjakova N, Sarti C, Smith AV, Smith JA, Stančáková A, Strawbridge RJ, Stringham HM, Sung YJ, Tanaka T, Teumer A, Trompet S, van der Laan SW, van der Most PJ, Van Vliet-Ostaptchouk JV, Vedantam SL, Verweij N, Vink JM, Vitart V, Wu Y, Yengo L, Zhang W, Hua Zhao J, Zimmermann ME, Zubair N, Abecasis GR, Adair LS, Afaq S, Afzal U, Bakker SJL, Bartz TM, Beilby J, Bergman RN, Bergmann S, Biffar R, Blangero J, Boerwinkle E, Bonnycastle LL, Bottinger E, Braga D, Buckley BM, Buyske S, Campbell H, Chambers JC, Collins FS, Curran JE, de Borst GJ, de Craen AJM, de Geus EJC, Dedoussis G, Delgado GE, den Ruijter HM, Eiriksdottir G, Eriksson AL, Esko T, Faul JD, Ford I, Forrester T, Gertow K, Gigante B, Glorioso N, Gong J, Grallert H, Grammer TB, Grarup N, Haitjema S, Hallmans G, Hamsten A, Hansen T, Harris TB, Hartman CA, Hassinen M, Hastie ND, Heath AC, Hernandez D, Hindorff L, Hocking LJ, Hollensted M, Holmen OL, Homuth G, Jan Hottenga J, Huang J, Hung J, Hutri-Kähönen N, Ingelsson E, James AL, Jansson JO, Jarvelin MR, Jhun MA, Jørgensen ME, Juonala M, Kähönen M, Karlsson M, Koistinen HA, Kolcic I, Kolovou G, Kooperberg C, Krämer BK, Kuusisto J, Kvaløy K, Lakka TA, Langenberg C, Launer LJ, Leander K, Lee NR, Lind L, Lindgren CM, Linneberg A, Lobbens S, Loh M, Lorentzon M, Luben R, Lubke G, Ludolph-Donislawski A, Lupoli S, Madden PAF, Männikkö R, Marques-Vidal P, Martin NG, McKenzie CA, McKnight B, Mellström D, Menni C, Montgomery GW, Musk AW(B, Narisu N, Nauck M, Nolte IM, Oldehinkel AJ, Olden M, Ong KK, Padmanabhan S, Peyser PA, Pisinger C, Porteous DJ, Raitakari OT, Rankinen T, Rao DC, Rasmussen-Torvik LJ, Rawal R, Rice T, Ridker PM, Rose LM, Bien SA, Rudan I, Sanna S, Sarzynski MA, Sattar N, Savonen K, Schlessinger D, Scholtens S, Schurmann C, Scott RA, Sennblad B, Siemelink MA, Silbernagel G, Slagboom PE, Snieder H, Staessen JA, Stott DJ, Swertz MA, Swift AJ, Taylor KD, Tayo BO, Thorand B, Thuillier D, Tuomilehto J, Uitterlinden AG, Vandenput L, Vohl MC, Völzke H, Vonk JM, Waeber G, Waldenberger M, Westendorp RGJ, Wild S, Willemsen G, Wolffenbuttel BHR, Wong A, Wright AF, Zhao W, Zillikens MC, Baldassarre D, Balkau B, Bandinelli S, Böger CA, Boomsma DI, Bouchard C, Bruinenberg M, Chasman DI, Chen YD, Chines PS, Cooper RS, Cucca F, Cusi D, Faire UD, Ferrucci L, Franks PW, Froguel P, Gordon-Larsen P, Grabe HJ, Gudnason V, Haiman CA, Hayward C, Hveem K, Johnson AD, Wouter Jukema J, Kardia SLR, Kivimaki M, Kooner JS, Kuh D, Laakso M, Lehtimäki T, Marchand LL, März W, McCarthy MI, Metspalu A, Morris AP, Ohlsson C, Palmer LJ, Pasterkamp G, Pedersen O, Peters A, Peters U, Polasek O, Psaty BM, Qi L, Rauramaa R, Smith BH, Sørensen TIA, Strauch K, Tiemeier H, Tremoli E, van der Harst P, Vestergaard H, Vollenweider P, Wareham NJ, Weir DR, Whitfield JB, Wilson JF, Tyrrell J, Frayling TM, Barroso I, Boehnke M, Deloukas P, Fox CS, Hirschhorn JN, Hunter DJ, Spector TD, Strachan DP, van Duijn CM, Heid IM, Mohlke KL, Marchini J, Loos RJF, Kilpeläinen TO, Liu CT, Borecki IB, North KE, Cupples LA. Genome-wide meta-analysis of 241,258 adults accounting for smoking behaviour identifies novel loci for obesity traits. Nat Commun 2017; 8:14977. [PMID: 28443625 PMCID: PMC5414044 DOI: 10.1038/ncomms14977] [Show More Authors] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 02/15/2017] [Indexed: 02/07/2023] Open
Abstract
Few genome-wide association studies (GWAS) account for environmental exposures, like smoking, potentially impacting the overall trait variance when investigating the genetic contribution to obesity-related traits. Here, we use GWAS data from 51,080 current smokers and 190,178 nonsmokers (87% European descent) to identify loci influencing BMI and central adiposity, measured as waist circumference and waist-to-hip ratio both adjusted for BMI. We identify 23 novel genetic loci, and 9 loci with convincing evidence of gene-smoking interaction (GxSMK) on obesity-related traits. We show consistent direction of effect for all identified loci and significance for 18 novel and for 5 interaction loci in an independent study sample. These loci highlight novel biological functions, including response to oxidative stress, addictive behaviour, and regulatory functions emphasizing the importance of accounting for environment in genetic analyses. Our results suggest that tobacco smoking may alter the genetic susceptibility to overall adiposity and body fat distribution.
