1
|
Szalanczy AM, Giorgio G, Goff E, Seshie O, Grzybowski M, Klotz J, Geurts AM, Redei EE, Solberg Woods LC. Changes in environmental stress over COVID-19 pandemic likely contributed to failure to replicate adiposity phenotype associated with Krtcap3. Physiol Genomics 2023; 55:452-467. [PMID: 37458463 PMCID: PMC10642928 DOI: 10.1152/physiolgenomics.00019.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/25/2023] [Accepted: 07/09/2023] [Indexed: 07/28/2023] Open
Abstract
We previously identified keratinocyte-associated protein 3, Krtcap3, as an obesity-related gene in female rats where a whole body Krtcap3 knockout (KO) led to increased adiposity compared to wild-type (WT) controls when fed a high-fat diet (HFD). We sought to replicate this work to better understand the function of Krtcap3 but were unable to reproduce the adiposity phenotype. In the current work, WT female rats ate more compared to WT in the prior study, with corresponding increases in body weight and fat mass, while there were no changes in these measures in KO females between the studies. The prior study was conducted before the COVID-19 pandemic, while the current study started after initial lockdown orders and was completed during the pandemic in a generally less stressful environment. We hypothesize that the environmental changes impacted stress levels and may explain the failure to replicate our results. Analysis of corticosterone (CORT) at euthanasia showed a significant study-by-genotype interaction where WT had significantly higher CORT relative to KO in study 1, with no differences in study 2. These data suggest that decreasing Krtcap3 expression may alter the environmental stress response to influence adiposity. We also found that KO rats in both studies, but not WT, experienced a dramatic increase in CORT after their cage mate was removed, suggesting a separate connection to social behavioral stress. Future work is necessary to confirm and elucidate the finer mechanisms of these relationships, but these data indicate the possibility of Krtcap3 as a novel stress gene.NEW & NOTEWORTHY Obesity is linked to both genetics and environmental factors such as stress. Krtcap3 has previously been identified as a gene associated with adiposity, and our work here demonstrates that environmental stress may influence the role of Krtcap3 on both food intake and adiposity. Obesity is strongly influenced by stress in humans, so the identification of novel genes that link stress and obesity will greatly advance our understanding of the disease.
Collapse
Affiliation(s)
- Alexandria M Szalanczy
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| | - Gina Giorgio
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| | - Emily Goff
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| | - Osborne Seshie
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| | - Michael Grzybowski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jason Klotz
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Eva E Redei
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Leah C Solberg Woods
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, North Carolina, United States
| |
Collapse
|
2
|
Witt A, Mateska I, Palladini A, Sinha A, Wölk M, Harauma A, Bechmann N, Pamporaki C, Dahl A, Rothe M, Kopaliani I, Adolf C, Riester A, Wielockx B, Bornstein SR, Kroiss M, Peitzsch M, Moriguchi T, Fedorova M, Grzybek M, Chavakis T, Mirtschink P, Alexaki VI. Fatty acid desaturase 2 determines the lipidomic landscape and steroidogenic function of the adrenal gland. SCIENCE ADVANCES 2023; 9:eadf6710. [PMID: 37478183 PMCID: PMC10361602 DOI: 10.1126/sciadv.adf6710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 06/16/2023] [Indexed: 07/23/2023]
Abstract
Corticosteroids regulate vital processes, including stress responses, systemic metabolism, and blood pressure. Here, we show that corticosteroid synthesis is related to the polyunsaturated fatty acid (PUFA) content of mitochondrial phospholipids in adrenocortical cells. Inhibition of the rate-limiting enzyme of PUFA synthesis, fatty acid desaturase 2 (FADS2), leads to perturbations in the mitochondrial lipidome and diminishes steroidogenesis. Consistently, the adrenocortical mitochondria of Fads2-/- mice fed a diet with low PUFA concentration are structurally impaired and corticoid levels are decreased. On the contrary, FADS2 expression is elevated in the adrenal cortex of obese mice, and plasma corticosterone is increased, which can be counteracted by dietary supplementation with the FADS2 inhibitor SC-26192 or icosapent ethyl, an eicosapentaenoic acid ethyl ester. In humans, FADS2 expression is elevated in aldosterone-producing adenomas compared to non-active adenomas or nontumorous adrenocortical tissue and correlates with expression of steroidogenic genes. Our data demonstrate that FADS2-mediated PUFA synthesis determines adrenocortical steroidogenesis in health and disease.
Collapse
Affiliation(s)
- Anke Witt
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Ivona Mateska
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Alessandra Palladini
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Centre Munich at the University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, 85764, Germany
| | - Anupam Sinha
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Akiko Harauma
- School of Life and Environmental Science, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa, 252-5201, Japan
| | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Christina Pamporaki
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Andreas Dahl
- DRESDEN-Concept Genome Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, 01307, Germany
| | | | - Irakli Kopaliani
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Christian Adolf
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, Munich, 80336, Germany
| | - Anna Riester
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, Munich, 80336, Germany
| | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Stefan R. Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Matthias Kroiss
- Department of Internal Medicine IV, University Hospital Munich, Ludwig-Maximilians-Universität München, Munich, 80336, Germany
- Department of Internal Medicine I, Division of Endocrinology and Diabetes, University Hospital, University of Wuerzburg, Wuerzburg, 97080, Germany
| | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Toru Moriguchi
- School of Life and Environmental Science, Azabu University, 1-17-71 Fuchinobe, Sagamihara, Kanagawa, 252-5201, Japan
| | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
| | - Michal Grzybek
- Center of Membrane Biochemistry and Lipid Research, Faculty of Medicine, Technische Universität Dresden, Dresden, 01307, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Centre Munich at the University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, 85764, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Peter Mirtschink
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 01307, Germany
| |
Collapse
|
3
|
Szalanczy AM, Giorgio G, Goff E, Seshie O, Grzybowski M, Klotz J, Geurts AM, Redei EE, Solberg Woods LC. Changes in Environmental Stress over COVID-19 Pandemic Likely Contributed to Failure to Replicate Adiposity Phenotype Associated with Krtcap3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532439. [PMID: 36993361 PMCID: PMC10055176 DOI: 10.1101/2023.03.15.532439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
We previously identified Keratinocyte-associated protein 3, Krtcap3, as an obesity-related gene in female rats where a whole-body Krtcap3 knock-out (KO) led to increased adiposity compared to wild-type (WT) controls when fed a high-fat diet (HFD). We sought to replicate this work to better understand the function of Krtcap3 but were unable to reproduce the adiposity phenotype. In the current work, WT female rats ate more compared to WT in the prior study, with corresponding increases in body weight and fat mass, while there were no changes in these measures in KO females between the studies. The prior study was conducted before the COVID-19 pandemic, while the current study started after initial lock-down orders and was completed during the pandemic with a generally less stressful environment. We hypothesize that the environmental changes impacted stress levels and may explain the failure to replicate our results. Analysis of corticosterone (CORT) at euthanasia showed a significant study by genotype interaction where WT had significantly higher CORT relative to KO in Study 1, with no differences in Study 2. These data suggest that decreasing Krtcap3 expression may alter the environmental stress response to influence adiposity. We also found that KO rats in both studies, but not WT, experienced a dramatic increase in CORT after their cage mate was removed, suggesting a separate connection to social behavioral stress. Future work is necessary to confirm and elucidate the finer mechanisms of these relationships, but these data indicate the possibility of Krtcap3 as a novel stress gene.
Collapse
Affiliation(s)
- Alexandria M Szalanczy
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| | - Gina Giorgio
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| | - Emily Goff
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| | - Osborne Seshie
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| | - Michael Grzybowski
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jason Klotz
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eva E Redei
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leah C Solberg Woods
- Department of Internal Medicine, School of Medicine, Wake Forest University, Winston Salem, NC USA
| |
Collapse
|
4
|
Šimon M, Mikec Š, Morton NM, Atanur SS, Konc J, Horvat S, Kunej T. Genome-wide screening for genetic variants in polyadenylation signal (PAS) sites in mouse selection lines for fatness and leanness. Mamm Genome 2023; 34:12-31. [PMID: 36414820 PMCID: PMC9684942 DOI: 10.1007/s00335-022-09967-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022]
Abstract
Alternative polyadenylation (APA) determines mRNA stability, localisation, translation and protein function. Several diseases, including obesity, have been linked to APA. Studies have shown that single nucleotide polymorphisms in polyadenylation signals (PAS-SNPs) can influence APA and affect phenotype and disease susceptibility. However, these studies focussed on associations between single PAS-SNP alleles with very large effects and phenotype. Therefore, we performed a genome-wide screening for PAS-SNPs in the polygenic mouse selection lines for fatness and leanness by whole-genome sequencing. The genetic variants identified in the two lines were overlapped with locations of PAS sites obtained from the PolyASite 2.0 database. Expression data for selected genes were extracted from the microarray expression experiment performed on multiple tissue samples. In total, 682 PAS-SNPs were identified within 583 genes involved in various biological processes, including transport, protein modifications and degradation, cell adhesion and immune response. Moreover, 63 of the 583 orthologous genes in human have been previously associated with human diseases, such as nervous system and physical disorders, and immune, endocrine, and metabolic diseases. In both lines, PAS-SNPs have also been identified in genes broadly involved in APA, such as Polr2c, Eif3e and Ints11. Five PAS-SNPs within 5 genes (Car, Col4a1, Itga7, Lat, Nmnat1) were prioritised as potential functional variants and could contribute to the phenotypic disparity between the two selection lines. The developed PAS-SNPs catalogue presents a key resource for planning functional studies to uncover the role of PAS-SNPs in APA, disease susceptibility and fat deposition.
Collapse
Affiliation(s)
- Martin Šimon
- grid.8954.00000 0001 0721 6013Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| | - Špela Mikec
- grid.8954.00000 0001 0721 6013Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| | - Nicholas M. Morton
- grid.511172.10000 0004 0613 128XUniversity of Edinburgh, The Queen’s Medical Research Institute, Centre for Cardiovascular Science, Edinburgh, UK
| | - Santosh S. Atanur
- grid.7445.20000 0001 2113 8111Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- grid.4305.20000 0004 1936 7988Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, UK
| | - Janez Konc
- grid.454324.00000 0001 0661 0844Laboratory for Molecular Modeling, National Institute of Chemistry, Ljubljana, Slovenia
| | - Simon Horvat
- grid.8954.00000 0001 0721 6013Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| | - Tanja Kunej
- grid.8954.00000 0001 0721 6013Biotechnical Faculty, Department of Animal Science, University of Ljubljana, Domžale, Slovenia
| |
Collapse
|
5
|
Tsaban G, Bilitzky-Kopit A, Yaskolka Meir A, Zelicha H, Gepner Y, Shelef I, Orr O, Chassidim Y, Sarusi B, Ceglarek U, Stumvoll M, Blüher M, Stampfer MJ, Shai I, Schwarzfuchs D. The Effect of Weight-Loss Interventions on Cervical and Chin Subcutaneous Fat Depots; the CENTRAL Randomized Controlled Trial. Nutrients 2021; 13:nu13113827. [PMID: 34836081 PMCID: PMC8617936 DOI: 10.3390/nu13113827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of cervical and chin subcutaneous adipose tissues (SAT) represent known phenotypes of obesity. We aimed to evaluate the sensitivity of these fat storages to long-term weight-loss directed lifestyle-intervention and to assess their relations to bodily-adiposity, insulin-resistance, and cardiometabolic risk; We randomly assigned 278 participants with abdominal-obesity/dyslipidemia to low-fat or Mediterranean/low-carbohydrate diets +/- physical-activity. All participants underwent an 18 month whole-body magnetic resonance imaging follow-up, from which we assessed cervical and chin SAT-areas; Participants (age = 48 years; 90% men; body-mass-index = 30.9 kg/m2) had an 18-month adherence-rate of 86%. Cervical-SAT and chin-SAT decreased after 6-months (-13.1% and -5.3%, respectively, p < 0.001). After 18-months only cervical-SAT remained decreased compared to baseline (-5%, p < 0.001). Cervical and chin-SAT 18-month changes were associated with changes in weight (r = 0.70, r = 0.66 respectively; <0.001 for both) and visceral-adipose-tissue (VAT; r = 0.35, r = 0.42 respectively; <0.001 for both). After adjustment to VAT, waist-circumference, or weight-changes, chin-SAT 18-month reduction was associated with favorable changes in fasting-glucose (β = 0.10; p = 0.05), HbA1c (β = 0.12; p = 0.03), and homeostasis-model-assessment-of-insulin-resistance (β = 0.12; p = 0.03). Cervical-SAT 18-month reduction was associated with decreased triglycerides (β = 0.16; p = 0.02) and leptin (β = 0.19; p = 0.01) independent of VAT; Cervical and chin-SATs are dynamic fat depots that correspond with weight-loss and are associated with changes in cardiometabolic profile. In long-term, chin-SAT displays a larger rebound compared with cervical-SAT. Chin-SAT accumulation is associated with in insulin-resistance, independent of central obesity. (ClinicalTrials identifier NCT01530724).
