51
|
Lin JZ, Chen CT, Ma JD, Mo YQ, Li QH, Chen LF, Yang ZH, Cheng WM, He XL, Zheng DH, Dai L. Neglected extra-articular manifestations in rheumatoid arthritis patients with normal body mass index: reduced skeletal muscle overlapping overfat. Ther Adv Chronic Dis 2020; 11:2040622320975241. [PMID: 33294150 PMCID: PMC7705189 DOI: 10.1177/2040622320975241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Chronic inflammation in rheumatoid arthritis (RA) can induce reduced muscle mass (myopenia) and ectopic fat deposition probably showing normal body mass index (BMI). We aimed to investigate their body composition (BC) characteristics and clinical significance. Methods: BMI and BC were collected in consecutive RA patients and control subjects. Myopenia was defined by appendicular skeletal muscle mass index (ASMI) ⩽7.0 kg/m2 in men and ⩽5.7 kg/m2 in women. Overfat was defined by body fat percentage (BF%) as ⩾25% for men and ⩾35% for women. Results: There were 620 RA patients (57.6% with normal BMI) and 2537 control subjects (62.5% with normal BMI) recruited. After 1:1 age and sex matching with control subjects, RA patients with normal BMI (n = 240) showed significantly higher prevalence of myopenia (43.3% versus 22.1%) and overfat (19.2% versus 7.1%) as well as myopenia overlapping overfat (17.1% versus 3.3%). In all RA patients with normal BMI (n = 357), there were 18.2% patients with myopenia overlapping overfat who had the worst radiographic scores and highest rates of previous glucocorticoid treatment and hypertension. Compared with those without, normal BMI RA patients with previous glucocorticoid treatment (24.4% versus 10.3%) or hypertension (27.8% versus 13.6%) had a higher rate of myopenia overlapping overfat. Previous glucocorticoid treatment [odds ratio (OR) = 2.844, 95% confidence interval (CI) 1.441–5.614] and hypertension (OR = 2.452, 95% CI 1.283–4.685) were potential associated factors of myopenia overlapping overfat in RA patients with normal BMI. Conclusion: Myopenia overlapping overfat is an important extra-articular manifestation which should not be ignored in RA patients with normal BMI, especially with glucocorticoid treatment and hypertension.
Collapse
Affiliation(s)
- Jian-Zi Lin
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou, Guangdong, PR China
| | - Chu-Tao Chen
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou, Guangdong, PR China
| | - Jian-Da Ma
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou, Guangdong, PR China
| | - Ying-Qian Mo
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou, Guangdong, PR China
| | - Qian-Hua Li
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou, Guangdong, PR China
| | - Le-Feng Chen
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou, Guangdong, PR China
| | - Ze-Hong Yang
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou, Guangdong, PR China
| | - Wan-Mei Cheng
- Shanghai Healthare Co. Ltd, Zhangjiang Innopark, Shanghai, PR China
| | - Xiao-Ling He
- Shanghai Healthare Co. Ltd, Zhangjiang Innopark, Shanghai, PR China
| | - Dong-Hui Zheng
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou, Guangdong 510120, PR China
| | - Lie Dai
- Department of Rheumatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 Yan Jiang West Road, Guangzhou, Guangdong 510120, PR China
| |
Collapse
|
52
|
Leachman JR, Rea MD, Cohn DM, Xu X, Fondufe-Mittendorf YN, Loria AS. Exacerbated obesogenic response in female mice exposed to early life stress is linked to fat depot-specific upregulation of leptin protein expression. Am J Physiol Endocrinol Metab 2020; 319:E852-E862. [PMID: 32830551 PMCID: PMC7790118 DOI: 10.1152/ajpendo.00243.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Early life stress (ELS) is an independent risk factor for increased BMI and cardiometabolic disease risk later in life. We have previously shown that a mouse model of ELS, maternal separation and early weaning (MSEW), exacerbates high-fat diet (HF)-induced obesity only in adult female mice. Therefore, the aim of this study was to investigate 1) whether the short- and long-term effects of HF on leptin expression are influenced by MSEW in a sex-specific manner and 2) the potential epigenetic mechanisms underlying the MSEW-induced changes in leptin expression. After 1 wk of HF, both MSEW male and female mice displayed increased fat mass compared with controls (P < 0.05). However, only MSEW female mice showed elevated leptin mRNA expression in gonadal white adipose tissue (gWAT; P < 0.05). After 12 wk of HF, fat mass remained increased only in female mice (P < 0.05). Moreover, plasma leptin and both leptin mRNA and protein expression in gWAT were augmented in MSEW female mice compered to controls (P < 0.05), but not in MSEW male mice. This association was not present in subcutaneous WAT. Furthermore, among 16 CpG sites in the leptin promoter, we identified three hypomethylated sites in tissue from HF-fed MSEW female mice compared with controls (3, 15, and 16, P < 0.05). These hypomethylated sites showed greater binding of key adipogenic factors such as PPARγ (P < 0.05). Taken together, our study reveals that MSEW superimposed to HF increases leptin protein expression in a sex- and fat depot-specific fashion. Our data suggest that the mechanism by which MSEW increases leptin expression could be epigenetic.
Collapse
Affiliation(s)
- Jacqueline R Leachman
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Mathew D Rea
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky
| | - Dianne M Cohn
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Xiu Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | | | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
53
|
Yip HYK, Chee A, Ang CS, Shin SY, Ooms LM, Mohammadi Z, Phillips WA, Daly RJ, Cole TJ, Bronson RT, Nguyen LK, Tiganis T, Hobbs RM, McLean CA, Mitchell CA, Papa A. Control of Glucocorticoid Receptor Levels by PTEN Establishes a Failsafe Mechanism for Tumor Suppression. Mol Cell 2020; 80:279-295.e8. [PMID: 33065020 DOI: 10.1016/j.molcel.2020.09.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 08/03/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022]
Abstract
The PTEN tumor suppressor controls cell death and survival by regulating functions of various molecular targets. While the role of PTEN lipid-phosphatase activity on PtdIns(3,4,5)P3 and inhibition of PI3K pathway is well characterized, the biological relevance of PTEN protein-phosphatase activity remains undefined. Here, using knockin (KI) mice harboring cancer-associated and functionally relevant missense mutations, we show that although loss of PTEN lipid-phosphatase function cooperates with oncogenic PI3K to promote rapid mammary tumorigenesis, the additional loss of PTEN protein-phosphatase activity triggered an extensive cell death response evident in early and advanced mammary tumors. Omics and drug-targeting studies revealed that PI3Ks act to reduce glucocorticoid receptor (GR) levels, which are rescued by loss of PTEN protein-phosphatase activity to restrain cell survival. Thus, we find that the dual regulation of GR by PI3K and PTEN functions as a rheostat that can be exploited for the treatment of PTEN loss-driven cancers.