Collapse
Affiliation(s)
- Anne E. Justice
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Thomas W. Winkler
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - Misa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Virginia A. Fisher
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Kristin Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Llilda Barata
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - Xuan Deng
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Jacek Czajkowski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - David Hadley
- Population Health Research Institute, St. George's, University of London, London, SW17 0RE, UK
- TransMed Systems, Inc., Cupertino, California 95014, USA
| | - Julius S. Ngwa
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore Maryland, USA
| | - Tarunveer S. Ahluwalia
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Audrey Y. Chu
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
| | - Nancy L. Heard-Costa
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Jeremiah Perez
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - John D. Eicher
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, USA
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss instititute of Bioinformatics
| | - Luting Xue
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Frida Renström
- Department of Biobank Research, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 02 Malmö, Sweden
| | - Joseph Wu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Shafqat Ahmad
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 02 Malmö, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Tamuno Alfred
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Najaf Amin
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015GE, The Netherlands
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Amelie Bonnefond
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Jennifer Bragg
- Internal Medicine - Nephrology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Gemma Cadby
- Centre for Genetic Origins of Health and Disease, University of Western Australia, Crawley, Australia
| | - Martina Chittani
- Department of Health Sciences, University of Milan,Via A. Di Rudiní, 8 20142, Milano, Italy
| | - Scott Coggeshall
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
| | - Tanguy Corre
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss instititute of Bioinformatics
| | - Nese Direk
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Psychiatry, Dokuz Eylul University, Izmir, Turkey
| | - Joel Eriksson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Mathias Gorski
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Marie Neergaard Harder
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Momoko Horikoshi
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Tao Huang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Epidemiology Domain, Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Jennifer E. Huffman
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, USA
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Anne U. Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Johanne Marie Justesen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Leena Kinnunen
- Department of Health, National Institute for Health and Welfare, Helsinki, FI-00271 Finland
| | - Marcus E. Kleber
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Meena Kumari
- ISER, University of Essex, Colchester CO43SQ, UK
- Department of Epidemiology and Public Health, UCL, London, WC1E 6BT, UK
| | - Unhee Lim
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre at Guy's and St. Thomas' Foundation Trust, London, UK
| | - Ani Manichaikul
- Center for Public Health Genomics and Biostatistics Section, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Jonathan Marten
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Rita P. S. Middelberg
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Department of Medicine I, University Hospital Grosshadern, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Louis Pérusse
- Department of Kinesiology, Faculty of Medicine, Université Laval, Québec, Canada
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Canada
| | - Natalia Pervjakova
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu 51010, Estonia
| | - Cinzia Sarti
- Department of Social and Health Care, City of Helsinki, Helsinki, Finland
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Alena Stančáková
- Department of Medicine, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Rona J. Strawbridge
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Heather M. Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore Maryland, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Germany
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, The Netherlands
| | - Sander W. van der Laan
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | - Peter J. van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | | | - Sailaja L. Vedantam
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Jacqueline M. Vink
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Ying Wu
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Loic Yengo
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Weihua Zhang
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Martina E. Zimmermann
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Niha Zubair
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Gonçalo R. Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Linda S. Adair
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Saima Afaq
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
| | - Uzma Afzal
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
| | - Stephan J. L. Bakker
- Department of Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Traci M. Bartz
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington 98101, USA
| | - John Beilby
- Busselton Population Medical Research Institute, Nedlands, Western Australia 6009, Australia
- PathWest Laboratory Medicine of WA, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
- School of Pathology and Laboraty Medicine, The University of Western Australia, 35 Stirling Hwy, Crawley, Western Australia 6009, Australia
| | - Richard N. Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss instititute of Bioinformatics