Collapse
Affiliation(s)
- Gal Tsaban
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (A.B.-K.); (A.Y.M.); (H.Z.); (O.O.); (I.S.)
- Department of Cardiology, Soroka University Medical Center, Beer-Sheva 84101, Israel
- Correspondence: ; Tel.: +972-8-647-7449/3
| | - Avital Bilitzky-Kopit
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (A.B.-K.); (A.Y.M.); (H.Z.); (O.O.); (I.S.)
| | - Anat Yaskolka Meir
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (A.B.-K.); (A.Y.M.); (H.Z.); (O.O.); (I.S.)
| | - Hila Zelicha
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (A.B.-K.); (A.Y.M.); (H.Z.); (O.O.); (I.S.)
| | - Yftach Gepner
- Department of Epidemiology and Preventive Medicine, Sackler Faculty of Medicine, and Sylvan Adams Sports Institute, School of Public Health, Tel-Aviv University, Tel-Aviv 96678, Israel;
| | - Ilan Shelef
- Division of Clinical Radiology, Soroka University Medical Center, Beer-Sheva 84101, Israel;
| | - Omri Orr
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (A.B.-K.); (A.Y.M.); (H.Z.); (O.O.); (I.S.)
| | - Yoash Chassidim
- Research Center, Soroka University Medical Center, Beer-Sheva 84101, Israel;
| | - Benjamin Sarusi
- Department of Medicine, Nuclear Research Center Negev, Dimona 84190, Israel; (B.S.); (D.S.)
| | - Uta Ceglarek
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany; (U.C.); (M.S.); (M.B.)
| | - Michael Stumvoll
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany; (U.C.); (M.S.); (M.B.)
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, 04103 Leipzig, Germany; (U.C.); (M.S.); (M.B.)
| | - Meir J. Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
| | - Iris Shai
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; (A.B.-K.); (A.Y.M.); (H.Z.); (O.O.); (I.S.)
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
| | - Dan Schwarzfuchs
- Department of Medicine, Nuclear Research Center Negev, Dimona 84190, Israel; (B.S.); (D.S.)
- Emergency Medicine Division, Soroka University Medical Center, Beer-Sheva 84101, Israel
| |
Collapse
|
6
|
Genomic and Non-Genomic Actions of Glucocorticoids on Adipose Tissue Lipid Metabolism. Int J Mol Sci 2021; 22:ijms22168503. [PMID: 34445209 PMCID: PMC8395154 DOI: 10.3390/ijms22168503] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/01/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022] Open
Abstract
Glucocorticoids (GCs) are hormones that aid the body under stress by regulating glucose and free fatty acids. GCs maintain energy homeostasis in multiple tissues, including those in the liver and skeletal muscle, white adipose tissue (WAT), and brown adipose tissue (BAT). WAT stores energy as triglycerides, while BAT uses fatty acids for heat generation. The multiple genomic and non-genomic pathways in GC signaling vary with exposure duration, location (adipose tissue depot), and species. Genomic effects occur directly through the cytosolic GC receptor (GR), regulating the expression of proteins related to lipid metabolism, such as ATGL and HSL. Non-genomic effects act through mechanisms often independent of the cytosolic GR and happen shortly after GC exposure. Studying the effects of GCs on adipose tissue breakdown and generation (lipolysis and adipogenesis) leads to insights for treatment of adipose-related diseases, such as obesity, coronary disease, and cancer, but has led to controversy among researchers, largely due to the complexity of the process. This paper reviews the recent literature on the genomic and non-genomic effects of GCs on WAT and BAT lipolysis and proposes research to address the many gaps in knowledge related to GC activity and its effects on disease.
Collapse
|
7
|
Diabetes Mellitus and Hypertension-A Case of Sugar and Salt? Int J Mol Sci 2020; 21:ijms21155200. [PMID: 32708014 PMCID: PMC7432106 DOI: 10.3390/ijms21155200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/29/2022] Open
Abstract
The majority of patients with diabetes mellitus (DM) have hypertension (HTN). A specific mechanism for the development of HTN in DM has not been described. In the Zucker, Endothel, und Salz (sugar, endothelium, and salt) study (ZEuS), indices of glucose metabolism and of volume regulation are recorded. An analysis of these parameters shows that glucose concentrations interfere with plasma osmolality and that changes in glycemic control have a significant impact on fluid status and blood pressure. The results of this study are discussed against the background of the striking similarities between the regulation of sugar and salt blood concentrations, introducing the view that DM is probably a sodium-retention disorder that leads to a state of hypervolemia.
Collapse
|
8
|
Prolonged use of finasteride-induced gonadal sex steroids alterations, DNA damage and menstrual bleeding in women. Biosci Rep 2020; 40:221928. [PMID: 31967291 PMCID: PMC7007407 DOI: 10.1042/bsr20191434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 01/07/2020] [Accepted: 01/13/2020] [Indexed: 11/30/2022] Open
Abstract
The aim of the present study was to examine the effect of prolonged use of finasteride on serum levels of dihydrotestosterone (DHT), estradiol (E2), progesterone, testosterone and androstenedione in women during the menstrual period. Further, to screen and compare the 5α-reductase activities through the expression of SRD5A1, SRD5A2 and AR gene and to determine the level of VEGF, VKOR and SAA gene expression and DNA damage. A total of 30 Saudi women aged between 25 and 35 years were enrolled in the study. The selected women were divided into two groups. The first group (n = 15) received 5 mg finasteride/day for prolonged period of one year and second group (n = 15) was taken as a healthy control. ELISA technique was used for measuring the serum levels of the targeted hormones, and Comet assay was used for checking the DNA integrity. Our findings revealed significant decrement of DHT, E2, progesterone and androstenedione levels and elevated levels of testosterone in group treated with daily oral doses of 5 mg finasteride/day compared with the control subjects. mRNA expression suggested that finasteride has concrete effects on the gene expression of the selected genes from the treated group in comparison with the control group. In addition, finasteride induced DNA damage, and heavy menstrual bleeding was noted in women treated with finasteride. In conclusion, the present findings revealed that finasteride has adverse health effects in women associated with gonadal sex steroids alterations, DNA damage and heavy menstrual bleeding with no consensus in the treatment of androgenetic alopecia in women.
Collapse
|
9
|
Onvani S, Mortazavi Najafabadi M, Haghighatdoost F, Larijani B, Azadbakht L. Short sleep duration is related to kidney-related biomarkers, but not lipid profile and diet quality in diabetic nephropathy patients. INT J VITAM NUTR RES 2019; 88:39-49. [PMID: 31038033 DOI: 10.1024/0300-9831/a000392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Objectives: Diabetic nephropathy may adversely influence on sleep duration. The quality of diet may also be affected by both sleep duration and diabetes nephropathy. Therefore, lower diet quality in short sleepers-diabetic nephropathy patients might be related to higher metabolic abnormalities. In the present study, we investigated if sleep duration is related to diet quality indices and biochemical markers in diabetic nephropathy patients. Method: This cross-sectional study was conducted among 237 diabetic nephropathy patients, randomly selected from patients attending in the Alzahra University Hospital, Isfahan, Iran. Usual dietary intake was assessed using validated food frequency questionnaire. Diet quality indices (including diet diversity score, dietary energy density, mean adequacy ratio, and nutrient adequacy ratio) were calculated based on standard definitions. Sleep duration was estimated using self-reported nocturnal hours of sleep. Results: Short sleepers (5-6 h) had higher blood urea nitrogen and creatinine compared with those who slept more than 7.5 h (20.26 + 0.23 mg/dl vs. 17.61 + 0.30 mg/dl, P < 0.0001, and 1.98 + 0.27 mg/dl vs. 1.90 + 0.24 mg/dl, P = 0.03, respectively). Serum triglyceride levels were positively correlated with sleep duration (P = 0.02). Diet quality indices were not significantly associated with sleep duration. Conclusion: Higher sleep duration is significantly related to lower kidney-related biomarkers in diabetic nephropathy patients. Diet quality indices were not associated with sleep duration in diabetic nephropathy patients. More longitudinal studies are required to evaluate the associations of sleep duration, diet quality and biochemical markers in diabetic nephropathy patients.
Collapse
Affiliation(s)
- Shokouh Onvani
- 1 Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.,2 Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Fahimeh Haghighatdoost
- 1 Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.,2 Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bagher Larijani
- 4 Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Azadbakht
- 1 Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.,2 Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.,5 Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medicl Sciences, Tehran, Iran.,6 Diabetes Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Papargyri P, Zapanti E, Salakos N, Papargyris L, Bargiota A, Mastorakos G. Links between HPA axis and adipokines: clinical implications in paradigms of stress-related disorders. Expert Rev Endocrinol Metab 2018; 13:317-332. [PMID: 30422016 DOI: 10.1080/17446651.2018.1543585] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION In the human organism, a constant interplay exists between the stress system [which includes the activity of the hypothalamic-pituitary-adrenal (HPA) axis] and the adipose tissue. This interplay is mediated by hormones of the HPA axis such as corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH) and glucocorticoids (GCs) and adipokines secreted by the adipose tissue. AREAS COVERED In this critical review, the bi-directional interactions between HPA axis and the most studied adipokines such as leptin and adiponectin, as well as the pro-inflammatory adipocytokines tumor necrosis factor (TNF) and interleukin (IL) 6 are presented. Furthermore, these interactions are described in normalcy as well as in specific clinical paradigms of stress-related disorders such as eating disorders, hypothalamic amenorrhea, and stress-related endogenous hypercortisolism states. Wherever new therapeutic strategies emerge, they are presented accordingly. EXPERT COMMENTARY Additional research is needed to clarify the mechanisms involved in the interplay between the HPA axis and the adipose tissue. Research should be focused, in particular, on the development of new therapeutic means targeting dysfunctional adipose tissue in stress-related situations.