Collapse
Affiliation(s)
- Hon Yan K Yip
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Annabel Chee
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Ching-Seng Ang
- Bio21 Mass Spectrometry and Proteomics Facility, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Sung-Young Shin
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Lisa M Ooms
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Zainab Mohammadi
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Wayne A Phillips
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia; Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Roger J Daly
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Timothy J Cole
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Roderick T Bronson
- Department of Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Lan K Nguyen
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Tony Tiganis
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia; Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Robin M Hobbs
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC 3800, Australia; Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| | - Catriona A McLean
- Department of Anatomical Pathology, Alfred Hospital, Prahran, VIC 3181, Australia
| | - Christina A Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | - Antonella Papa
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia.
| |
Collapse
|
54
|
Anderson WD, Soh JY, Innis SE, Dimanche A, Ma L, Langefeld CD, Comeau ME, Das SK, Schadt EE, Björkegren JLM, Civelek M. Sex differences in human adipose tissue gene expression and genetic regulation involve adipogenesis. Genome Res 2020; 30:1379-1392. [PMID: 32967914 PMCID: PMC7605264 DOI: 10.1101/gr.264614.120] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
Sex differences in adipose tissue distribution and function are associated with sex differences in cardiometabolic disease. While many studies have revealed sex differences in adipocyte cell signaling and physiology, there is a relative dearth of information regarding sex differences in transcript abundance and regulation. We investigated sex differences in subcutaneous adipose tissue transcriptional regulation using omic-scale data from ∼3000 geographically and ethnically diverse human samples. We identified 162 genes with robust sex differences in expression. Differentially expressed genes were implicated in oxidative phosphorylation and adipogenesis. We further determined that sex differences in gene expression levels could be related to sex differences in the genetics of gene expression regulation. Our analyses revealed sex-specific genetic associations, and this finding was replicated in a study of 98 inbred mouse strains. The genes under genetic regulation in human and mouse were enriched for oxidative phosphorylation and adipogenesis. Enrichment analysis showed that the associated genetic loci resided within binding motifs for adipogenic transcription factors (e.g., PPARG and EGR1). We demonstrated that sex differences in gene expression could be influenced by sex differences in genetic regulation for six genes (e.g., FADS1 and MAP1B). These genes exhibited dynamic expression patterns during adipogenesis and robust expression in mature human adipocytes. Our results support a role for adipogenesis-related genes in subcutaneous adipose tissue sex differences in the genetic and environmental regulation of gene expression.
Collapse
Affiliation(s)
- Warren D Anderson
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Joon Yuhl Soh
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Sarah E Innis
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Alexis Dimanche
- Physics Department, Southwestern University, Georgetown, Texas 78626, USA
| | - Lijiang Ma
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Carl D Langefeld
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, USA
| | - Mary E Comeau
- Department of Biostatistics and Data Science, Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, USA
| | - Swapan K Das
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, USA
| | - Eric E Schadt
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Johan L M Björkegren
- Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia 22908, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia 22904, USA
| |
Collapse
|
55
|
Willmer T, Goedecke JH, Dias S, Louw J, Pheiffer C. DNA methylation of FKBP5 in South African women: associations with obesity and insulin resistance. Clin Epigenetics 2020; 12:141. [PMID: 32958048 PMCID: PMC7507280 DOI: 10.1186/s13148-020-00932-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Disruption of the hypothalamic-pituitary-adrenal (HPA) axis, a neuroendocrine system associated with the stress response, has been hypothesized to contribute to obesity development. This may be mediated through epigenetic modulation of HPA axis-regulatory genes in response to metabolic stressors. The aim of this study was to investigate adipose tissue depot-specific DNA methylation differences in the glucocorticoid receptor (GR) and its co-chaperone, FK506-binding protein 51 kDa (FKBP5), both key modulators of the HPA axis. METHODS Abdominal subcutaneous adipose tissue (ASAT) and gluteal subcutaneous adipose tissue (GSAT) biopsies were obtained from a sample of 27 obese and 27 normal weight urban-dwelling South African women. DNA methylation and gene expression were measured by pyrosequencing and quantitative real-time PCR, respectively. Spearman's correlation coefficients, orthogonal partial least-squares discriminant analysis and multivariable linear regression were performed to evaluate the associations between DNA methylation, messenger RNA (mRNA) expression and key indices of obesity and metabolic dysfunction. RESULTS Two CpG dinucleotides within intron 7 of FKBP5 were hypermethylated in both ASAT and GSAT in obese compared to normal weight women, while no differences in GR methylation were observed. Higher percentage methylation of the two FKBP5 CpG sites correlated with adiposity (body mass index and waist circumference), insulin resistance (homeostasis model for insulin resistance, fasting insulin and plasma adipokines) and systemic inflammation (c-reactive protein) in both adipose depots. GR and FKBP5 mRNA levels were lower in GSAT, but not ASAT, of obese compared to normal weight women. Moreover, FKBP5 mRNA levels were inversely correlated with DNA methylation and positively associated with adiposity, metabolic and inflammatory parameters. CONCLUSIONS These findings associate dysregulated FKBP5 methylation and mRNA expression with obesity and insulin resistance in South African women. Additional studies are required to assess the longitudinal association of FKBP5 with obesity and associated co-morbidities in large population-based samples.
Collapse
Affiliation(s)
- Tarryn Willmer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, 7505, South Africa.
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, 7505, South Africa.
| | - Julia H Goedecke
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Tygerberg, 7505, South Africa
- Division of Exercise Science and Sports Medicine, Department of Human Biology, University of Cape Town, Boundary Road, Newlands, 7700, South Africa
| | - Stephanie Dias
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, 7505, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, 7505, South Africa
- Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa, 3886, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, 7505, South Africa
- Division of Medical Physiology, Faculty of Health Sciences, Stellenbosch University, Tygerberg, 7505, South Africa
| |
Collapse
|
56
|
Hinds TD, Creeden JF, Gordon DM, Spegele AC, Britton SL, Koch LG, Stec DE. Rats Genetically Selected for High Aerobic Exercise Capacity Have Elevated Plasma Bilirubin by Upregulation of Hepatic Biliverdin Reductase-A (BVRA) and Suppression of UGT1A1. Antioxidants (Basel) 2020; 9:antiox9090889. [PMID: 32961782 PMCID: PMC7554716 DOI: 10.3390/antiox9090889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/12/2020] [Accepted: 09/16/2020] [Indexed: 12/15/2022] Open
Abstract
Exercise in humans and animals increases plasma bilirubin levels, but the mechanism by which this occurs is unknown. In the present study, we utilized rats genetically selected for high capacity running (HCR) and low capacity running (LCR) to determine pathways in the liver that aerobic exercise modifies to control plasma bilirubin. The HCR rats, compared to the LCR, exhibited significantly higher levels of plasma bilirubin and the hepatic enzyme that produces it, biliverdin reductase-A (BVRA). The HCR also had reduced expression of the glucuronyl hepatic enzyme UGT1A1, which lowers plasma bilirubin. Recently, bilirubin has been shown to activate the peroxisome proliferator-activated receptor-α (PPARα), a ligand-induced transcription factor, and the higher bilirubin HCR rats had significantly increased PPARα-target genes Fgf21, Abcd3, and Gys2. These are known to promote liver function and glycogen storage, which we found by Periodic acid–Schiff (PAS) staining that hepatic glycogen content was higher in the HCR versus the LCR. Our results demonstrate that exercise stimulates pathways that raise plasma bilirubin through alterations in hepatic enzymes involved in bilirubin synthesis and metabolism, improving liver function, and glycogen content. These mechanisms may explain the beneficial effects of exercise on plasma bilirubin levels and health in humans.