| | - Reiner Biffar
- Clinic for Prosthetic Dentistry, Gerostomatology and Material Science, University Medicine Greifswald, Germany
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Eric Boerwinkle
- Human Genetics Center, The University of Texas Health Science Center, PO Box 20186, Houston, Texas 77225, USA
| | - Lori L. Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Erwin Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Daniele Braga
- Department of Health Sciences, University of Milan,Via A. Di Rudiní, 8 20142, Milano, Italy
| | - Brendan M. Buckley
- Department of Pharmacology and Therapeutics, University College Cork, Ireland
| | - Steve Buyske
- Department of Genetics, Rutgers University, Piscataway, New Jersey 08854, USA
- Department of Statistics and Biostatistics, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Harry Campbell
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - John C. Chambers
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Francis S. Collins
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Joanne E. Curran
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, Texas, USA
| | - Gert J. de Borst
- Department of Vascular Surgery, Division of Surgical Specialties, UMC Utrecht, The Netherlands
| | - Anton J. M. de Craen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, The Netherlands
| | - Eco J. C. de Geus
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- EMGO+ Institute Vrije Universiteit & Vrije Universiteit Medical Center
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Graciela E. Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | | | - Anna L. Eriksson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Tõnu Esko
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
| | - Jessica D. Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, UK
| | - Terrence Forrester
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, JMAAW15 Jamaica
| | - Karl Gertow
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Bruna Gigante
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nicola Glorioso
- Hypertension and Related Disease Centre, AOU-University of Sassari
| | - Jian Gong
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- German Center for Diabetes Research, D-85764 Neuherberg, Germany
| | - Tanja B. Grammer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Saskia Haitjema
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | - Göran Hallmans
- Department of Public Health and Clinical Medicine, Section for Nutritional Research, Umeå University, Umeå, Sweden
| | - Anders Hamsten
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tamara B. Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Catharina A. Hartman
- Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Maija Hassinen
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Nicholas D. Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Andrew C. Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Lucia Hindorff
- Division of Genomic Medicine, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Lynne J. Hocking
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Mette Hollensted
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Germany
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jie Huang
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Joseph Hung
- School of Medicine and Pharmacology, The University of Western Australia, 25 Stirling Hwy, Crawley, Western Australia 6009, Australia
- Department of Cardiovascular Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Nina Hutri-Kähönen
- Department of Pediatrics, Tampere University Hospital, Tampere 33521, Finland
- Department of Pediatrics, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology, Uppsala University, Uppsala, 751 85, Sweden
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
- Science for Life Laboratory, Uppsala University, Uppsala, 750 85, Sweden
| | - Alan L. James
- Busselton Population Medical Research Institute, Nedlands, Western Australia 6009, Australia
- School of Medicine and Pharmacology, The University of Western Australia, 25 Stirling Hwy, Crawley, Western Australia 6009, Australia
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC–PHE Centre for Environment & Health, School of Public Health, Imperial College London, UK
- Center for Life Course Epidemiology, Faculty of Medicine, University of OuluP.O.Box 5000, FI-90014, Finland
- Biocenter Oulu, University of Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Kajaanintie 50, P.O.Box 20, FI-90220, 90029 Oulu, Finland
| | - Min A. Jhun
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Markus Juonala
- Department of Medicine, University of Turku, Turku 20520 Finland
- Division of Medicine, Turku University Hospital, Turku 20521, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere 33521, Finland
- Department of Clinical Physiology, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Magnus Karlsson
- Clinical and Molecular Osteoporosis Research Unit, Department of Orthopedics and Clinical Sciences, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Heikki A. Koistinen
- Department of Health, National Institute for Health and Welfare, Helsinki, FI-00271 Finland
- Department of Medicine and Abdominal Center: Endocrinology, University of Helsinki and Helsinki University Central Hospital, Helsinki, FI-00029 Finland
- Minerva Foundation Institute for Medical Research, Biomedicum 2U, Helsinki, FI-00290 Finland
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, Croatia
| | - Genovefa Kolovou
- Department of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Bernhard K. Krämer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Kirsti Kvaløy
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Timo A. Lakka
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine/Physiology, University of Eastern Finland, Kuopio Campus, Finland
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Lenore J. Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Karin Leander
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Nanette R. Lee
- USC-Office of Population Studies Foundation, Inc., University of San Carlos, Cebu City 6000, Philippines
- Department of Anthropology, Sociology and History, University of San Carlos, Cebu City 6000, Philippines