Collapse
Affiliation(s)
- Panagiota Papargyri
- a Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieion Hospital, School of Medicine , National and Kapodistrian University of Athens , Athens , Greece
| | - Evangelia Zapanti
- b Department of Endocrinology , Alexandra Hospital , Athens , Greece
| | - Nicolaos Salakos
- c Second Department of Obstetrics and Gynecology, Aretaieion Hospital, School of Medicine , National and Kapodistrian University of Athens , Athens , Greece
| | - Loukas Papargyris
- d CRCINA, INSERM, Université de Nantes, Université d'Angers , Angers , France
- e LabEx IGO "Immunotherapy, Graft, Oncology," , Angers , France
| | - Alexandra Bargiota
- f Department of Endocrinology and Metabolic Diseases, Larissa University Hospital, School of Medicine , University of Thessaly , Larissa , Greece
| | - George Mastorakos
- a Unit of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieion Hospital, School of Medicine , National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
11
|
Insulin Regulates Adrenal Steroidogenesis by Stabilizing SF-1 Activity. Sci Rep 2018; 8:5025. [PMID: 29567944 PMCID: PMC5864882 DOI: 10.1038/s41598-018-23298-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/08/2018] [Indexed: 01/21/2023] Open
Abstract
Development of metabolic syndrome is associated with hyperactivity of the HPA axis characterized by elevated levels of circulating adrenal hormones including cortisol and aldosterone. However, the molecular mechanism leading to the dysregulation of the HPA axis is not well elucidated. In this study, we found that insulin regulates adrenal steroidogenesis by increasing the expression and activity of steroidogenic factor 1 (SF-1) both in vitro and in vivo and this insulin effect was partly through inhibition of FoxO1. Specifically, insulin increased the protein and RNA levels of SF-1 and steroidogenic target genes. Further, adrenal SF-1 expression was significantly increased by hyperactivation of insulin signaling in mice. Together with the elevated SF-1 expression in adrenal glands, hyperactivation of insulin signaling led to increased aldosterone and corticosterone levels. On the other hand, suppressing the insulin signaling using streptozotocin markedly reduced the expression of adrenal SF-1 in mice. In addition, overexpression of FoxO1 significantly suppressed SF-1 and its steroidogenic target genes implying that the positive effect of insulin on SF-1 activity might be through suppression of FoxO1 in the adrenal gland. Taken together, these results indicate that insulin regulates adrenal steroidogenesis through coordinated control of SF-1 and FoxO1.
Collapse
|
12
|
|
13
|
Foster MT. So as we worry we weigh: Visible burrow system stress and visceral adiposity. Physiol Behav 2017; 178:151-156. [DOI: 10.1016/j.physbeh.2017.01.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/20/2016] [Accepted: 01/11/2017] [Indexed: 10/20/2022]
|
14
|
Cannarella R, La Vignera S, Condorelli RA, Calogero AE. Glycolipid and Hormonal Profiles in Young Men with Early-Onset Androgenetic Alopecia: A meta-analysis. Sci Rep 2017; 7:7801. [PMID: 28798373 PMCID: PMC5552767 DOI: 10.1038/s41598-017-08528-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/10/2017] [Indexed: 11/25/2022] Open
Abstract
Hormonal and metabolic abnormalities have been reported in men with early-onset androgenetic alopecia (AGA). Although this has been ascribed to the existence of a male polycystic ovary syndrome (PCOS)-equivalent, data on this topic are inconsistent and this syndrome has not been already acknowledged. To evaluate if, already before the age of 35 years, any difference occurs in the glycolipid and hormonal profiles and in the body weight in men with AGA compared to age-matched controls, we performed a comprehensive meta-analysis of all the available observational case-control studies of literature, using MEDLINE, Google Schoolar and Scopus databases. Among 10596 papers retrieved, seven studies were finally included, enrolling a total of 1009 participants. Our findings demonstrate that young men with AGA have a slightly but significantly worse glycolipid profile compared to controls and a hormonal pattern resembling those of women with PCOS, already before the age of 35 years. Therefore, early-onset AGA might represent a phenotypic sign of the male PCOS-equivalent. The acknowledgement of this syndrome would be of importance to prevent the long-term consequences on health in the affected men. The glycolipid profile and the body weight should be monitored in men with AGA starting from the second decade of life.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
15
|
Fedoseeva LA, Klimov LO, Ershov NI, Alexandrovich YV, Efimov VM, Markel AL, Redina OE. Molecular determinants of the adrenal gland functioning related to stress-sensitive hypertension in ISIAH rats. BMC Genomics 2016; 17:989. [PMID: 28105924 PMCID: PMC5249038 DOI: 10.1186/s12864-016-3354-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background The adrenals are known as an important link in pathogenesis of arterial hypertensive disease. The study was directed to the adrenal transcriptome analysis in ISIAH rats with stress-sensitive arterial hypertension and predominant involvement in pathogenesis of the hypothalamic-pituitary-adrenal and sympathoadrenal systems. Results The RNA-Seq approach was used to perform the comparative adrenal transcriptome profiling in hypertensive ISIAH and normotensive WAG rats. Multiple differentially expressed genes (DEGs) related to different biological processes and metabolic pathways were detected. The discussion of the results helped to prioritize the several DEGs as the promising candidates for further studies of the genetic background underlying the stress-sensitive hypertension development in the ISIAH rats. Two of these were transcription factor genes (Nr4a3 and Ppard), which may be related to the predominant activation of the sympathetic-adrenal medullary axis in ISIAH rats. The other genes are known as associated with hypertension and were defined in the current study as DEGs making the most significant contribution to the inter-strain differences. Four of them (Avpr1a, Hsd11b2, Agt, Ephx2) may provoke the hypertension development, and Mpo may contribute to insulin resistance and inflammation in the ISIAH rats. Conclusions The study strongly highlighted the complex nature of the pathogenesis of stress-sensitive hypertension. The data obtained may be useful for identifying the common molecular determinants in different animal models of arterial hypertension, which may be potentially used as therapeutic targets for pharmacological intervention. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3354-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Larisa A Fedoseeva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Leonid O Klimov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Nikita I Ershov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Yury V Alexandrovich
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Vadim M Efimov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation.,Novosibirsk State University, Novosibirsk, Russian Federation
| | - Arcady L Markel
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation.,Novosibirsk State University, Novosibirsk, Russian Federation
| | - Olga E Redina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation.
| |
Collapse
|
16
|
Chen WC, Chen YM, Huang CC, Tzeng YD. Dehydroepiandrosterone Supplementation Combined with Whole-Body Vibration Training Affects Testosterone Level and Body Composition in Mice. Int J Med Sci 2016; 13:730-740. [PMID: 27766021 PMCID: PMC5069407 DOI: 10.7150/ijms.16132] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/19/2016] [Indexed: 11/05/2022] Open
Abstract
Dehydroepiandrosterone (DHEA), the most abundant sex steroid, is primarily secreted by the adrenal gland and a precursor hormone used by athletes for performance enhancement. Whole-body vibration (WBV) is a well-known light-resistance exercise by automatic adaptations to rapid and repeated oscillations from a vibrating platform, which is also a simple and convenient exercise for older adults. However, the potential effects of DHEA supplementation combined with WBV training on to body composition, exercise performance, and hormone regulation are currently unclear. The objective of the study is to investigate the effects of DHEA supplementation combined with WBV training on body composition, exercise performance, and physical fatigue-related biochemical responses and testosterone content in young-adult C57BL/6 mice. In this study, male C57BL/6 mice were divided into four groups (n = 8 per group) for 6-weeks treatment: sedentary controls with vehicle (SC), DHEA supplementation (DHEA, 10.2 mg/kg), WBV training (WBV; 5.6 Hz, 2 mm, 0.13 g), and WBV training with DHEA supplementation (WBV+DHEA; WBV: 5.6 Hz, 2 mm, 0.13 g and DHEA: 10.2 mg/kg). Exercise performance was evaluated by forelimb grip strength and exhaustive swimming time, as well as changes in body composition and anti-fatigue levels of serum lactate, ammonia, glucose, creatine kinase (CK), and blood urea nitrogen (BUN) after a 15-min swimming exercise. In addition, the biochemical parameters and the testosterone content were measured at the end of the experiment. Six-week DHEA supplementation alone significantly increased mice body weight (BW), muscle weight, testosterone level, and glycogen contents (liver and muscle) when compared with SC group. DHEA supplementation alone had no negative impact on all tissue and biochemical profiles, but could not improve exercise performance. However, WBV+DHEA supplementation also significantly decreased BW, testosterone level and glycogen content of liver, as well as serum lactate and ammonia levels after the 15-min swimming exercise when compared with DHEA supplementation alone. Although DHEA supplementation alone had no beneficial effect in the exercise performance of mice, the BW, testosterone level and glycogen content significantly increased. On the other hand, WBV training combined with DHEA decreased the BW gain, testosterone level and glycogen content caused by DHEA supplementation. Therefore, WBV training could inhibit DHEA supplementation to synthesis the testosterone level or may decrease the DHEA supplement absorptive capacity in young-adult mice.
Collapse
Affiliation(s)
- Wen-Chyuan Chen
- Center for General Education, Chang Gung University of Science and Technology, Taoyuan 33301, Taiwan;; Department of Otorhinolaryngology-Head and Neck Surgery, Sleep Center, Linkou-Chang Gung Memorial Hospital, Taoyuan 33301, Taiwan
| | - Yi-Ming Chen
- Center for General Education, Chang Gung University of Science and Technology, Taoyuan 33301, Taiwan;; Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 33301, Taiwan
| | - Chi-Chang Huang
- Graduate Institute of Sports Science, National Taiwan Sport University, Taoyuan 33301, Taiwan
| | - Yen-Dun Tzeng
- Division of General Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, 813 Kaohsiung, Taiwan
| |
Collapse
|
17
|
Abstract
The purpose of this article is to review fundamentals in adrenal gland histophysiology. Key findings regarding the important signaling pathways involved in the regulation of steroidogenesis and adrenal growth are summarized. We illustrate how adrenal gland morphology and function are deeply interconnected in which novel signaling pathways (Wnt, Sonic hedgehog, Notch, β-catenin) or ionic channels are required for their integrity. Emphasis is given to exploring the mechanisms and challenges underlying the regulation of proliferation, growth, and functionality. Also addressed is the fact that while it is now well-accepted that steroidogenesis results from an enzymatic shuttle between mitochondria and endoplasmic reticulum, key questions still remain on the various aspects related to cellular uptake and delivery of free cholesterol. The significant progress achieved over the past decade regarding the precise molecular mechanisms by which the two main regulators of adrenal cortex, adrenocorticotropin hormone (ACTH) and angiotensin II act on their receptors is reviewed, including structure-activity relationships and their potential applications. Particular attention has been given to crucial second messengers and how various kinases, phosphatases, and cytoskeleton-associated proteins interact to ensure homeostasis and/or meet physiological demands. References to animal studies are also made in an attempt to unravel associated clinical conditions. Many of the aspects addressed in this article still represent a challenge for future studies, their outcome aimed at providing evidence that the adrenal gland, through its steroid hormones, occupies a central position in many situations where homeostasis is disrupted, thus highlighting the relevance of exploring and understanding how this key organ is regulated. © 2014 American Physiological Society. Compr Physiol 4:889-964, 2014.