Collapse
Affiliation(s)
- Terry D. Hinds
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40508, USA
- Correspondence: (T.D.H.J.); (D.E.S.)
| | - Justin F. Creeden
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH 43614, USA; (J.F.C.); (D.M.G.)
| | - Darren M. Gordon
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, OH 43614, USA; (J.F.C.); (D.M.G.)
| | - Adam C. Spegele
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA; (A.C.S.); (L.G.K.)
| | - Steven L. Britton
- Department of Anesthesiology, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Lauren G. Koch
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH 43614, USA; (A.C.S.); (L.G.K.)
| | - David E. Stec
- Center for Excellence in Cardiovascular-Renal Research, Department of Physiology & Biophysics, University of Mississippi Medical Center, 2500 North State St, Jackson, MS 392161, USA
- Correspondence: (T.D.H.J.); (D.E.S.)
| |
Collapse
|
57
|
Schneider MO, Hübner T, Pretscher J, Goecke TW, Schwitulla J, Häberle L, Kornhuber J, Ekici AB, Beckmann MW, Fasching PA, Schwenke E. Genetic variants in the glucocorticoid pathway genes and birth weight. Arch Gynecol Obstet 2020; 303:427-434. [PMID: 32886236 DOI: 10.1007/s00404-020-05761-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 08/24/2020] [Indexed: 12/01/2022]
Abstract
PURPOSE The aim of this study was to examine associations between single nucleotide polymorphisms (SNPs) that tag genetic variation in the glucocorticoid pathways (particularly in maternal genes FKBP5, NR3C1, and CRHR1) and birth weight. METHODS The Franconian Maternal Health Evaluation Study (FRAMES) recruited healthy pregnant women prospectively for the assessment of maternal and fetal health. Germline DNA was collected from 375 pregnant women. Nine SNPs in the above-mentioned genes were genotyped. After reconstruction of haplotypes for each gene, a linear regression model was applied to the data to describe the association between haplotypes and birth weight. RESULTS Female sex in the newborn (compared to male) was associated with lower birth weight, whereas a later week of gestation, higher body mass index pre-pregnancy, and higher parity were associated with higher birth weight. No association with birthweight was shown for the haplotypes of the selected SNPs. CONCLUSIONS In this cohort of healthy unselected pregnant women, the analyzed candidate haplotypes in FKBP5, NR3C1, and CRHR1 did not show any association with birth weight. This might be in line with several other studies that have found no influence of fetal polymorphisms in the glucocorticoid receptor gene or triggers of the maternal HPA axis such as stress and psychosocial problems on birth weight. However, the small sample size in this study and the lack of consideration of individual risk factors and levels of stress in this cohort needs to be taken into account when interpreting the results.
Collapse
Affiliation(s)
- Michael O Schneider
- Department of Gynecology and Obstetrics, Erlangen University Perinatal Center, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany.
| | - Theresa Hübner
- Department of Gynecology and Obstetrics, Erlangen University Perinatal Center, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Department of Gynecology and Obstetrics, Würzburg University Hospital, Würzburg, Germany
| | - Jutta Pretscher
- Department of Gynecology and Obstetrics, Erlangen University Perinatal Center, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
| | - Tamme W Goecke
- Department of Gynecology and Obstetrics, Erlangen University Perinatal Center, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
- Department of Obstetrics, RoMed Clinic Rosenheim, Rosenheim, Germany
| | - Judith Schwitulla
- Biostatistics Unit, Department of Gynecology and Obstetrics, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Lothar Häberle
- Biostatistics Unit, Department of Gynecology and Obstetrics, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Erlangen University Perinatal Center, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
| | - Peter A Fasching
- Department of Gynecology and Obstetrics, Erlangen University Perinatal Center, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
| | - Eva Schwenke
- Department of Gynecology and Obstetrics, Erlangen University Perinatal Center, Erlangen University Hospital, Friedrich Alexander University of Erlangen-Nuremberg, Universitätsstrasse 21-23, 91054, Erlangen, Germany
| |
Collapse
|
58
|
Linderborg KM, Kortesniemi M, Aatsinki AK, Karlsson L, Karlsson H, Yang B, Uusitupa HM. Interactions between cortisol and lipids in human milk. Int Breastfeed J 2020; 15:66. [PMID: 32690057 PMCID: PMC7370511 DOI: 10.1186/s13006-020-00307-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 07/09/2020] [Indexed: 12/05/2022] Open
Abstract
Background Human breast milk is one of the key early postnatal biological exposures for the developing child. It includes bioactive compounds, such as cortisol and fatty acids, which may be linked via the mother’s lipid metabolism. Methods This study investigated the associations between cortisol and lipids in human milk at the infant age of 2.5 months. Human milk cortisol concentrations were measured using luminescence immunoassay, and two groups of milks (n = 50 each) were formed based on either high (> 10 nmol/L) or low (< 3 nmol/L) cortisol levels. Lipids, as fatty acid content and composition of neutral (triacylglycerol-rich) and polar (phospholipid-rich) lipids, were measured with gas chromatography. The samples originated from the FinnBrain Birth Cohort Study. Results The percentage of phospholipid-rich lipids of total lipids was 33.08% ± 1.33%. In triacylglycerol-rich lipids, high cortisol level in milk was associated with higher lauric (12:0, mass % and mg/mL), myristic (14:0, mass % and mg/mL), eicosenoic (20:1n − 9, mass %), docosenoic (22:1n − 9, mass %, and mg/mL) acids, and to lower palmitic acid (16:0, mass %) compared with low cortisol levels in milk. In phospholipid-rich lipids, high cortisol level was associated with higher myristic (14:0, mass %) and docosenoic (22:1n − 9, mass %) acids. After adjusting for pre-pregnancy BMI and sampling time by linear regression, the milk cortisol remained a significant predictor for lauric and myristic acids in triacylglycerol-rich lipids, and myristic and docosenoic acid in phospholipid-rich lipids (β = 0.23 to 0.38 and p < 0.05 for each). Conclusions This study revealed certain significant associations between milk cortisol and the fatty acid composition of human milk, indicating that cortisol might be one of the factors affecting the origin of the lipids in human milk.