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University, Uppsala 751 85, Sweden
| | - Cecilia M. Lindgren
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Li Ka Shing Centre for Health Information and Discovery, The Big Data Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Allan Linneberg
- Research Centre for Prevention and Health, the Capital Region of Denmark, Copenhagen, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stephane Lobbens
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Marie Loh
- Dept Epidemiology and Biostatistics, School of Public Health, Imperical College London, UK
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Level 5, Singapore 138648, Singapore
| | - Mattias Lorentzon
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Robert Luben
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Gitta Lubke
- Department of Psychology, University of Notre Dame, Notre Dame, USA
| | - Anja Ludolph-Donislawski
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Sara Lupoli
- Department of Health Sciences, University of Milan,Via A. Di Rudiní, 8 20142, Milano, Italy
| | - Pamela A. F. Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Reija Männikkö
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne university hospital (CHUV), Lausanne, Switzerland
| | - Nicholas G. Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Colin A. McKenzie
- Tropical Metabolism Research Unit, Tropical Medicine Research Institute, University of the West Indies, Mona, JMAAW15 Jamaica
| | - Barbara McKnight
- Department of Biostatistics, University of Washington, Seattle, Washington 98195, USA
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington 98101, USA
- Program in Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Dan Mellström
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Grant W. Montgomery
- Molecular Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - AW (Bill) Musk
- Busselton Population Medical Research Institute, Nedlands, Western Australia 6009, Australia
- School of Population Health, The University of Western Australia, 35 Stirling Hwy, Crawley, Western Australia 6009, Australia
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia 6009, Australia
| | - Narisu Narisu
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Germany
| | - Ilja M. Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Albertine J. Oldehinkel
- Interdisciplinary Center Psychopathology and Emotion Regulation (ICPE), University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Matthias Olden
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Sandosh Padmanabhan
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Scotland
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Charlotta Pisinger
- Research Center for Prevention and Health, Glostrup Hospital, Glostrup Denmark
- Department of Public Health, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - David J. Porteous
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20521, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - D. C. Rao
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Laura J. Rasmussen-Torvik
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rajesh Rawal
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Treva Rice
- Division of Biostatistics, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
- Division of Cardiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lynda M. Rose
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
| | - Stephanie A. Bien
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Igor Rudan
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Serena Sanna
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, 09042, Monserrato, Italy
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow, UK
| | - Kai Savonen
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Salome Scholtens
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - Bengt Sennblad
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Marten A. Siemelink
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
| | - Günther Silbernagel
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Austria
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Jan A. Staessen
- Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Science , University of Leuven, Campus Sint Rafael, Kapucijnenvoer 35, Leuven; Belgium
- R&D VitaK Group, Maastricht University, Brains Unlimited Building, Oxfordlaan 55, Maastricht, The Netherlands
| | - David J. Stott
- Institute of Cardiovascular and Medical Sciences, Faculty of Medicine, University of Glasgow, UK
| | - Morris A. Swertz
- Department of Genetics, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Amy J. Swift
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Kent D. Taylor
- Center for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute at Harbor/UCLA Medical Center, Torrance, California, USA
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California, USA
| | - Bamidele O. Tayo
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University of Chicago, Maywood, Illinois 61053, USA
| | - Barbara Thorand
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- German Center for Diabetes Research, D-85764 Neuherberg, Germany
| | - Dorothee Thuillier
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
| | - Jaakko Tuomilehto
- Research Division, Dasman Diabetes Institute, Dasman, Kuwait
- Department of Neurosciences and Preventive Medicine, Danube-University Krems, 3500 Krems, Austria
- Saudi Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Andre G. Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Canada
- School of Nutrition, Université Laval, Québec, Canada
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Germany
| | - Judith M. Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Gérard Waeber
- Department of Medicine, Internal Medicine, Lausanne university hospital (CHUV), Lausanne, Switzerland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - R. G. J. Westendorp
- Department of Public Health, and Center for Healthy Ageing, University of Copenhagen, Denmark
| | - Sarah Wild
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Bruce H. R. Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, 33 Bedford Place, London, WC1B 5JU, UK
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - M Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Damiano Baldassarre