Collapse
Affiliation(s)
- Nicole Gallo-Payet
- Division of Endocrinology, Department of Medicine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, and Centre de Recherche Clinique Étienne-Le Bel of the Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Quebec, Canada
| | | |
Collapse
|
18
|
Cortisol, platelet serotonin content, and platelet activity in patients with major depression and type 2 diabetes: an exploratory investigation. Psychosom Med 2015; 77:145-55. [PMID: 25626989 DOI: 10.1097/psy.0000000000000145] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Hypothalamic-pituitary-adrenal system dysfunction, serotonergic system alterations, and enhanced platelet activity may contribute to the increased cardiac risk in depression. This exploratory study examined associations between cortisol parameters, platelet serotonin (5-HT) content, and platelet activity markers in patients with newly diagnosed major depression (MD) and/or Type 2 diabetes (T2DM) compared with healthy controls. METHODS We compared cortisol awakening response (CAR), diurnal decrease in salivary cortisol concentrations (slope), platelet 5-HT, and platelet markers (CD40, CD40 ligand [CD40L], soluble CD40L, CD62P, β-thromboglobulin, and platelet factor-4) in 22 T2DM patients, 20 MD patients, 18 T2DM patients with MD, and 24 healthy controls. RESULTS Platelet markers were elevated in MD (F(6,60) = 11.14, p < .001) and T2DM (F(6,60) = 13.07, p < .001). Subgroups did not differ in 5-HT or cortisol slope, whereas T2DM patients without depression had significantly lower CAR than did healthy controls (F(1,61) = 7.46, p = .008). In healthy controls, cortisol slope correlated with platelet activity for CD40 (r = -0.43, p = .048) and 5-HT was correlated with CD40L (r = 0.53, p = .007). In patients with both T2DM and MD, 5-HT and CD62P were correlated (r = 0.52, p = .033). CONCLUSIONS Increased platelet activity in T2DM and MD may play a role in the association between diabetes, depression, and coronary artery disease. The present data suggest that group differences in cortisol or 5-HT as well as group-specific associations of cortisol or 5-HT with platelet markers might be of limited importance in the shared pathways of T2DM and depression in the pathophysiology of coronary artery disease.
Collapse
|
19
|
Fokidis HB, Adomat HH, Kharmate G, Hosseini-Beheshti E, Guns ES, Soma KK. Regulation of local steroidogenesis in the brain and in prostate cancer: lessons learned from interdisciplinary collaboration. Front Neuroendocrinol 2015; 36:108-29. [PMID: 25223867 DOI: 10.1016/j.yfrne.2014.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 11/16/2022]
Abstract
Sex steroids play critical roles in the regulation of the brain and many other organs. Traditionally, researchers have focused on sex steroid signaling that involves travel from the gonads via the circulation to intracellular receptors in target tissues. This classic concept has been challenged, however, by the growing number of cases in which steroids are synthesized locally and act locally within diverse tissues. For example, the brain and prostate carcinoma were previously considered targets of gonadal sex steroids, but under certain circumstances, these tissues can upregulate their steroidogenic potential, particularly when circulating sex steroid concentrations are low. We review some of the similarities and differences between local sex steroid synthesis in the brain and prostate cancer. We also share five lessons that we have learned during the course of our interdisciplinary collaboration, which brought together neuroendocrinologists and cancer biologists. These lessons have important implications for future research in both fields.
Collapse
Affiliation(s)
- H Bobby Fokidis
- Department of Biology, Rollins College, Winter Park, FL 37289, USA; Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada.
| | - Hans H Adomat
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada
| | | | | | - Emma S Guns
- Vancouver Prostate Centre, Vancouver, BC V6H 3Z6, Canada; Department of Urological Sciences, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Kiran K Soma
- Department of Psychology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Brain Research Centre, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
20
|
Changes in morphology and function of adrenal cortex in mice fed a high-fat diet. Int J Obes (Lond) 2014; 39:321-30. [PMID: 24919565 DOI: 10.1038/ijo.2014.102] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 05/09/2014] [Accepted: 05/15/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND/OBJECTIVES Obesity is a major risk factor for the development of type 2 diabetes and other debilitating diseases. Obesity and diabetes are intimately linked with altered levels of adrenal steroids. Elevated levels of these hormones induce insulin resistance and cause cardiovascular diseases. The mechanisms underlying obesity-related alterations in adrenal steroids are still not well understood. Here, we investigated how diet-induced obesity affects the morphology and function of the mouse adrenal cortex. METHODS We fed animals either a high-fat diet (HFD) or a normal diet (60% kcal from fat or 10% kcal from fat, respectively) for 18 weeks. We then assessed various aspects of adrenal gland morphology and function, as well as basal plasma concentrations of steroid hormones and ACTH. RESULTS We show that adrenal glands of mice fed a HFD release more corticosterone and aldosterone, resulting in higher plasma levels. This increase is driven by adrenal cortical hyperplasia, and by increased expression of multiple genes involved in steroidogenesis. We demonstrate that diet-induced obesity elevates Sonic hedgehog signaling in Gli1-positive progenitors, which populate the adrenal capsule and give rise to the steroidogenic cells of the adrenal cortex. Feeding animals with a HFD depletes Gli1-positive progenitors, as the adrenal cortex expands. CONCLUSIONS This work provides insight into how diet-induced obesity changes the biology of the adrenal gland. The association of these changes with increased Shh signaling suggests possible therapeutic strategies for obesity-related steroid hormone dysfunction.
Collapse
|
21
|
Ou CY, Chen TC, Lee JV, Wang JC, Stallcup MR. Coregulator cell cycle and apoptosis regulator 1 (CCAR1) positively regulates adipocyte differentiation through the glucocorticoid signaling pathway. J Biol Chem 2014; 289:17078-86. [PMID: 24811171 DOI: 10.1074/jbc.m114.548081] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glucocorticoids contribute to adipocyte differentiation by cooperating with transcription factors, such as CCAAT/enhancer-binding protein β (C/EBPβ), to stimulate transcription of the gene encoding peroxisome proliferator-activated receptor (PPARγ), a master regulator of adipogenesis. However, the mechanism of PPARγ gene regulation by glucocorticoids, the glucocorticoid receptor (GR), and its coregulators is poorly understood. Here we show that two GR binding regions (GBRs) in the mouse PPARγ gene were responsive to glucocorticoid, and treatment of 3T3-L1 preadipocytes with glucocorticoid alone induced GR occupancy and chromatin remodeling at PPARγ GBRs, which also contain binding sites for C/EBP and PPARγ proteins. GR recruited cell cycle and apoptosis regulator 1 (CCAR1), a transcription coregulator, to the PPARγ gene GBRs. Notably, CCAR1 was required for GR occupancy and chromatin remodeling at one of the PPARγ gene GBRs. Moreover, depletion of CCAR1 markedly suppressed differentiation of mouse mesenchymal stem cells and 3T3-L1 preadipocytes to mature adipocytes and decreased induction of PPARγ, C/EBPα, and C/EBPδ. Although CCAR1 was required for stimulation of several GR-regulated adipogenic genes in 3T3-L1 preadipocytes by glucocorticoid, it was not required for GR-activated transcription of certain anti-inflammatory genes in human A549 lung epithelial cells. Overall, our results highlighted the novel and specific roles of GR and CCAR1 in adipogenesis.
Collapse
Affiliation(s)
- Chen-Yin Ou
- From the Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California 90089 and
| | - Tzu-Chieh Chen
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California 94720
| | - Joyce V Lee
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California 94720
| | - Jen-Chywan Wang
- Department of Nutritional Science and Toxicology, University of California, Berkeley, California 94720
| | - Michael R Stallcup
- From the Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California 90089 and
| |
Collapse
|
22
|
Zhang BL, Song LX, Li YF, Li YL, Guo YZ, Zhang E, Liu HM. Synthesis and biological evaluation of dehydroepiandrosterone-fused thiazole, imidazo[2,1-b]thiazole, pyridine steroidal analogues. Steroids 2014; 80:92-101. [PMID: 24355392 DOI: 10.1016/j.steroids.2013.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/02/2013] [Accepted: 12/09/2013] [Indexed: 11/24/2022]
Abstract
A series of steroidal[17,16-d]thiazole, steroidal[1,2-b]pyridine and steroidal[17,16-d]thiazole[2,1-b]imidazo products were synthesized through a convenient and productive method. Anti-proliferation activity against EC109 (human esophageal carcinoma), EC9706 (human esophageal carcinoma) and MGC-803 (human gastric carcinoma) cell lines was examined in vitro. Among the screened compounds, several highly potential compounds were located.
Collapse
Affiliation(s)
- Bao-Le Zhang
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China
| | - Li-Xing Song
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ya-Fei Li
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China
| | - Yi-Lei Li
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China
| | - Ya-Zhuo Guo
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China
| | - En Zhang
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China
| | - Hong-Min Liu
- School of Pharmaceutical Sciences and New Drug Research & Development Center, Zhengzhou University, Zhengzhou 450001, PR China.
| |
Collapse
|
23
|
de Oliveira C, Scarabelot VL, de Souza A, de Oliveira CM, Medeiros LF, de Macedo IC, Marques Filho PR, Cioato SG, Caumo W, Torres ILS. Obesity and chronic stress are able to desynchronize the temporal pattern of serum levels of leptin and triglycerides. Peptides 2014; 51:46-53. [PMID: 24184591 DOI: 10.1016/j.peptides.2013.10.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/21/2013] [Accepted: 10/21/2013] [Indexed: 12/20/2022]
Abstract
Disruption of the circadian system can lead to metabolic dysfunction as a response to environmental alterations. This study assessed the effects of the association between obesity and chronic stress on the temporal pattern of serum levels of adipogenic markers and corticosterone in rats. We evaluated weekly weight, delta weight, Lee index, and weight fractions of adipose tissue (mesenteric, MAT; subcutaneous, SAT; and pericardial, PAT) to control for hypercaloric diet-induced obesity model efficacy. Wistar rats were divided into four groups: standard chow (C), hypercaloric diet (HD), stress plus standard chow (S), and stress plus hypercaloric diet (SHD), and analyzed at three time points: ZT0, ZT12, and ZT18. Stressed animals were subjected to chronic stress for 1h per day, 5 days per week, during 80 days. The chronic exposure to a hypercaloric diet was an effective model for the induction of obesity and metabolic syndrome, increasing delta weight, Lee index, weight fractions of adipose tissue, and triglycerides and leptin levels. We confirmed the presence of a temporal pattern in the release of triglycerides, corticosterone, leptin, and adiponectin in naïve animals. Chronic stress reduced delta weight, MAT weight, and levels of triglycerides, total cholesterol, and leptin. There were interactions between chronic stress and obesity and serum total cholesterol levels, between time points and obesity and adiponectin and corticosterone levels, and between time points and chronic stress and serum leptin levels. In conclusion, both parameters were able to desynchronize the temporal pattern of leptin and triglyceride release, which could contribute to the development of metabolic diseases such as obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Carla de Oliveira
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Post Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Vanessa Leal Scarabelot
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil; Post Graduate Program in Biological Sciences - Physiology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil
| | - Andressa de Souza
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Post Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil; Laboratório de Bioquímica, Centro de Ciências Básicas da Saúde, Centro Universitário Univates, Lajeado, RS 95900-000, Brazil
| | - Cleverson Moraes de Oliveira
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Post Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Liciane Fernandes Medeiros
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil; Post Graduate Program in Biological Sciences - Physiology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil
| | - Isabel Cristina de Macedo
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil; Post Graduate Program in Biological Sciences - Physiology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil
| | - Paulo Ricardo Marques Filho
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Post Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Stefania Giotti Cioato
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Post Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil
| | - Wolnei Caumo
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Post Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Iraci L S Torres
- Pain Pharmacology and Neuromodulation, Animal Models Laboratory, Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil; Post Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil; Animal Experimentation Unit and Graduate Research Group, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS 90035-003, Brazil; Post Graduate Program in Biological Sciences - Physiology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS 90050-170, Brazil.