Collapse
Affiliation(s)
- Kaisa M Linderborg
- Department of Biochemistry, Food Chemistry and Food Development, University of Turku, Itäinen Pitkäkatu 4C, FI-20014, Turun yliopisto, Turku, Finland.
| | - Maaria Kortesniemi
- Department of Biochemistry, Food Chemistry and Food Development, University of Turku, Itäinen Pitkäkatu 4C, FI-20014, Turun yliopisto, Turku, Finland
| | - Anna-Katariina Aatsinki
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
| | - Linnea Karlsson
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland.,Department of Child Psychiatry, Turku University Hospital and University of Turku, Turku, Finland
| | - Hasse Karlsson
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland.,Department of Psychiatry, Turku University Hospital and University of Turku, Turku, Finland
| | - Baoru Yang
- Department of Biochemistry, Food Chemistry and Food Development, University of Turku, Itäinen Pitkäkatu 4C, FI-20014, Turun yliopisto, Turku, Finland
| | - Henna-Maria Uusitupa
- The FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine, University of Turku, Turku, Finland
| |
Collapse
|
59
|
Sex-dependent metabolic effects of pregestational exercise on prenatally stressed mice. J Dev Orig Health Dis 2020; 12:271-279. [PMID: 32406352 DOI: 10.1017/s2040174420000343] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Stressful events during the prenatal period have been related to hyperactive hypothalamic-pituitary-adrenal (HPA) axis responses as well as metabolic changes in adult life. Moreover, regular exercise may contribute to the improvement of the symptoms associated with stress and stress-related chronic diseases. Therefore, this study aims to investigate the effects of exercise, before the gestation period, on the metabolic changes induced by prenatal stress in adult mice. Female Balb/c mice were divided into three groups: control (CON), prenatal restraint stress (PNS) and exercise before the gestational period plus PNS (EX + PNS). When adults, the plasmatic biochemical analysis, oxidative stress, gene expression of metabolic-related receptors and sex differences were assessed in the offspring. Prenatal stress decreased neonatal and adult body weight when compared to the pregestational exercise group. Moreover, prenatal stress was associated with reduced body weight in adult males. PNS and EX + PNS females showed decreased hepatic catalase. Pregestational exercise prevented the stress-induced cholesterol increase in females but did not prevent the liver mRNA expression reduction on the peroxisome proliferator-activated receptors (PPARs) α and γ in PNS females. Conversely, PNS and EX + PNS males showed an increased PPARα mRNA expression. In conclusion, pregestational exercise prevented some effects of prenatal stress on metabolic markers in a sex-specific manner.
Collapse
|
60
|
Gordon DM, Neifer KL, Hamoud ARA, Hawk CF, Nestor-Kalinoski AL, Miruzzi SA, Morran MP, Adeosun SO, Sarver JG, Erhardt PW, McCullumsmith RE, Stec DE, Hinds TD. Bilirubin remodels murine white adipose tissue by reshaping mitochondrial activity and the coregulator profile of peroxisome proliferator-activated receptor α. J Biol Chem 2020; 295:9804-9822. [PMID: 32404366 DOI: 10.1074/jbc.ra120.013700] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/11/2020] [Indexed: 12/18/2022] Open
Abstract
Activation of lipid-burning pathways in the fat-storing white adipose tissue (WAT) is a promising strategy to improve metabolic health and reduce obesity, insulin resistance, and type II diabetes. For unknown reasons, bilirubin levels are negatively associated with obesity and diabetes. Here, using mice and an array of approaches, including MRI to assess body composition, biochemical assays to measure bilirubin and fatty acids, MitoTracker-based mitochondrial analysis, immunofluorescence, and high-throughput coregulator analysis, we show that bilirubin functions as a molecular switch for the nuclear receptor transcription factor peroxisome proliferator-activated receptor α (PPARα). Bilirubin exerted its effects by recruiting and dissociating specific coregulators in WAT, driving the expression of PPARα target genes such as uncoupling protein 1 (Ucp1) and adrenoreceptor β 3 (Adrb3). We also found that bilirubin is a selective ligand for PPARα and does not affect the activities of the related proteins PPARγ and PPARδ. We further found that diet-induced obese mice with mild hyperbilirubinemia have reduced WAT size and an increased number of mitochondria, associated with a restructuring of PPARα-binding coregulators. We conclude that bilirubin strongly affects organismal body weight by reshaping the PPARα coregulator profile, remodeling WAT to improve metabolic function, and reducing fat accumulation.
Collapse
Affiliation(s)
- Darren M Gordon
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA.,Center for Diabetes and Endocrine Research (CeDER), University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Kari L Neifer
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Abdul-Rizaq Ali Hamoud
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Charles F Hawk
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Andrea L Nestor-Kalinoski
- Advanced Microscopy and Imaging Center, Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Scott A Miruzzi
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Michael P Morran
- Advanced Microscopy and Imaging Center, Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Samuel O Adeosun
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jeffrey G Sarver
- Center for Drug Design and Development (CD3), Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, Ohio, USA
| | - Paul W Erhardt
- Center for Drug Design and Development (CD3), Department of Medicinal and Biological Chemistry, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, Ohio, USA
| | - Robert E McCullumsmith
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA.,ProMedica, Toledo, Ohio, USA
| | - David E Stec
- Department of Physiology and Biophysics, Cardiorenal and Metabolic Diseases Research Center, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Terry D Hinds
- Department of Neurosciences, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA .,Center for Diabetes and Endocrine Research (CeDER), University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| |
Collapse
|
61
|
Alhaddad H, Gordon DM, Bell RL, Jarvis EE, Kipp ZA, Hinds TD, Sari Y. Chronic Ethanol Consumption Alters Glucocorticoid Receptor Isoform Expression in Stress Neurocircuits and Mesocorticolimbic Brain Regions of Alcohol-Preferring Rats. Neuroscience 2020; 437:107-116. [PMID: 32353460 DOI: 10.1016/j.neuroscience.2020.04.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 01/17/2023]
Abstract
Evidence suggests the hypothalamic-pituitary-adrenal (HPA) axis is involved in Alcohol Use Disorders (AUDs), which might be mediated by an imbalance of glucocorticoid receptor (GR), GRα and GRβ, activity. GRβ antagonizes the GRα isoform to cause glucocorticoid (GC) resistance. In the present study, we aimed to investigate the effects of chronic continuous free-choice access to ethanol on GR isoform expression in subregions of the mesocorticolimbic reward circuit. Adult male alcohol-preferring (P) rats had concurrent access to 15% and 30% ethanol solutions, with ad lib access to lab chow and water, for six weeks. Quantitative Real-time PCR (RT-PCR) analysis showed that chronic ethanol consumption reduced GRα expression in the nucleus accumbens shell (NAcsh) and hippocampus, whereas ethanol drinking reduced GRβ in the nucleus accumbens core (NAcc), prefrontal cortex (PFC), and hippocampus. An inhibitor of GRα, microRNA-124-3p (miR124-3p) was significantly higher in the NAcsh, and GC-induced gene, GILZ, as a measure of GC-responsiveness, was significantly lower. These were not changed in the NAcc. Likewise, genes associated with HPA axis activity were not significantly changed by ethanol drinking [i.e., corticotrophin-releasing hormone (Crh), adrenocorticotrophic hormone (Acth), and proopiomelanocortin (Pomc)] in these brain regions. Serum corticosterone levels were not changed by ethanol drinking. These data indicate that the expression of GRα and GRβ isoforms are differentially affected by ethanol drinking despite HPA-associated peptides remaining unchanged, at least at the time of tissue harvesting. Moreover, the results suggest that GR changes may stem from ethanol-induced GC-resistance in the NAcsh. These findings confirm a role for stress in high ethanol drinking, with GRα and GRβ implicated as targets for the treatment of AUDs.