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | | | - Carsten A. Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Dorret I. Boomsma
- Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Marcel Bruinenberg
- Lifelines Cohort Study, PO Box 30001, 9700 RB Groningen, The Netherlands
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts USA
- Division of Genetics, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Yii-DerIda Chen
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute and Department of Pediatrics, Harbor-UCLA, Torrance, California 90502, USA
| | - Peter S. Chines
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland 20892, USA
| | - Richard S. Cooper
- Department of Public Health Sciences, Stritch School of Medicine, Loyola University of Chicago, Maywood, Illinois 61053, USA
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale Delle Ricerche (CNR), Cittadella Universitaria di Monserrato, 09042, Monserrato, Italy
- Dipartimento di Scienze Biomediche, Universita' degli Studi di Sassari, Sassari, Italy
| | - Daniele Cusi
- Sanipedia srl, Bresso (Milano), Italy and Institute of Biomedical Technologies National Centre of Research Segrate, Milano, Italy
| | - Ulf de Faire
- Unit of Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore Maryland, USA
| | - Paul W. Franks
- Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Lund University, SE-205 02 Malmö, Sweden
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Public Health & Clinical Medicine, Umeå University, Umeå, Sweden
| | - Philippe Froguel
- University of Lille, CNRS, Institut Pasteur of Lille, UMR 8199 - EGID, Lille, France
- Department of Genomics of Common Disease, Imperial College London, London, UK
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill North Carolina, 27516, USA
| | - Hans- Jörgen Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Christopher A. Haiman
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, 90089, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
| | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and Nursing, Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Andrew D. Johnson
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, The Framingham Heart Study, Framingham, Massachusetts, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, The Netherlands
- Durrer Center for Cardiogenetic Research, Amsterdam, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, UCL, London, WC1E 6BT, UK
| | - Jaspal S. Kooner
- Cardiology, Ealing Hospital NHS Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
- Faculty of Med, National Heart & Lung Institute, Cardiovascular Science, Hammersmith Campus, Hammersmith Hospital, Hammersmith Campus, Imperial College London, UK
| | - Diana Kuh
- MRC Unit for Lifelong Health and Ageing at UCL, 33 Bedford Place, London, WC1B 5JU, UK
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, 70210 Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, University of Tampere, Tampere 33014, Finland
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii 96813, USA
| | - Winfried März
- Synlab Academy, Synlab Services GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
- Oxford National Institute for Health Research (NIHR) Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool L69 3GL, UK
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Lyle J. Palmer
- School of Public Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, Department of Cardiology, Division Heart & Lungs, UMC Utrecht, The Netherlands
- Laboratory of Clinical Chemistry and Hematology, Division Laboratories & Pharmacy, UMC Utrecht, The Netherlands
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- German Center for Diabetes Research, D-85764 Neuherberg, Germany
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle Washington USA
| | - Ozren Polasek
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
- Department of Public Health, Faculty of Medicine, University of Split, Croatia
| | - Bruce M. Psaty
- Department of Medicine, University of Washington, Seattle, Washington 98195, USA
- Department of Epidemiology, University of Washington, Seattle, Washington 98101, USA
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington 98101, USA
| | - Lu Qi
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Rainer Rauramaa
- Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Blair H. Smith
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland
- Division of Population Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4RB, Scotland
| | - Thorkild I. A. Sørensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Epidemiology (formerly Institute of Preventive Medicine), Bispebjerg and Frederiksberg Hospital (2000 Frederiksberg), The Capital Region, Copenhagen, Denmark
- MRC Integrative Epidemiology Unit, Bristol University, Bristol, UK
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, D-85764 Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, D-81377 Munich, Germany
| | - Henning Tiemeier
- Department of Psychiatry Erasmus Medical Center, Rotterdam, The Netherlands
| | - Elena Tremoli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Milan, Italy
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, The Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands
| | - Henrik Vestergaard
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center, Gentofte, Denmark
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne university hospital (CHUV), Lausanne, Switzerland
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
| | - David R. Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - John B. Whitfield
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Scotland, UK
| | - Jessica Tyrrell
- Genetics of Complex Traits, University of Exeter Medical School, RILD Building University of Exeter, Exeter, EX2 5DW, UK
- European Centre for Environment and Human Health, University of Exeter Medical School, The Knowledge Spa, Truro TR1 3HD, UK
| | - Timothy M. Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter EX1 2LU, UK
| | - Inês Barroso
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Level 4, Institute of Metabolic Science Box 289 Addenbrooke's Hospital Cambridge CB2 OQQ, UK