| |
Collapse
|
24
|
Swierczynska MM, Lamounier-Zepter V, Bornstein SR, Eaton S. Lipoproteins and Hedgehog signalling--possible implications for the adrenal gland function. Eur J Clin Invest 2013; 43:1178-83. [PMID: 23992253 DOI: 10.1111/eci.12145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 07/27/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Metabolic syndrome is a common metabolic disorder that is associated with an increased risk of type 2 diabetes and cardiovascular diseases. Disturbances in adrenal steroid hormone production significantly contribute to the development of this disorder. Therefore, it is extremely important to fully understand the mechanisms governing adrenal gland function, both in physiological and pathological conditions. RESULTS Recently, Sonic hedgehog has emerged as an important regulator of adrenal development, with a possible role in adult gland homeostasis. Recent work of our group shows that lipoproteins are important regulators of Hedgehog signaling; they act as carriers for the spread of Hedgehog proteins, but also contain lipid(s) that inhibit the pathway. CONCLUSIONS We propose that lipoproteins may affect Sonic hedgehog signaling in the adult adrenal gland at multiple levels. Understanding the interplay between lipoprotein metabolism and adrenal Hedgehog signaling may improve our understanding of how adrenal gland disorders contribute to the metabolic syndrome.
Collapse
Affiliation(s)
- Marta M Swierczynska
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | | | | |
Collapse
|
25
|
Vasiljević A, Veličković N, Bursać B, Djordjevic A, Milutinović DV, Nestorović N, Matić G. Enhanced prereceptor glucocorticoid metabolism and lipogenesis impair insulin signaling in the liver of fructose-fed rats. J Nutr Biochem 2013; 24:1790-7. [DOI: 10.1016/j.jnutbio.2013.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/18/2013] [Accepted: 04/01/2013] [Indexed: 12/19/2022]
|
26
|
Li P, Pan F, Hao Y, Feng W, Song H, Zhu D. SGK1 is regulated by metabolic-related factors in 3T3-L1 adipocytes and overexpressed in the adipose tissue of subjects with obesity and diabetes. Diabetes Res Clin Pract 2013; 102:35-42. [PMID: 24035040 DOI: 10.1016/j.diabres.2013.08.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 07/05/2013] [Accepted: 08/23/2013] [Indexed: 12/26/2022]
Abstract
AIMS The present study aimed to investigate the pathophysiological role of SGK1 in the development of metabolic syndrome by investigating the expression and regulation of serum and glucocorticoid-inducible kinase 1 (SGK1) in adipose tissues in obesity and diabetes. METHODS SGK1 expression in adipose tissue was investigated using reverse transcription polymerase chain reaction (RT-PCR) and immunohistochemistry. SGK1 regulation in differentiated 3T3-L1 adipocytes by metabolic-related factors was assessed using Northern blot analysis. Humans with obesity and type 2 diabetes and KKAy and db/db mice were used to evaluate SGK1 expression in the adipose tissue of subjects with obesity and diabetes using quantitative real-time PCR and Western blot analysis. RESULTS SGK1 was expressed in white adipose tissue as shown by mRNA and protein levels. Aldosterone and glucocorticoids stimulated SGK1 expression in a time- and dose-dependent manner, whereas PPAR-γ agonists inhibited SGK1 expression in differentiated 3T3-L1 adipocytes. Furthermore, SGK1 mRNA and protein were overexpressed in the adipose tissue of mice and humans with obesity and type 2 diabetes. CONCLUSION Aldosterone, glucocorticoids and other factors contribute to excessive SGK1 expression in adipose tissue. This excessive SGK1 expression may be related to adipose tissue dysfunction, which may contribute to the development of obesity, diabetes and metabolic syndrome.
Collapse
Affiliation(s)
- Ping Li
- Department of Endocrinology, Drum Tower Hospital Affiliated to Nanjing University Medical School, Nanjing, PR China
| | | | | | | | | | | |
Collapse
|
27
|
Cherkasova OP, Seliatitskaia VG. [Adrenocortical and renin-angiotensin systems in dynamics of experimental diabetes]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2013; 59:183-91. [PMID: 23789345 DOI: 10.18097/pbmc20135902183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Components of the adrenocortical system (adrenal and blood corticosteroid hormones and hepatic and renal 11beta-hydroxysteroid dehydrogenase activity) and also activity of the most important enzyme of the renin-angiotensin system, tissue and blood angiotensin converting enzyme (ACE) have been investigated in dynamics of alloxan diabetes. The study has shown that the initial period of diabetes is characterized by activation of synthesis and secretion of adrenocortical hormones into blood. High blood glucose and glucocorticoid honnones increase activity of the renin-angiotensin system in lungs and decrease ACE secretion into blood. This is accompanied by a decrease of activity of the renin-angiotensin system in kidneys. Subsequent progression of diabetes resulted in impairments of physiologically determined correlations between the components of these systems. Development of experimental diabetes for 30 days was accompanied by sign of a decrease of the adrenal glucocorticoid function regardless of stable impairments of carbohydrate metabolism. Under these conditions increased adrenal and hepatic 11beta-hydroxysteroid dehydrogenase activity may be responsible for maintenance of elevated levels of the main glucocorticoid in blood and tissues. Factor analysis revealed impairments in intersystem relationships between the adrenocortical and renin-angiotensin systems in experimental diabetes thus suggesting disintegration of regulatory systems.
Collapse
|
28
|
Fu L, Kettner NM. The circadian clock in cancer development and therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 119:221-82. [PMID: 23899600 PMCID: PMC4103166 DOI: 10.1016/b978-0-12-396971-2.00009-9] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Most aspects of mammalian function display circadian rhythms driven by an endogenous clock. The circadian clock is operated by genes and comprises a central clock in the brain that responds to environmental cues and controls subordinate clocks in peripheral tissues via circadian output pathways. The central and peripheral clocks coordinately generate rhythmic gene expression in a tissue-specific manner in vivo to couple diverse physiological and behavioral processes to periodic changes in the environment. However, with the industrialization of the world, activities that disrupt endogenous homeostasis with external circadian cues have increased. This change in lifestyle has been linked to an increased risk of diseases in all aspects of human health, including cancer. Studies in humans and animal models have revealed that cancer development in vivo is closely associated with the loss of circadian homeostasis in energy balance, immune function, and aging, which are supported by cellular functions important for tumor suppression including cell proliferation, senescence, metabolism, and DNA damage response. The clock controls these cellular functions both locally in cells of peripheral tissues and at the organismal level via extracellular signaling. Thus, the hierarchical mammalian circadian clock provides a unique system to study carcinogenesis as a deregulated physiological process in vivo. The asynchrony between host and malignant tissues in cell proliferation and metabolism also provides new and exciting options for novel anticancer therapies.
Collapse
Affiliation(s)
- Loning Fu
- Department of Pediatrics/U.S. Department of Agriculture/Agricultural Research Service/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Nicole M. Kettner
- Department of Pediatrics/U.S. Department of Agriculture/Agricultural Research Service/Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
29
|
Wan Mahmood WA, Draman Yusoff MS, Behan LA, Di Perna A, Kyaw Tun T, McDermott J, Sreenan S. Association between Sleep Disruption and Levels of Lipids in Caucasians with Type 2 Diabetes. Int J Endocrinol 2013; 2013:341506. [PMID: 24072996 PMCID: PMC3773446 DOI: 10.1155/2013/341506] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 07/22/2013] [Indexed: 11/17/2022] Open
Abstract
Aim. To investigate the association between sleep quality and duration with lipid and glycaemic control in Caucasian subjects with type 2 diabetes. Methods. Sleep quality was assessed using the Pittsburgh Sleep Quality Index (PSQI) in 114 type 2 diabetes (T2DM) subjects. Comparisons were made between subjects with different sleep quality and sleep duration. Hierarchical multiple regression analyses were used to determine contributors to metabolic parameters. Results. Subjects with poor sleep quality (PQ; PSQI ≥ 6) had higher systolic blood pressure, glycated haemoglobin, urine albumin : creatinine ratio (UAC), total cholesterol (TC), and triglycerides (TG) (P < 0.05 for all) compared to those with good sleep quality (GQ; PSQI ≤ 5). Long sleep duration (LSD) subjects had higher TC and short sleep duration (SSD) subjects had higher TG compared to those with medium sleep duration. Sleep duration and PSQI score were independent predictors of TC and low-density lipoprotein cholesterol (LDL), contributing to 14.0% and 6.1% of the total variance, respectively. Conclusions. In this Caucasian T2DM population, PQ is associated with adverse cardiovascular risk markers, and long and short sleep disruptions have an independent negative impact on lipids. Sleep assessment should be included as part of a diabetes clinic review.
Collapse
Affiliation(s)
- Wan Aizad Wan Mahmood
- Department of Endocrinology and Diabetes, Royal College of Surgeons in Ireland, Connolly Hospital, Blanchardstown, Dublin 15, Ireland
| | - Mohd Shazli Draman Yusoff
- Department of Endocrinology and Diabetes, Royal College of Surgeons in Ireland, Connolly Hospital, Blanchardstown, Dublin 15, Ireland
| | - Lucy Ann Behan
- Department of Endocrinology and Diabetes, Royal College of Surgeons in Ireland, Connolly Hospital, Blanchardstown, Dublin 15, Ireland
| | - Andrea Di Perna
- Department of Endocrinology and Diabetes, Royal College of Surgeons in Ireland, Connolly Hospital, Blanchardstown, Dublin 15, Ireland
| | - Tommy Kyaw Tun
- Department of Endocrinology and Diabetes, Royal College of Surgeons in Ireland, Connolly Hospital, Blanchardstown, Dublin 15, Ireland
| | - John McDermott
- Department of Endocrinology and Diabetes, Royal College of Surgeons in Ireland, Connolly Hospital, Blanchardstown, Dublin 15, Ireland
| | - Seamus Sreenan
- Department of Endocrinology and Diabetes, Royal College of Surgeons in Ireland, Connolly Hospital, Blanchardstown, Dublin 15, Ireland
- *Seamus Sreenan:
| |
Collapse
|
30
|
Macedo IC, Medeiros LF, Oliveira C, Oliveira CM, Rozisky JR, Scarabelot VL, Souza A, Silva FR, Santos VS, Cioato SG, Caumo W, Torres ILS. Cafeteria diet-induced obesity plus chronic stress alter serum leptin levels. Peptides 2012; 38:189-96. [PMID: 22940203 DOI: 10.1016/j.peptides.2012.08.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 08/10/2012] [Accepted: 08/10/2012] [Indexed: 12/19/2022]
Abstract
Obesity is a disease that has become a serious public health issue worldwide, and chronic stressors, which are a problem for modern society, cause neuroendocrine changes with alterations in food intake. Obesity and chronic stress are associated with the development of cardiovascular diseases and metabolic disorders. In this study, a rat model was used to evaluate the effects of a hypercaloric diet plus chronic restraint stress on the serum leptin and lipids levels and on the weight of specific adipose tissue (mesenteric, MAT; subcutaneous, SAT and visceral, VAT). Wistar rats were divided into the following 4 groups: standard chow (C), hypercaloric diet (HD), stress plus standard chow (S), and stress plus hypercaloric diet (SHD). The animals in the stress groups were subjected to chronic stress (placed inside a 25 cm × 7 cm plastic tube for 1h per day, 5 days per week for 6 weeks). The following parameters were evaluated: the weight of the liver, adrenal glands and specific adipose tissue; the delta weight; the Lee index; and the serum levels of leptin, corticosterone, glucose, total cholesterol, and triglycerides. The hypercaloric diet induced obesity in rats, increasing the Lee index, weight, leptin, triglycerides, and cholesterol levels. The stress decreased weight gain even in animals fed a hypercaloric diet but did not prevent a significant increase in the Lee index. However, an interaction between the independent factors (hypercaloric diet and stress) was observed, which is demonstrated by the increased serum leptin levels in the animals exposed to both protocols.