Collapse
Affiliation(s)
- Hasan Alhaddad
- Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA
| | - Darren M Gordon
- Department of Neurosciences, Center for Diabetes and Endocrine Research (CeDER), University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Richard L Bell
- Indiana University School of Medicine, Department of Psychiatry, Neurosciences Research Building, 320 West 15th Street, Indianapolis, IN 46202, USA
| | - Erin E Jarvis
- Indiana University School of Medicine, Department of Psychiatry, Neurosciences Research Building, 320 West 15th Street, Indianapolis, IN 46202, USA
| | - Zachary A Kipp
- Department of Neurosciences, Center for Diabetes and Endocrine Research (CeDER), University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Terry D Hinds
- Department of Neurosciences, Center for Diabetes and Endocrine Research (CeDER), University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA.
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA.
| |
Collapse
|
62
|
Nuclear Receptors as Regulators of Pituitary Corticotroph Pro-Opiomelanocortin Transcription. Cells 2020; 9:cells9040900. [PMID: 32272677 PMCID: PMC7226830 DOI: 10.3390/cells9040900] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 12/16/2022] Open
Abstract
The hypothalamic–pituitary–adrenal (HPA) axis plays a critical role in adaptive stress responses and maintaining organism homeostasis. The pituitary corticotroph is the central player in the HPA axis and is regulated by a plethora of hormonal and stress related factors that synergistically interact to activate and temper pro-opiomelanocortin (POMC) transcription, to either increase or decrease adrenocorticotropic hormone (ACTH) production and secretion as needed. Nuclear receptors are a family of highly conserved transcription factors that can also be induced by various physiologic signals, and they mediate their responses via multiple targets to regulate metabolism and homeostasis. In this review, we summarize the modulatory roles of nuclear receptors on pituitary corticotroph cell POMC transcription, describe the unique and complex role these factors play in hypothalamic–pituitary–adrenal axis (HPA) regulation and discuss potential therapeutic targets in disease states.
Collapse
|
63
|
Hill JL, Solomon MB, Nguyen ET, Caldwell JL, Wei Y, Foster MT. Glucocorticoids regulate adipose tissue protein concentration in a depot- and sex-specific manner. Stress 2020; 23:243-247. [PMID: 31441358 DOI: 10.1080/10253890.2019.1658736] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Preclinical and clinical findings indicate that glucocorticoids (GC) induce lipid accumulation in visceral depots, while inhibiting lipid stores from subcutaneous depots. Whereas some suggest that this is due to adipose depot specific concentration of glucocorticoid receptors (GR) or 11beta-hydroxysteroid dehydrogenase 1 (11β-HSD1), others demonstrate these events emerge from increases in interleukin-1 beta (IL-1β) from macrophages within distinct depots. Regardless of the mechanisms, most of these studies occur in males and thus lack evaluation of sex differences. Here, we examined the impact of 2-week corticosterone (CORT) (3 mg/kg/day) or saline treatment on GR, 11β-HSD1 and IL-1β protein concentration in intra-abdominal (epididymal/parametrial, and visceral) and subcutaneous (inguinal) depots in male and female Sprague Dawley rats. The objective was to examine if factors that regulate GC-induced adipose depot metabolism and distribution, differ between males and females. CORT inhibited, but did not decrease, body weight gain in both sexes. 11β-HSD1 was similar between the sexes in all adipose depots. CORT increased IL-1β in both sexes only in gonadal adipose tissue. Overall, males had greater GR protein concentration in all adipose depots, whereas females had more IL-1β in intra-abdominal adipose depots. Given the male-biased increase in intra-abdominal GR protein concentration, the data suggest that males may be more prone to CORT-induced increases in visceral obesity, which may have implications for increased risk for metabolic diseases. Overall, the data suggest that the effects of GC signaling in adipose tissue are multifaceted, dependent on sex, and the inherent adipocyte characteristics.Lay summaryResearch supports that glucocorticoids (GC) induce visceral adipose tissue accumulation, however few studies have examined if these GC-mediated outcomes are similar between males and females. This study investigates if female rats differentially respond to corticosterone treatment. Results indicate that male rats may have an increased susceptibility to CORT-induced accumulation of visceral adipose tissue compared with females, which may have implication for sex-specific risk for metabolic diseases.
Collapse
Affiliation(s)
- Jessica L Hill
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Matia B Solomon
- Department of Psychology Experimental Psychology Program, Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Elizabeth T Nguyen
- Department of Psychology Experimental Psychology Program, Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Jody L Caldwell
- Department of Psychology Experimental Psychology Program, Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - Yuren Wei
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA
| | - Michelle T Foster
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
64
|
Akalestou E, Genser L, Rutter GA. Glucocorticoid Metabolism in Obesity and Following Weight Loss. Front Endocrinol (Lausanne) 2020; 11:59. [PMID: 32153504 PMCID: PMC7045057 DOI: 10.3389/fendo.2020.00059] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/30/2020] [Indexed: 01/08/2023] Open
Abstract
Glucocorticoids are steroid hormones produced by the adrenal cortex and are essential for the maintenance of various metabolic and homeostatic functions. Their function is regulated at the tissue level by 11β-hydroxysteroid dehydrogenases and they signal through the glucocorticoid receptor, a ligand-dependent transcription factor. Clinical observations have linked excess glucocorticoid levels with profound metabolic disturbances of intermediate metabolism resulting in abdominal obesity, insulin resistance and dyslipidaemia. In this review, we discuss the physiological mechanisms of glucocorticoid secretion, regulation and function, and survey the metabolic consequences of excess glucocorticoid action resulting from elevated release and activation or up-regulated signaling. Finally, we summarize the reported impact of weight loss by diet, exercise, or bariatric surgery on circulating and tissue-specific glucocorticoid levels and examine the therapeutic possibility of reversing glucocorticoid-associated metabolic disorders.