- University of Cambridge Metabolic Research Laboratories, Level 4, Institute of Metabolic Science Box 289 Addenbrooke's Hospital Cambridge CB2 OQQ, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Panagiotis Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Caroline S. Fox
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
| | - Joel N. Hirschhorn
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Department of Genetics, Harvard Medical School, Boston Massachusetts 02115, USA
| | - David J. Hunter
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - David P. Strachan
- Population Health Research Institute, St. George's, University of London, London, SW17 0RE, UK
- Division of Population Health Sciences and Education, St George's, University of London, London SW17 0RE, UK
| | - Cornelia M. van Duijn
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus University Medical Center, Rotterdam 3015GE, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands Consortium for Healthy Aging (NCHA). Leiden, The Netherlands
- Center for Medical Systems Biology, Leiden, The Netherlands
| | - Iris M. Heid
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, D-93053 Regensburg, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
- The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, USA
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
- Mount Sinai School of Medicine, New York 10029, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Tuomas O. Kilpeläinen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge CB2 0QQ, UK
- Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine; St. Louis, Missouri, 63108 USA
| | - Kari E. North
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts 02118, USA
- NHLBI Framingham Heart Study, Framingham, Massachusetts, 01702 USA
| |
Collapse
|
43
|
Lee Y, Fluckey JD, Chakraborty S, Muthuchamy M. Hyperglycemia- and hyperinsulinemia-induced insulin resistance causes alterations in cellular bioenergetics and activation of inflammatory signaling in lymphatic muscle. FASEB J 2017; 31:2744-2759. [PMID: 28298335 DOI: 10.1096/fj.201600887r] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/22/2017] [Indexed: 12/27/2022]
Abstract
Insulin resistance is a well-known risk factor for obesity, metabolic syndrome (MetSyn) and associated cardiovascular diseases, but its mechanisms are undefined in the lymphatics. Mesenteric lymphatic vessels from MetSyn or LPS-injected rats exhibited impaired intrinsic contractile activity and associated inflammatory changes. Hence, we hypothesized that insulin resistance in lymphatic muscle cells (LMCs) affects cell bioenergetics and signaling pathways that consequently alter contractility. LMCs were treated with different concentrations of insulin or glucose or both at various time points to determine insulin resistance. Onset of insulin resistance significantly impaired glucose uptake, mitochondrial function, oxygen consumption rates, glycolysis, lactic acid, and ATP production in LMCs. Hyperglycemia and hyperinsulinemia also impaired the PI3K/Akt while enhancing the ERK/p38MAPK/JNK pathways in LMCs. Increased NF-κB nuclear translocation and macrophage chemoattractant protein-1 and VCAM-1 levels in insulin-resistant LMCs indicated activation of inflammatory mechanisms. In addition, increased phosphorylation of myosin light chain-20, a key regulator of lymphatic muscle contraction, was observed in insulin-resistant LMCs. Therefore, our data elucidate the mechanisms of insulin resistance in LMCs and provide the first evidence that hyperglycemia and hyperinsulinemia promote insulin resistance and impair lymphatic contractile status by reducing glucose uptake, altering cellular metabolic pathways, and activating inflammatory signaling cascades.-Lee, Y., Fluckey, J. D., Chakraborty, S., Muthuchamy, M. Hyperglycemia- and hyperinsulinemia-induced insulin resistance causes alterations in cellular bioenergetics and activation of inflammatory signaling in lymphatic muscle.
Collapse
Affiliation(s)
- Yang Lee
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| | - James D Fluckey
- Department of Health and Kinesiology, Texas A&M University, College Station, Texas, USA
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA;
| | - Mariappan Muthuchamy
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA;
| |
Collapse
|
44
|
Prenatal phthalate exposure and 8-isoprostane among Mexican-American children with high prevalence of obesity. J Dev Orig Health Dis 2016; 8:196-205. [PMID: 28031075 DOI: 10.1017/s2040174416000763] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oxidative stress has been linked to many obesity-related conditions among children including cardiovascular disease, diabetes mellitus and hypertension. Exposure to environmental chemicals such as phthalates, ubiquitously found in humans, may also generate reactive oxygen species and subsequent oxidative stress. We examined longitudinal changes of 8-isoprostane urinary concentrations, a validated biomarker of oxidative stress, and associations with maternal prenatal urinary concentrations of phthalate metabolites for 258 children at 5, 9 and 14 years of age participating in a birth cohort residing in an agricultural area in California. Phthalates are endocrine disruptors, and in utero exposure has been also linked to altered lipid metabolism, as well as adverse birth and neurodevelopmental outcomes. We found that median creatinine-corrected 8-isoprostane concentrations remained constant across all age groups and did not differ by sex. Total cholesterol, systolic and diastolic blood pressure were positively associated with 8-isoprostane in 14-year-old children. No associations were observed between 8-isoprostane and body mass index (BMI), BMI Z-score or waist circumference at any age. Concentrations of three metabolites of high molecular weight phthalates measured at 13 weeks of gestation (monobenzyl, monocarboxyoctyl and monocarboxynonyl phthalates) were negatively associated with 8-isoprostane concentrations among 9-year olds. However, at 14 years of age, isoprostane concentrations were positively associated with two other metabolites (mono(2-ethylhexyl) and mono(2-ethyl-5-carboxypentyl) phthalates) measured in early pregnancy. Longitudinal data on 8-isoprostane in this pediatric population with a high prevalence of obesity provides new insight on certain potential cardiometabolic risks of prenatal exposure to phthalates.