Collapse
Affiliation(s)
- I C Macedo
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul Institute of Basic Health Sciences, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Cordoba-Chacon J, Gahete MD, Pozo-Salas AI, Moreno-Herrera A, Castaño JP, Kineman RD, Luque RM. Peripubertal-onset but not adult-onset obesity increases IGF-I and drives development of lean mass, which may lessen the metabolic impairment in adult obesity. Am J Physiol Endocrinol Metab 2012; 303:E1151-7. [PMID: 22932784 PMCID: PMC3774069 DOI: 10.1152/ajpendo.00340.2012] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
It has been suggested that adult metabolic dysfunction may be more severe in individuals who become obese as children compared with those who become obese later in life. To determine whether adult metabolic function differs if diet-induced weight gain occurs during the peripubertal age vs. if excess weight gain occurs after puberty, male C57Bl/6J mice were fed a low-fat (LF; 10% kcal from fat) or high-fat (HF; 60% kcal from fat) diet starting during the peripubertal period (pHF; 4 wk of age) or as adults (aHF; 12 wk of age). Both pHF and aHF mice were hyperinsulinemic and hyperglycemic, and both showed impaired glucose tolerance and insulin resistance compared with their LF-fed controls. However, despite a longer time on diet, pHF mice were relatively more insulin sensitive than aHF mice, which was associated with higher lean mass and circulating IGF-I levels. In addition, HF feeding had an overall stimulatory effect on circulating corticosterone levels; however, this rise was associated only with elevated plasma ACTH in the aHF mice. Despite the belief that adult metabolic dysfunction may be more severe in individuals who become obese as children, data generated using a diet-induced obese mouse model suggest that adult metabolic dysfunction associated with peripubertal onset of obesity is not worse than that associated with adult-onset obesity.
Collapse
Affiliation(s)
- Jose Cordoba-Chacon
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba/Hospital Universitario Reina Sofia, University of Cordoba Centro de Investigacion Biomedica en Red Fisiopatologia de Obesidad y Nutricion, Córdoba, Spain
| | | | | | | | | | | | | |
Collapse
|
32
|
Atalar F, Gormez S, Caynak B, Akan G, Tanriverdi G, Bilgic-Gazioglu S, Gunay D, Duran C, Akpinar B, Ozbek U, Buyukdevrim AS, Yazici Z. The role of mediastinal adipose tissue 11β-hydroxysteroid d ehydrogenase type 1 and glucocorticoid expression in the development of coronary atherosclerosis in obese patients with ischemic heart disease. Cardiovasc Diabetol 2012; 11:115. [PMID: 23009206 PMCID: PMC3515420 DOI: 10.1186/1475-2840-11-115] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 09/10/2012] [Indexed: 11/12/2022] Open
Abstract
Background Visceral fat deposition and its associated atherogenic complications are mediated by glucocorticoids. Cardiac visceral fat comprises mediastinal adipose tissue (MAT) and epicardial adipose tissue (EAT), and MAT is a potential biomarker of risk for obese patients. Aim Our objective was to evaluate the role of EAT and MAT 11beta-hydroxysteroid dehydrogenase type 1 (11β-HSD-1) and glucocorticoid receptor (GCR) expression in comparison with subcutaneous adipose tissue (SAT) in the development of coronary atherosclerosis in obese patients with coronary artery disease (CAD), and to assess their correlations with CD68 and fatty acids from these tissues. Methods and results Expression of 11β-HSD-1 and GCR was measured by qRT-PCR in EAT, MAT and SAT of thirty-one obese patients undergoing coronary artery bypass grafting due to CAD (obese CAD group) and sixteen obese patients without CAD undergoing heart valve surgery (controls). 11β-HSD-1 and GCR expression in MAT were found to be significantly increased in the obese CAD group compared with controls (p < 0.05). In the obese CAD group, 11β-HSD-1 and GCR mRNA levels were strongly correlated in MAT. Stearidonic acid was significantly increased in EAT and MAT of the obese CAD group and arachidonic acid was significantly expressed in MAT of the obese male CAD group (p < 0.05). Conclusions We report for the first time the increased expression of 11β-HSD-1 and GCR in MAT compared with EAT and SAT, and also describe the interrelated effects of stearidonic acid, HOMA-IR, plasma cortisol and GCR mRNA levels, explaining 40.2% of the variance in 11β-HSD-1 mRNA levels in MAT of obese CAD patients. These findings support the hypothesis that MAT contributes locally to the development of coronary atherosclerosis via glucocorticoid action.
Collapse
Affiliation(s)
- Fatmahan Atalar
- Department Growth-Development and Pediatric Endocrinology, Child Health Institute, Istanbul University, Istanbul, Turkey.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Garlic (Allium sativum) down-regulates the expression of angiotensin II AT1 receptor in adrenal and renal tissues of streptozotocin-induced diabetic rats. Inflammopharmacology 2012; 21:147-59. [DOI: 10.1007/s10787-012-0139-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 05/16/2012] [Indexed: 01/11/2023]
|
34
|
Donner NC, Montoya CD, Lukkes JL, Lowry CA. Chronic non-invasive corticosterone administration abolishes the diurnal pattern of tph2 expression. Psychoneuroendocrinology 2012; 37:645-61. [PMID: 21924839 PMCID: PMC3249349 DOI: 10.1016/j.psyneuen.2011.08.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Revised: 07/20/2011] [Accepted: 08/22/2011] [Indexed: 12/25/2022]
Abstract
Both hypothalamic-pituitary-adrenal (HPA) axis activity and serotonergic systems are commonly dysregulated in stress-related psychiatric disorders. We describe here a non-invasive rat model for hypercortisolism, as observed in major depression, and its effects on physiology, behavior, and the expression of tph2, the gene encoding tryptophan hydroxylase 2, the rate-limiting enzyme for brain serotonin (5-hydroxytryptamine; 5-HT) synthesis. We delivered corticosterone (40 μg/ml, 100 μg/ml or 400 μg/ml) or vehicle to adrenal-intact adult, male rats via the drinking water for 3 weeks. On days 15, 16, 17 and 18, respectively, the rats' emotionality was assessed in the open-field (OF), social interaction (SI), elevated plus-maze (EPM), and forced swim tests (FST). On day 21, half of the rats in each group were killed 2h into the dark phase of a 12/12 h reversed light/dark cycle; the other half were killed 2h into the light phase. We then measured indices of HPA axis activity, plasma glucose and interleukin-6 (IL-6) availability, and neuronal tph2 expression at each time point. Chronic corticosterone intake was sufficient to cause increased anxiety- and depressive-like behavior in a dose-dependent manner. It also disrupted the diurnal pattern of plasma adrenocorticotropin (ACTH), corticosterone, and glucose concentrations, caused adrenal atrophy, and prevented regular weight gain. No diurnal or treatment-dependent changes were found for plasma concentrations of IL-6. Remarkably, all doses of corticosterone treatment abolished the diurnal variation of tph2 mRNA expression in the brainstem dorsal raphe nucleus (DR) by elevating the gene's expression during the animals' inactive (light) phase. Our data demonstrate that chronic elevation of corticosterone creates a vulnerability to a depression-like syndrome that is associated with increased tph2 expression, similar to that observed in depressed patients.
Collapse
Affiliation(s)
- Nina C Donner
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO 80309-0354, USA.
| | | | | | | |
Collapse
|
35
|
Reduced serotonin reuptake transporter (SERT) function causes insulin resistance and hepatic steatosis independent of food intake. PLoS One 2012; 7:e32511. [PMID: 22412882 PMCID: PMC3297606 DOI: 10.1371/journal.pone.0032511] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 01/27/2012] [Indexed: 01/06/2023] Open
Abstract
Serotonin reuptake transporter (SERT) is a key regulator of serotonin neurotransmission and a major target of antidepressants. Antidepressants, such as selectively serotonin reuptake inhibitors (SSRIs), that block SERT function are known to affect food intake and body weight. Here, we provide genetic evidence that food intake and metabolism are regulated by separable mechanisms of SERT function. SERT-deficient mice ate less during both normal diet and high fat diet feeding. The reduced food intake was accompanied with markedly elevated plasma leptin levels. Despite reduced food intake, SERT-deficient mice exhibited glucose intolerance and insulin resistance, and progressively developed obesity and hepatic steatosis. Several lines of evidence indicate that the metabolic deficits of SERT-deficient mice are attributable to reduced insulin-sensitivity in peripheral tissues. First, SERT-deficient mice exhibited beta-cell hyperplasia and islet-mass expansion. Second, biochemical analyses revealed constitutively elevated JNK activity and diminished insulin-induced AKT activation in the liver of SERT-deficient mice. SERT-deficient mice exhibited hyper-JNK activity and hyperinsulinemia prior to the development of obesity. Third, enhancing AKT signaling by PTEN deficiency corrected glucose tolerance in SERT-deficient mice. These findings have potential implications for designing selective SERT drugs for weight control and the treatment of metabolic syndromes.
Collapse
|
36
|
Mattei J, Noel SE, Tucker KL. A meat, processed meat, and French fries dietary pattern is associated with high allostatic load in Puerto Rican older adults. JOURNAL OF THE AMERICAN DIETETIC ASSOCIATION 2011; 111:1498-506. [PMID: 21963016 PMCID: PMC3185297 DOI: 10.1016/j.jada.2011.07.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 03/01/2011] [Indexed: 01/09/2023]
Abstract
BACKGROUND Consumption of certain dietary patterns, such as a Western diet, has been associated with unfavorable physiologic outcomes. Diet has been proposed as a contributor to allostatic load, a composite measure of physiological dysregulation. OBJECTIVE To determine the association of dietary patterns, defined by "meat and french fries," "traditional Puerto Rican foods" (rice, beans, and oils), or "sweets," with allostatic load, and with the 10 individual physiologic parameters that comprise it. PARTICIPANTS Baseline data collected from participants of the Boston Puerto Rican Health Study (n=1,117; aged 45 to 75 years) was used to run linear and logistic regression models, adjusting for age, sex, alcohol intake, smoking, medications, energy intake, and body mass index or physical activity. RESULTS Significant trends across increasing quintiles of the meat and french fries pattern were observed for higher allostatic load score (P=0.002), waist circumference (P=0.032), systolic blood pressure (P=0.008), and diastolic blood pressure (P<0.0001). Participants in the highest quintile of the meat and french fries pattern had significantly higher allostatic load score than those in the lowest quintile (mean 4.3±0.11 vs 3.9±0.12, P=0.030), and had higher odds of having high allostatic load (odds ratio [95% confidence interval]: 1.8 [1.2 to 2.9]), low dehydroepiandrosterone sulfate (odds ratio 1.9 [95% confidence interval: 1.2 to 3.1]), and high glycosylated hemoglobin (odds ratio 1.7 (95% confidence interval: 1.0 to 2.9]). The traditional pattern was not associated with allostatic load, whereas a significant association between the sweets pattern and allostatic load disappeared after restricting analysis to participants without diabetes. CONCLUSIONS A meat, processed meat, and french fries dietary pattern may contribute to the deregulation of multiple physiologic parameters in Puerto Rican adults. Efforts to limit consumption of this pattern may help prevent further cumulative physiological dysregulation in this high risk population.