Collapse
Affiliation(s)
- Elina Akalestou
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
| | - Laurent Genser
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- Department of Digestive and Hepato-Pancreato-Biliary Surgery, Liver Transplantation, Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière University Hospital, Institut Hospitalo-Universitaire ICAN, Sorbonne Université, Paris, France
| | - Guy A. Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial Centre for Translational and Experimental Medicine, Imperial College London, London, United Kingdom
- *Correspondence: Guy A. Rutter
| |
Collapse
|
65
|
Miranda RA, Pietrobon CB, Bertasso IM, Rodrigues VST, Lopes BP, Calvino C, de Oliveira E, de Moura EG, Lisboa PC. Early weaning leads to specific glucocorticoid signalling in fat depots of adult rats. Endocrine 2020; 67:180-189. [PMID: 31494802 DOI: 10.1007/s12020-019-02080-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/30/2019] [Indexed: 12/25/2022]
Abstract
PURPOSE Early weaning (EW) is a stressful condition that programmes a child to be overweight in adult life. Fat mass depends on glucocorticoids (GC) to regulate adipogenesis and lipogenesis. We hypothesised that the increased adiposity in models of EW was due to a disturbed HPA axis and/or disrupted GC function. METHODS We used two experimental models, pharmacological early weaning (PEW, dams were bromocriptine-treated) and non-pharmacological early weaning (NPEW, dams' teats were wrapped with a bandage), which were initiated during the last 3 days of lactation. Offspring from both genders was analysed on postnatal day 180. RESULTS Offspring in both models were overweight with increased visceral fat mass, but plasma corticosterone was increased in both genders in the PEW group but not the NPEW group. NPEW males had increased GRα expression in visceral adipose tissue (VAT), and GRα expression decreased in PEW males in subcutaneous adipose tissue (SAT). Females in both EW groups had increased 11βHSD1 expression in SAT. PEW males had increased C/EBPβ expression in SAT. PEW females had lower PPARy and FAS expression in VAT than the NPEW females. We detected a sex dimorphism in VAT and SAT in the EW groups regarding 11βHSD1, GRα and C/EBPβ expression. CONCLUSIONS The accumulated adiposity induced by EW exhibited distinct mechanisms depending on gender, specific fat deposition and GC metabolism and action. The higher proportion of VAT/SAT in both sets of EW males may be related to the action of GC in these tissues, and the higher conversion of GC in SAT in females may explain the differences in the fat distribution.
Collapse
Affiliation(s)
- Rosiane Aparecida Miranda
- Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Carla Bruna Pietrobon
- Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Iala Milene Bertasso
- Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Vanessa S Tavares Rodrigues
- Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Bruna Pereira Lopes
- Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Camila Calvino
- Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Elaine de Oliveira
- Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Patrícia C Lisboa
- Laboratory of Endocrine Physiology, Biology Institute, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
66
|
Wu L, Song Y, Zhang Y, Liang B, Deng Y, Tang T, Ye YC, Hou HY, Wang CC. Novel Genetic Variants of PPARγ2 Promoter in Gestational Diabetes Mellitus and its Molecular Regulation in Adipogenesis. Front Endocrinol (Lausanne) 2020; 11:499788. [PMID: 33551986 PMCID: PMC7862745 DOI: 10.3389/fendo.2020.499788] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 11/25/2020] [Indexed: 11/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ2 (PPARγ2) is a nuclear hormone receptor of ligand-dependent transcription factor with a key role in adipogenesis and insulin sensitization in diabetes mellitus. In this study, we investigated genetic variants in PPARγ2 promoter, its association with gestational diabetes mellitus (GDM), and its molecular regulation. PPARγ2 promoter and start codon (-2,091 to +82 bp) from 400 pregnancies with GDM and 400 gestational-age matched control pregnancies were sequenced. Association and linkage disequilibrium of the identified polymorphisms with GDM was determined. ChIP-seq, gene silencing, and dual-luciferase reporter assays were performed to confirm transcription factor binding sites and promoter activity of the variants. Transfection experiments were carried out to determine the effects of variants on gene expression and adipogenesis. Among 15 variants identified, 7 known variants were not significantly associated with the risk of GDM (odds ratio: 0.710-1.208, 95% confidence interval: 0.445-0.877 to 1.132-1.664, P > 0.05) while linkage disequilibrium was significant (D' > 0.7, R2 > 0.9). However, T-A-A-T-G haplotype was not significantly associated with GDM (χ2 = 2.461, P = 0.117). Five rare variants and 3 novel variants (rs948820149, rs1553638909, and rs1553638903) were only found in GDM. Transcription factor glucocorticoid receptor β (GRβ) bound to -807A/C (rs948820149) and knockdown of GRβ suppressed PPARγ2 promoter activity. This mutation significantly down-regulated PPARγ2 expression and alleviated adipogenesis. In conclusion, a novel -807A/C in PPARγ2 promoter was identified in Chinese women with GDM and the mutation affected GRβ binding and transcription of PPARγ2 for adipogenesis.
Collapse
Affiliation(s)
- Ling Wu
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Yi Song
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Bo Liang
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Yan Deng
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tao Tang
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Yan Chou Ye
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hong Ying Hou
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chi Chiu Wang
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Development and Reproduction Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Chinese University of Hong Kong-Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- *Correspondence: Chi Chiu Wang,
| |
Collapse
|
67
|
Sharma AK, Shi X, Isales CM, McGee-Lawrence ME. Endogenous Glucocorticoid Signaling in the Regulation of Bone and Marrow Adiposity: Lessons from Metabolism and Cross Talk in Other Tissues. Curr Osteoporos Rep 2019; 17:438-445. [PMID: 31749087 DOI: 10.1007/s11914-019-00554-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW The development of adiposity in the bone marrow, known as marrow adipose tissue (MAT), is often associated with musculoskeletal frailty. Glucocorticoids, which are a key component of the biological response to stress, affect both bone and MAT. These molecules signal through receptors such as the glucocorticoid receptor (GR), but the role of the GR in regulation of MAT is not yet clear from previous studies. The purpose of this review is to establish and determine the role of GR-mediated signaling in marrow adiposity by comparing and contrasting what is known against other energy-storing tissues like adipose tissue, liver, and muscle, to provide better insight into the regulation of MAT during times of metabolic stress (e.g., dietary challenges, aging). RECENT FINDINGS GR-mediated glucocorticoid signaling is critical for proper storage and utilization of lipids in cells such as adipocytes and hepatocytes and proteolysis in muscle, impacting whole-body composition, energy utilization, and homeostasis through a complex network of tissue cross talk between these systems. Loss of GR signaling in bone promotes increased MAT and decreased bone mass. GR-mediated signaling in the liver, adipose tissue, and muscle is critical for whole-body energy and metabolic homeostasis, and both similarities and differences in GR-mediated GC signaling in MAT as compared with these tissues are readily apparent. It is clear that GC-induced pathways work together through these tissues to affect systemic biology, and understanding the role of bone in these patterns of tissue cross talk may lead to a better understanding of MAT-bone biology that improves treatment strategies for frailty-associated diseases.