Collapse
|
45
|
Carmona-Montesinos E, Velazquez-Perez R, Pichardo Aguirre E, Rivas-Arancibia S. Obesity, Oxidative Stress, and Their Effect on Serum Heme Oxygenase-1 Concentrations and Insulin in Children Aged 3 to 5 Years in a Pediatric Hospital of the Ministry of Health CDMX. Child Obes 2016; 12:474-481. [PMID: 27728771 DOI: 10.1089/chi.2016.0155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Obesity during early stages of life may condition states of oxidative stress. Heme oxygenase-1 (HO-1) is an enzyme involved in oxidative metabolism; it has antioxidant and anti-inflammatory functions and is related in sensitivity to insulin. However, a high concentration of this enzyme has been described to cause alterations such as insulin resistance. The objective of this work was to study the relationship between obesity, oxidative stress, HO-1, and insulin in children aged 3 to 5 years. METHODS To achieve our objective, we studied a control group of children (n = 50) and a group of obese children (n = 50) who underwent an anthropometric evaluation. Additionally, we quantified peroxidized lipids, oxidized low-density lipoproteins (Ox-LDLs), oxidized and reduced glutathione, HO-1, and insulin. We also calculated the homeostasis model assessment of insulin resistance (HOMA-IR), HOMA-%B, and HOMA-%S indices. According to the data distribution, nonparametric and Spearman's rank correlation coefficient tests were conducted. RESULTS The results demonstrate that obese children show a statistically relevant increase in BMI/age, serum concentrations of peroxidized lipids, Ox-LDLs, oxidized glutathione, HO-1, and insulin (p < 0.005). In addition, there was an increase in the HOMA-IR and HOMA-%B (p < 0.0001) indices and a decrease of reduced glutathione, as well as a reduction in the HOMA-%S, compared with the children of the control group (p < 0.003). CONCLUSIONS With the results obtained, we can conclude that obese preschool children show a chronic state of oxidative stress, an increase of HO-1, and an incipient state of insulin resistance. Finally, the increased reactive oxygen species could be one of the leading factors involved in insulin resistance and Ox-LDL increase from the preschool stage.
Collapse
Affiliation(s)
- Enrique Carmona-Montesinos
- 1 Departamento de Fisiologia, Facultad de Medicina, UNAM , Coyoacan, Mexico .,2 Hospital Pediatrico San Juan de Aragon , Secretaria de Salud CDMX, Gustavo A. Madero, Mexico
| | | | - Edna Pichardo Aguirre
- 3 Servicio de Laboratorio Clínico, Hospital Materno Pediatrico Xochimilco , Secretaria de Salud CDMX, Xochimilco, Mexico
| | | |
Collapse
|
46
|
Middlemiss JE, McEniery CM. Feeling the pressure: (patho) physiological mechanisms of weight gain and weight loss in humans. Hypertens Res 2016; 40:226-236. [PMID: 27760999 DOI: 10.1038/hr.2016.142] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/30/2016] [Accepted: 09/12/2016] [Indexed: 12/12/2022]
Abstract
Obesity is an ongoing global epidemic and has adverse consequences for cardiovascular health. Obesity is often associated with hypertension, which is, itself, a common condition and an important cause of morbidity and mortality worldwide. Although animal models of obesity have provided extensive data on the links between obesity and hypertension, a greater understanding of the pathways linking obesity and hypertension in humans is likely to assist translation of animal data, and may, itself, identify important treatment strategies. Ultimately, this could have a substantial impact on human health, both at an individual and population level. The current review will focus specifically on studies of experimental weight gain and weight loss in humans and the following key areas, which are strongly related to blood pressure: cardiovascular function, autonomic nervous system function, metabolic function and the impact of cardiorespiratory fitness.