Collapse
Affiliation(s)
- Josiemer Mattei
- Bouvé College of Health Sciences, Northeastern University, 360 Huntington Avenue, 316 Robinson, Boston, MA, 02115, Tel: 617-373-4273, Fax: 617-373-2968
| | - Sabina E. Noel
- Department of Pediatrics, Boston University School of Medicine, Boston Medical Center, 88 East Newton St. Vose Hall 301A Boston, MA 02118, Tel: Phone: 617-414-3643, Fax:
| | - Katherine L. Tucker
- Department of Health Sciences, Bouvé College of Health Sciences, Northeastern University, 360 Huntington Avenue, 316 Robinson, Boston, MA, 02115, Tel: 617-373-7952, Fax: 617-373-2968
| |
Collapse
|
37
|
Clock genes and sleep. Pflugers Arch 2011; 463:3-14. [DOI: 10.1007/s00424-011-1003-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 07/08/2011] [Accepted: 07/22/2011] [Indexed: 11/26/2022]
|
38
|
Dowd JB, Ranjit N, Do DP, Young EA, House JS, Kaplan GA. Education and levels of salivary cortisol over the day in US adults. Ann Behav Med 2011; 41:13-20. [PMID: 20812036 DOI: 10.1007/s12160-010-9224-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Dysregulation of the hypothalamic-pituitary-adrenal axis is hypothesized to be an important pathway linking socioeconomic position and chronic disease. PURPOSE This paper tests the association between education and the diurnal rhythm of salivary cortisol. METHODS Up to eight measures of cortisol (mean of 5.38 per respondent) over 2 days were obtained from 311 respondents, aged 18-70, drawn from the 2001-2002 Chicago Community Adult Health Study. Multi-level models with linear splines were used to estimate waking level, rates of cortisol decline, and area-under-the-curve over the day, by categories of education. RESULTS Lower education (0-11 years) was associated with lower waking levels of cortisol, but not the rate of decline of cortisol, resulting in a higher area-under-the-curve for more educated respondents throughout the day. CONCLUSIONS This study found evidence of lower cortisol exposure among individuals with less education and thus does not support the hypothesis that less education is associated with chronic over-exposure to cortisol.
Collapse
Affiliation(s)
- Jennifer B Dowd
- School of Public Health, Hunter College, City University of New York (CUNY), 425 E. 25th St. Box 926, New York, NY, 10010, USA,
| | | | | | | | | | | |
Collapse
|
39
|
Hannemann A, Meisinger C, Bidlingmaier M, Döring A, Thorand B, Heier M, Belcredi P, Ladwig KH, Wallaschofski H, Friedrich N, Schipf S, Lüdemann J, Rettig R, Peters J, Völzke H, Seissler J, Beuschlein F, Nauck M, Reincke M. Association of plasma aldosterone with the metabolic syndrome in two German populations. Eur J Endocrinol 2011; 164:751-8. [PMID: 21357289 DOI: 10.1530/eje-10-1074] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVE The aim of this study was to analyze the potential association of the plasma aldosterone concentration (PAC) with the metabolic syndrome (MetS) and its components in two German population-based studies. METHODS We selected 2830 and 2901 participants (31-80 years) from the follow-ups of the Study of Health in Pomerania (SHIP)-1 and the Cooperative Health Research in the Region of Augsburg (KORA) F4 respectively. MetS was defined as the presence of at least three out of the following five criteria: waist circumference ≥94 cm (men (m)) and ≥80 cm (women (w)); high-density lipoprotein (HDL) cholesterol <1.0 mmol/l (m) and <1.3 mmol/l (w); blood pressure ≥130/85 mmHg or antihypertensive treatment; non-fasting glucose (SHIP-1) ≥8 mmol/l, fasting glucose (KORA F4) ≥5.55 mmol/l or antidiabetic treatment; non-fasting triglycerides (SHIP-1) ≥2.3 mmol/l, fasting triglycerides (KORA F4) ≥1.7 mmol/l, or lipid-lowering treatment. We calculated logistic regression models by comparing the highest study- and sex-specific PAC quintiles versus all lower quintiles. RESULTS MetS was common with 48.1% (m) and 34.8% (w) in SHIP-1 and 42.7% (m) and 27.5% (w) in KORA F4. Our logistic regression models revealed associations of PAC with MetS, elevated triglycerides, and decreased HDL cholesterol in SHIP-1 and KORA F4. CONCLUSIONS Our findings add to the increasing evidence supporting a relation between aldosterone and MetS and suggest that aldosterone may be involved in the pathophysiology of MetS and lipid metabolism disorders.
Collapse
Affiliation(s)
- Anke Hannemann
- Institute of Clinical Chemistry and Laboratory Medicine, Ernst-Moritz-Arndt-University Greifswald, Ferdinand-Sauerbruch-Strasse, Greifswald, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Auci DL, Ahlem CN, Kennedy MR, Page TM, Reading CL, Frincke JM. A potential role for 5-androstene-3β,7β,17β-triol in obesity and metabolic syndrome. Obesity (Silver Spring) 2011; 19:806-11. [PMID: 20847733 DOI: 10.1038/oby.2010.204] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Metabolic syndrome is marked by perturbed glucocorticoid (GC) signaling, systemic inflammation, and altered immune status. Dehydroepiandrosterone (DHEA), a major circulating adrenal steroid and dietary supplement, demonstrates antiobesity, anti-inflammatory, GC-opposing and immune-modulating activity when administered to rodents. However, plasma DHEA levels failed to correlate with metabolic syndrome and oral replacement therapy provided only mild benefits to patients. Androstene-3β,7β,17β-triol (β-AET) an anti-inflammatory metabolite of DHEA, also exhibits GC-opposing and immune-modulating activity when administered to rodents. We hypothesized a role for β-AET in obesity. We now report that plasma levels of β-AET positively correlate with BMI in healthy men and women. Together with previous studies, the observations reported here may suggest a compensatory role for β-AET in preventing the development of metabolic syndrome. The β-AET structural core may provide the basis for novel pharmaceuticals to treat this disease.
Collapse
|
41
|
Kagerer SM, Eichholz C, Jöhren O. Orexins/hypocretins increase the promoter activity of selective steroidogenic enzymes. Peptides 2011; 32:839-43. [PMID: 21256172 DOI: 10.1016/j.peptides.2011.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 01/05/2011] [Accepted: 01/05/2011] [Indexed: 10/18/2022]
Abstract
Orexins (hypocretins) regulate multiple physiological functions, including central regulation of energy homeostasis and sleep-wake behavior but also peripheral hormonal actions. Recent data suggest specific effects of orexins at adrenal glands. To further assess the mechanism by which orexins regulate steroidogenesis we analyzed the effect of orexin A and B on the transcriptional activity of the luciferase reporter gene driven by the human steroid 21-hydroxylase (CYP21), 3β-hydroxysteroid dehydrogenase (HSD3B2), 11β-hydroxylase (CYP11B1), and aldosterone synthase (CYP11B2) gene promoter regions. After transient transfection of the reporter gene constructs into human NCI H295R cells, treatment with orexin A and B for 6 and 12h increased the promoter activity of the CYP11B2, HSD3B2 and, to a lesser extend, CYP21 genes. The activity of the CYP11B1 was increased by both orexins after 3h of treatment. Compared to the effects of forskolin or angiotensin II, however, the effect of orexins on the transcriptional activity of the steroidogenic enzyme genes was moderate. Our results suggest that orexins increase the expression of steroidogenic enzymes at the transcriptional level and that orexins play a role in the long term regulation of adrenal steroid production.
Collapse
Affiliation(s)
- Sonja M Kagerer
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany
| | | | | |
Collapse
|
42
|
Abstract
Systematic screening studies performed mainly in patients with diabetes mellitus have revealed an unexpectedly high prevalence of occult Cushing syndrome. Such studies may provide a rationale for systematically screening obese patients with type 2 diabetes mellitus. However, a screening strategy is only justified if it is supported by enough evidence of its efficacy and if the benefits will outweigh drawbacks. To date, the natural history of occult Cushing syndrome and its possible effect on long-term morbidity are unknown. The clinical spectrum of occult Cushing syndrome and its relatively low prevalence may potentially negatively affect the performance of endocrine tests used to diagnose overt Cushing syndrome and generate false positives. Whether the cure of occult Cushing syndrome favorably influences clinical outcomes and is more beneficial than treatment of diabetes mellitus and cardiovascular risk factors with currently available pharmacological tools remains to be demonstrated. Last, the acceptability of a screening program by professionals and the health-care system in terms of workload and costs is highly questionable. Thus, an assessment of the indications for and against screening for occult Cushing syndrome on the basis of currently available data suggests that, to date, the cons surpass the pros.
Collapse
Affiliation(s)
- Antoine Tabarin
- Department of Endocrinology, Université Bordeaux 2, Centre Hospitalier Universitaire de Bordeaux, France.
| | | |
Collapse
|
43
|
Barone MTU, Menna-Barreto L. Diabetes and sleep: a complex cause-and-effect relationship. Diabetes Res Clin Pract 2011; 91:129-37. [PMID: 20810183 DOI: 10.1016/j.diabres.2010.07.011] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 07/26/2010] [Accepted: 07/29/2010] [Indexed: 12/22/2022]
Abstract
Strong associations of diabetes with sleep impairment have been frequently reported. In the present review, we discuss current evidence and hypotheses for how type 1 and type 2 diabetes mellitus are associated with sleep impairment. This association may be described as a vicious circle, where sleep disorders favor the development of type 2 diabetes or exacerbate the metabolic control of both types of diabetes, whereas diabetes itself, especially when associated with poor metabolic control, is often followed by sleep disorders. In this review, novel findings concerning the neuro-endocrine-metabolic mediation of the mentioned circle are highlighted. Understanding how this association occurs, the impact of sleep impairment on diabetes, and the impact of diabetes on the development or exacerbation of sleep disorders should lead to potential new therapeutic strategies for treating both conditions.