Collapse
Affiliation(s)
- Anuj K Sharma
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB1101, Augusta, GA, USA
| | - Xingming Shi
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, USA
| | - Carlos M Isales
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, USA
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Augusta University, Augusta, GA, USA
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, 1460 Laney Walker Blvd., CB1101, Augusta, GA, USA.
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA.
| |
Collapse
|
68
|
Pacholko AG, Wotton CA, Bekar LK. Poor Diet, Stress, and Inactivity Converge to Form a "Perfect Storm" That Drives Alzheimer's Disease Pathogenesis. NEURODEGENER DIS 2019; 19:60-77. [PMID: 31600762 DOI: 10.1159/000503451] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/17/2019] [Indexed: 11/19/2022] Open
Abstract
North American incidence of Alzheimer's disease (AD) is expected to more than double over the coming generation. Although genetic factors surrounding the production and clearance of amyloid-β and phosphorylated tau proteins are known to be responsible for a subset of early-onset AD cases, they do not explain the pathogenesis of the far more prevalent sporadic late-onset variant of the disease. It is thus likely that lifestyle and environmental factors contribute to neurodegenerative processes implicated in the pathogenesis of AD. Herein, we review evidence that (1) excess sucrose consumption induces AD-associated liver pathologies and brain insulin resistance, (2) chronic stress overdrives activity of locus coeruleus neurons, leading to loss of function (a common event in neurodegeneration), (3) high-sugar diets and stress promote the loss of neuroprotective sex hormones in men and women, and (4) Western dietary trends set the stage for a lithium-deficient state. We propose that these factors may intersect as part of a "perfect storm" to contribute to the widespread prevalence of neurodegeneration and AD. In addition, we put forth the argument that exercise and supplementation with trace lithium can counteract many of the deleterious consequences associated with excessive caloric intake and perpetual stress. We conclude that lifestyle and environmental factors likely contribute to AD pathogenesis and that simple lifestyle and dietary changes can help counteract their effects.
Collapse
Affiliation(s)
- Anthony G Pacholko
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Caitlin A Wotton
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Lane K Bekar
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada,
| |
Collapse
|
69
|
Faught E, Vijayan MM. Postnatal triglyceride accumulation is regulated by mineralocorticoid receptor activation under basal and stress conditions. J Physiol 2019; 597:4927-4941. [DOI: 10.1113/jp278088] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/26/2019] [Indexed: 12/17/2022] Open
Affiliation(s)
- Erin Faught
- Department of Biological SciencesUniversity of Calgary Calgary Alberta Canada
| | | |
Collapse
|
70
|
Lee IT, Atuahene A, Egritag HE, Wang L, Donovan M, Buettner C, Geer EB. Active Cushing Disease Is Characterized by Increased Adipose Tissue Macrophage Presence. J Clin Endocrinol Metab 2019; 104:2453-2461. [PMID: 30722035 PMCID: PMC6510019 DOI: 10.1210/jc.2018-02552] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/30/2019] [Indexed: 01/04/2023]
Abstract
CONTEXT Although glucocorticoids (GCs) have potent anti-inflammatory actions, patients with hypercortisolism due to Cushing disease (CD) have increased circulating proinflammatory cytokines that may contribute to their insulin resistance and cardiovascular disease. The mechanisms and tissues that account for the increased systemic inflammation in patients with CD are unknown. OBJECTIVE To determine whether chronic endogenous GC exposure due to CD is associated with adipose tissue (AT) inflammation in humans. DESIGN, SETTING, PARTICIPANTS Abdominal subcutaneous AT samples from 10 patients with active CD and 10 age-, sex-, and body mass index‒matched healthy subjects were assessed for macrophage infiltration and mRNA expression of proinflammatory cytokines. MAIN OUTCOME MEASURE Using immunohistochemistry, AT samples were analyzed for the expression of vimentin, caspase, CD3, CD4, CD8, CD11c, CD20, CD31, CD56, CD68, and CD163. Quantitative PCR was used to assess the mRNA gene expression of arginase, CD11b, CD68, EMR-1, IL-6, IL-10, MCP-1, and TNF-α. RESULTS Immunohistochemistry revealed higher mean percentage infiltration of CD68+ macrophages and CD4+ T lymphocytes, increased mean area of CD11c+ M1 macrophages, higher number of CD11c+ crownlike structures, and decreased vimentin in the AT of patients with active CD compared with controls. PCR revealed no differences in mRNA expression of any analyzed markers in patients with CD. CONCLUSIONS Chronic exposure to GCs due to CD increases the presence of AT macrophages, a hallmark of AT inflammation. Hence, AT inflammation may be the source of the systemic inflammation seen in CD, which in turn may contribute to obesity, insulin resistance, and cardiovascular disease in these patients.
Collapse
Affiliation(s)
- Irene T Lee
- Division of Endocrinology, Metabolism and Diabetes, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexandria Atuahene
- Division of Endocrinology, Metabolism and Diabetes, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hale Ergin Egritag
- Division of Endocrinology, Metabolism and Diabetes, Icahn School of Medicine at Mount Sinai, New York, New York
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ling Wang
- Division of Endocrinology, Metabolism and Diabetes, Icahn School of Medicine at Mount Sinai, New York, New York
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michael Donovan
- Division of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christoph Buettner
- Division of Endocrinology, Metabolism and Diabetes, Icahn School of Medicine at Mount Sinai, New York, New York
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eliza B Geer
- Division of Endocrinology, Metabolism and Diabetes, Icahn School of Medicine at Mount Sinai, New York, New York
- Division of Endocrinology, Department of Medicine, and Multidisciplinary Pituitary and Skull Base Tumor Center, Memorial Sloan-Kettering Cancer Center, New York, New York
- Correspondence and Reprint Requests: Eliza B. Geer, MD, Multidisciplinary Pituitary and Skull Base Tumor Center, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, Box 419, New York, New York 10065. E-mail:
| |
Collapse
|
71
|
Lv J, Ma Q, Dasgupta C, Xu Z, Zhang L. Antenatal Hypoxia and Programming of Glucocorticoid Receptor Expression in the Adult Rat Heart. Front Physiol 2019; 10:323. [PMID: 31001129 PMCID: PMC6454194 DOI: 10.3389/fphys.2019.00323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/11/2019] [Indexed: 12/26/2022] Open
Abstract
Glucocorticoid receptor (GR) signaling is critical for development and function of the heart. Our previous study demonstrated that gestational hypoxia induced epigenetic repression of the GR gene in the developing heart. The present study aims to determine that the alterations of promoter methylation level and epigenetic repression of the GR gene in the developing heart in response to maternal hypoxia is sustained in adult offspring and potential gender differences in the programming of GR gene. Pregnant rats were treated with 10.5% O2 from gestational day 15 (E15) to 21 (E21). Hearts were isolated from 5-month-old male and female offspring with the developing stage being equivalent to 18-year-old human. GR mRNA and protein abundance was determined with real time qRT-PCR and Western blot. GR gene promoter methylation and binding of transcription factors were measured with methylated DNA immunoprecipitation (MeDIP) and Chromatin immunoprecipitation (ChIP). The results showed that antenatal hypoxia significantly decreased the expression of GR mRNA and protein in the hearts of adult male offspring, but not in females, which is ascribed to the differential changes of alternative exon1 mRNA variants of GR gene in male and female hearts in response to prenatal hypoxia. In addition, the downregulation of GR expression in the male heart was correlated with increased methylation levels of CpG dinucleotides in promoters of exon 14, 15, 16, 17, and 110, which resulted in a decrease in the binding of their transcription factors. Thus, the study reveals that antenatal hypoxia results in a reprogramming and long-term change in GR gene expression in the heart by hypermethylation of GR promoter in a sex-differential pattern, which provides a novel mechanism regarding the increased vulnerability of heart later in life with exposure of prenatal hypoxia.