Collapse
Affiliation(s)
- Jessica E Middlemiss
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| | - Carmel M McEniery
- Division of Experimental Medicine and Immunotherapeutics, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Conceição EPS, Moura EG, Carvalho JC, Oliveira E, Lisboa PC. Early redox imbalance is associated with liver dysfunction at weaning in overfed rats. J Physiol 2016; 593:4799-811. [PMID: 26332355 DOI: 10.1113/jp271189] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/12/2015] [Indexed: 12/30/2022] Open
Abstract
Neonatal overfeeding induced by litter size reduction leads to further obesity and other metabolic disorders, such as liver oxidative stress and microsteatosis at adulthood. We hypothesized that overfeeding causes an early redox imbalance at weaning, which could programme the animals to future liver dysfunction. Thus, we studied lipogenesis, adipogenesis, catecholamine status and oxidative balance in weaned overfed pups. To induce early overfeeding, litters were adjusted to three pups at the 3rd day of lactation (SL group). The control group contained 10 pups per litter until weaning (NL group). Peripheral autonomic nerve function was determined in vivo at 21 days old. Thereafter, pups were killed for further analysis. Differences were considered significant when P < 0.05. The SL pups presented with a higher visceral adipocyte area, higher content of lipogenic enzymes (ACC, FAS) and with a lower content of adipogenic factors (CEBP, PPARγ) in visceral adipose tissue (VAT). Although autonomic nerve activity and adrenal catecholamine production were not significantly altered, catecholamine receptor (β3ADR) content was lower in VAT. The SL pups also presented with higher triglyceride, PPARγ, PPARα and PGC1α contents in liver. In plasma and liver, the SL pups showed an oxidative imbalance, with higher lipid peroxidation and protein oxidation. The SL group presented with a higher serum alanine aminotransferase (ALT). The early increase in lipogenesis in adipose tissue and liver in weaned overfed rats suggests that the higher oxidative stress and lower catecholamine content in VAT are associated with the early development of liver dysfunction and adipocyte hypertrophy.
Collapse
Affiliation(s)
- E P S Conceição
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - E G Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - J C Carvalho
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - E Oliveira
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| | - P C Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, RJ, Brazil
| |
Collapse
|
48
|
Anti-Dyslipidemic Properties of Saffron: Reduction in the Associated Risks of Atherosclerosis and Insulin Resistance. IRANIAN RED CRESCENT MEDICAL JOURNAL 2016. [DOI: 10.5812/ircmj.36226] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
49
|
Turner JE. Is immunosenescence influenced by our lifetime "dose" of exercise? Biogerontology 2016; 17:581-602. [PMID: 27023222 PMCID: PMC4889625 DOI: 10.1007/s10522-016-9642-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 03/03/2016] [Indexed: 02/06/2023]
Abstract
The age-associated decline in immune function, referred to as immunosenescence, is well characterised within the adaptive immune system, and in particular, among T cells. Hallmarks of immunosenescence measured in the T cell pool, include low numbers and proportions of naïve cells, high numbers and proportions of late-stage differentiated effector memory cells, poor proliferative responses to mitogens, and a CD4:CD8 ratio <1.0. These changes are largely driven by infection with Cytomegalovirus, which has been directly linked with increased inflammatory activity, poor responses to vaccination, frailty, accelerated cognitive decline, and early mortality. It has been suggested however, that exercise might exert an anti-immunosenescence effect, perhaps delaying the onset of immunological ageing or even rejuvenating aged immune profiles. This theory has been developed on the basis of evidence that exercise is a powerful stimulus of immune function. For example, in vivo antibody responses to novel antigens can be improved with just minutes of exercise undertaken at the time of vaccination. Further, lymphocyte immune-surveillance, whereby cells search tissues for antigens derived from viruses, bacteria, or malignant transformation, is thought to be facilitated by the transient lymphocytosis and subsequent lymphocytopenia induced by exercise bouts. Moreover, some forms of exercise are anti-inflammatory, and if repeated regularly over the lifespan, there is a lower morbidity and mortality from diseases with an immunological and inflammatory aetiology. The aim of this article is to discuss recent theories for how exercise might influence T cell immunosenescence, exploring themes in the context of hotly debated issues in immunology.
Collapse
Affiliation(s)
- James E Turner
- Department for Health, University of Bath, Claverton Down, Bath, BA2 7AY, UK.
| |
Collapse
|
50
|
Li C, Feng F, Xiong X, Li R, Chen N. Exercise coupled with dietary restriction reduces oxidative stress in male adolescents with obesity. J Sports Sci 2016; 35:663-668. [DOI: 10.1080/02640414.2016.1183807] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|