Collapse
Affiliation(s)
- Mark T U Barone
- Instituto de Ciências Biomédicas, Universidade de São Paulo, Brazil.
| | | |
Collapse
|
44
|
Costa MJ, So AYL, Kaasik K, Krueger KC, Pillsbury ML, Fu YH, Ptacek LJ, Yamamoto KR, Feldman BJ. Circadian rhythm gene period 3 is an inhibitor of the adipocyte cell fate. J Biol Chem 2011; 286:9063-70. [PMID: 21228270 DOI: 10.1074/jbc.m110.164558] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glucocorticoids rapidly and robustly induce cell fate decisions in various multipotent cells, although the precise mechanisms of these important cellular events are not understood. Here we showed that glucocorticoids repressed Per3 expression and that this repression was critical for advancing mesenchymal stem cells to the adipocyte fate. Exogenous expression of Per3 inhibited adipogenesis, whereas knocking out Per3 enhanced that fate. Moreover, we found that PER3 formed a complex with PPARγ and inhibited PPARγ-mediated transcriptional activation via Pparγ response elements. Consistent with these findings, Per3 knock-out mice displayed alterations in body composition, with both increased adipose and decreased muscle tissue compared with wild-type mice. Our findings identify Per3 as potent mediator of cell fate that functions by altering the transcriptional activity of PPARγ.
Collapse
Affiliation(s)
- Maria Jose Costa
- Department of Pediatrics, University, Stanford, California 94305, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Ahlem CN, Page TM, Auci DL, Kennedy MR, Mangano K, Nicoletti F, Ge Y, Huang Y, White SK, Villegas S, Conrad D, Wang A, Reading CL, Frincke JM. Novel components of the human metabolome: the identification, characterization and anti-inflammatory activity of two 5-androstene tetrols. Steroids 2011; 76:145-55. [PMID: 20974164 DOI: 10.1016/j.steroids.2010.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 10/13/2010] [Accepted: 10/13/2010] [Indexed: 01/24/2023]
Abstract
Two natural 5-androstene steroid tetrols, androst-5-ene-3β,7β,16α,17β-tetrol (HE3177) and androst-5-ene-3α,7β,16α,17β-tetrol (HE3413), were discovered in human plasma and urine. These compounds had significant aqueous solubility, did not bind or transactivate steroid-binding nuclear hormone receptors, and were not immunosuppressive in murine mixed-lymphocyte studies. Both compounds appear to be metabolic end products, as they were resistant to primary and secondary metabolism. Both were orally bioavailable, and were very well tolerated in a two-week dose-intensive toxicity study in mice. Anti-inflammatory properties were found with exogenous administration of these compounds in rodent disease models of multiple sclerosis, lung injury, chronic prostatitis, and colitis.
Collapse
Affiliation(s)
- Clarence N Ahlem
- Harbor Biosciences, Inc., 9171 Towne Centre Drive, Suite 180, San Diego, CA 92122, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Shi L, Remer T, Buyken AE, Hartmann MF, Hoffmann P, Wudy SA. Prepubertal urinary estrogen excretion and its relationship with pubertal timing. Am J Physiol Endocrinol Metab 2010; 299:E990-7. [PMID: 20858752 DOI: 10.1152/ajpendo.00374.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Whether prepubertal estrogen production impacts on the timing of puberty is not clear. We aimed to investigate prepubertal 24-h estrogen excretion levels and their association with early and late pubertal markers. Daily urinary excretion rates of estrogens of 132 healthy children, who provided 24-h urine samples 1 and 2 yr before the start of the pubertal growth spurt [age at takeoff (ATO)], were quantified by stable isotope dilution/GC-MS. E-sum3 (estrone + estradiol + estriol) was used as a marker for potentially bioactive estrogen metabolites and E-sum5 (E-sum3 + 16-epiestriol + 16-ketoestradiol) for total estrogen production. Pubertal outcomes were ATO, age at peak height velocity (APHV), duration of pubertal growth acceleration (APHV-ATO), age at Tanner stage 2 for pubic hair (PH2), genital (G2, boys) and breast (B2, girls) development, and age at menarche. Prepubertal urinary estrogen excretions (E-sum3 and E-sum5) were not associated with ATO, APHV, and age at PH2 but with duration of pubertal growth acceleration (P < 0.01) in both sexes. Girls with higher E-sum3 reached B2 0.9 yr (P = 0.04) and menarche 0.3 yr earlier (P = 0.04) than girls with lower E-sum3. E-sum3 was not associated with age at G2 in boys (P = 0.6). For most pubertal variables, the associations with E-sum3 were stronger than with E-sum5. In conclusion, prepubertal estrogens may not be critical for the onset of the pubertal growth spurt but are correlated with its duration in both boys and girls. Prepubertal estrogen levels may already predict the timing of girls' menstruation and breast development but do not appear to affect sexual maturation in boys.
Collapse
Affiliation(s)
- Lijie Shi
- Research Institute of Child Nutrition, Dept. of Nutrition and Health, Heinstueck 11, 44225, Dortmund, Germany.
| | | | | | | | | | | |
Collapse
|
47
|
Chen MJ, Chen CD, Yang JH, Chen CL, Ho HN, Yang WS, Yang YS. High serum dehydroepiandrosterone sulfate is associated with phenotypic acne and a reduced risk of abdominal obesity in women with polycystic ovary syndrome. Hum Reprod 2010; 26:227-34. [PMID: 21088016 DOI: 10.1093/humrep/deq308] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Women with polycystic ovary syndrome (PCOS) are known to have high prevalence of acne and elevated androgen levels. The current study aims to determine if dehydroepiandrosterone sulfate (DHEAS) level is associated with the presence of acne and reduced risk of abdominal obesity in women with PCOS, after considering the concurrent high testosterone level and insulin resistance (IR). METHODS Three hundred and eighteen untreated consecutive Taiwanese women with PCOS were enrolled. Phenotypic hyperandrogenism was recorded, and BMI, waist circumference, waist-to-hip ratio, lipid profiles, fasting glucose and insulin levels and hormone profiles were measured. RESULTS Women with acne were younger, had higher serum DHEAS levels (6.01 ± 3.45 versus 4.87 ± 2.49 μmol/l, P = 0.002) and a lower BMI (P = 0.0006), but comparable serum testosterone levels, in comparison with women without acne. The aggravating effect of elevated DHEAS on the risk of acne (odds ratio = 2.15, 95% confidence interval: 1.25-3.68, P = 0.005 for DHEAS cut-off of 6.68 μmol/l) still exited after adjustment for age and BMI. The DHEAS level was positively correlated with the testosterone level, but inversely related to waist circumference, waist-to-hip ratio, BMI, IR index, low-density lipoprotein-cholesterol and triglycerides. Women with PCOS in the highest quartile of DHEAS had the lowest risk of abdominal obesity after adjustment for age, IR, dyslipidemia, testosterone and estradiol levels. CONCLUSIONS Our results demonstrated the high serum DHEAS in women with PCOS was associated with the presence of acne and a significantly reduced risk of abdominal obesity, independent of serum testosterone concentration and IR.
Collapse
Affiliation(s)
- Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Classically, sleep has been considered to serve an essential restorative function for the brain. However, there are an increasing number of studies linking decreased sleep quantity and/or quality in humans to an increased obesity and diabetes risk. Reductions in sleep quantity or quality lead to an increase in hunger and appetite, which chronically could predispose an individual to obesity. Carefully controlled studies have shown that two nights of insufficient sleep is causally linked to a decrease in disposition index, the most commonly used predictor of an individual's diabetes risk, and impairments in glucose tolerance and insulin sensitivity. Thus, sleep appears to play a critical role in modulating energy metabolism in peripheral tissues. Here we will discuss recent work implicating adipose tissue as a potential direct target of disruption of sleep quality, and explore the potential mechanistic links between sleep, adipose tissue and the global control of energy metabolism in humans.
Collapse
Affiliation(s)
- Josiane Broussard
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, 1333 San Pablo Street, MMR 626, Los Angeles, CA 90089, Tel: 323-442-1919; Fax: 323-442-1918
| | - Matthew J. Brady
- Department of Medicine, Committee on Molecular Metabolism and Nutrition, University of Chicago, 5841 S. Maryland Ave, MC1027, Chicago, IL 60637, Tel: 773-702-2346; Fax: 773-834-0486
| |
Collapse
|
49
|
Low stress reactivity and neuroendocrine factors in the BTBR T+tf/J mouse model of autism. Neuroscience 2010; 171:1197-208. [PMID: 20888890 DOI: 10.1016/j.neuroscience.2010.09.059] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Revised: 09/02/2010] [Accepted: 09/28/2010] [Indexed: 12/31/2022]
Abstract
Autism is a neurodevelopmental disorder characterized by abnormal reciprocal social interactions, communication deficits, and repetitive behaviors with restricted interests. BTBR T+tf/J (BTBR) is an inbred mouse strain that displays robust behavioral phenotypes with analogies to all three of the diagnostic symptoms of autism, including low social interactions, reduced vocalizations in social settings, and high levels of repetitive self-grooming. Autism-relevant phenotypes in BTBR offer translational tools to discover neurochemical mechanisms underlying unusual mouse behaviors relevant to symptoms of autism. Because repetitive self-grooming in mice may be a displacement behavior elevated by stressors, we investigated neuroendocrine markers of stress and behavioral reactivity to stressors in BTBR mice, as compared to C57BL/6J (B6), a standard inbred strain with high sociability. Radioimmunoassays replicated previous findings that circulating corticosterone is higher in BTBR than in B6. Higher basal glucocorticoid receptor mRNA and higher oxytocin peptide levels were detected in the brains of BTBR as compared to B6. No significant differences were detected in corticotrophin releasing factor (CRF) peptide or CRF mRNA. In response to behavioral stressors, BTBR and B6 were generally similar on behavioral tasks including stress-induced hyperthermia, elevated plus-maze, light ↔ dark exploration, tail flick, acoustic startle and prepulse inhibition. BTBR displayed less reactivity than B6 to a noxious thermal stimulus in the hot plate, and less immobility than B6 in both the forced swim and tail suspension depression-related tasks. BTBR, therefore, exhibited lower depression-like scores than B6 on two standard tests sensitive to antidepressants, did not differ from B6 on two well-validated anxiety-like behaviors, and did not exhibit unusual stress reactivity to sensory stimuli. Our findings support the interpretation that autism-relevant social deficits, vocalizations, and repetitive behaviors are not the result of abnormal stress reactivity in the BTBR mouse model of autism.
Collapse
|
50
|
Chronic inflammation in obesity and the metabolic syndrome. Mediators Inflamm 2010; 2010. [PMID: 20706689 PMCID: PMC2913796 DOI: 10.1155/2010/289645] [Citation(s) in RCA: 725] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Accepted: 06/17/2010] [Indexed: 12/14/2022] Open
Abstract
The increasing incidence of obesity and the metabolic syndrome is disturbing. The activation of inflammatory pathways, used normally as host defence, reminds the seriousness of this condition. There is probably more than one cause for activation of inflammation. Apparently, metabolic overload evokes stress reactions, such as oxidative, inflammatory, organelle and cell hypertrophy, generating vicious cycles. Adipocyte hypertrophy, through physical reasons, facilitates cell rupture, what will evoke an inflammatory reaction. Inability of adipose tissue development to engulf incoming fat leads to deposition in other organs, mainly in the liver, with consequences on insulin resistance. The oxidative stress which accompanies feeding, particularly when there is excessive ingestion of fat and/or other macronutrients without concomitant ingestion of antioxidant-rich foods/beverages, may contribute to inflammation attributed to obesity. Moreover, data on the interaction of microbiota with food and obesity brought new hypothesis for the obesity/fat diet relationship with inflammation. Beyond these, other phenomena, for instance psychological and/or circadian rhythm disturbances, may likewise contribute to oxidative/inflammatory status. The difficulty in the management of obesity/metabolic syndrome is linked to their multifactorial nature where environmental, genetic and psychosocial factors interact through complex networks.
Collapse
|