Collapse
Affiliation(s)
- Juanxiu Lv
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qingyi Ma
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Chiranjib Dasgupta
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Zhice Xu
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Lubo Zhang
- Institute for Fetology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, United States
| |
Collapse
|
72
|
Deyarmin JS, McCormley MC, Champagne CD, Stephan AP, Busqueta LP, Crocker DE, Houser DS, Khudyakov JI. Blubber transcriptome responses to repeated ACTH administration in a marine mammal. Sci Rep 2019; 9:2718. [PMID: 30804370 PMCID: PMC6390094 DOI: 10.1038/s41598-019-39089-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/16/2019] [Indexed: 01/03/2023] Open
Abstract
Chronic physiological stress impacts animal fitness by catabolizing metabolic stores and suppressing reproduction. This can be especially deleterious for capital breeding carnivores such as marine mammals, with potential for ecosystem-wide effects. However, the impacts and indicators of chronic stress in animals are currently poorly understood. To identify downstream mediators of repeated stress responses in marine mammals, we administered adrenocorticotropic hormone (ACTH) once daily for four days to free-ranging juvenile northern elephant seals (Mirounga angustirostris) to stimulate endogenous corticosteroid release, and compared blubber tissue transcriptome responses to the first and fourth ACTH administrations. Gene expression profiles were distinct between blubber responses to single and repeated ACTH administration, despite similarities in circulating cortisol profiles. We identified 61 and 12 genes that were differentially expressed (DEGs) in response to the first ACTH and fourth administrations, respectively, 24 DEGs between the first and fourth pre-ACTH samples, and 12 DEGs between ACTH response samples from the first and fourth days. Annotated DEGs were associated with functions in redox and lipid homeostasis, suggesting potential negative impacts of repeated stress on capital breeding, diving mammals. DEGs identified in this study are potential markers of repeated stress in marine mammals, which may not be detectable by endocrine profiles alone.
Collapse
Affiliation(s)
- Jared S Deyarmin
- Department of Biological Sciences, University of the Pacific, Stockton, CA, 95211, USA
| | - Molly C McCormley
- Department of Biological Sciences, University of the Pacific, Stockton, CA, 95211, USA
| | - Cory D Champagne
- Conservation and Biological Research Program, National Marine Mammal Foundation, San Diego, CA, 92106, USA
| | - Alicia P Stephan
- Department of Biological Sciences, University of the Pacific, Stockton, CA, 95211, USA
| | - Laura Pujade Busqueta
- Department of Biological Sciences, University of the Pacific, Stockton, CA, 95211, USA
| | - Daniel E Crocker
- Biology Department, Sonoma State University, Rohnert Park, CA, 94928, USA
| | - Dorian S Houser
- Conservation and Biological Research Program, National Marine Mammal Foundation, San Diego, CA, 92106, USA
| | - Jane I Khudyakov
- Department of Biological Sciences, University of the Pacific, Stockton, CA, 95211, USA.
- Conservation and Biological Research Program, National Marine Mammal Foundation, San Diego, CA, 92106, USA.
| |
Collapse
|
73
|
Leventhal SM, Lim D, Green TL, Cantrell AE, Cho K, Greenhalgh DG. Uncovering a multitude of human glucocorticoid receptor variants: an expansive survey of a single gene. BMC Genet 2019; 20:16. [PMID: 30736733 PMCID: PMC6368729 DOI: 10.1186/s12863-019-0718-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 01/23/2019] [Indexed: 12/26/2022] Open
Abstract
Background Glucocorticoids are commonly used in the clinical setting for their potent anti-inflammatory effects; however, significant variations in response to treatment have been demonstrated. Although the underlying mechanisms have yet to be fully understood, this variable response may be a result of alterations in human glucocorticoid receptor (hGR) expression and function. In addition to hGRα, the biologically active isoform, a screening of current databases and publications revealed five alternative splice isoforms and hundreds of variants that have been reported to date. Many of these changes in the hGR-coding gene, NR3C1, have been linked to pathophysiology. However, many studies focus on evaluating hGR expression in vitro or detecting previously reported variants. Results In this study, blood from healthy volunteers, burn and asthma patients, as well as from peripheral blood mononuclear cells isolated from leukoreduced donor whole blood, were screened for NR3C1 isoforms. We identified more than 1500 variants, including an additional 21 unique splice isoforms which contain 15 new cryptic exons. A dynamic database, named the Universal hGR (UhGR), was created to annotate and visualize the variants. Conclusion This identification of naturally occurring and stress-induced hGR isoforms, as well as the establishment of an hGR-specific database, may reveal new patterns or suggest areas of interest that will lead to the improved understanding of the human stress response system. Electronic supplementary material The online version of this article (10.1186/s12863-019-0718-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stacey M Leventhal
- Shriners Hospitals for Children Northern California, Sacramento, California, USA
| | - Debora Lim
- Department of Surgery, University of California, Davis, Sacramento, California, USA
| | - Tajia L Green
- Shriners Hospitals for Children Northern California, Sacramento, California, USA
| | - Anna E Cantrell
- Department of Surgery, University of California, Davis, Sacramento, California, USA
| | - Kiho Cho
- Shriners Hospitals for Children Northern California, Sacramento, California, USA. .,Department of Surgery, University of California, Davis, Sacramento, California, USA.
| | - David G Greenhalgh
- Shriners Hospitals for Children Northern California, Sacramento, California, USA. .,Department of Surgery, University of California, Davis, Sacramento, California, USA.
| |
Collapse
|
74
|
Kassotis CD, Stapleton HM. Endocrine-Mediated Mechanisms of Metabolic Disruption and New Approaches to Examine the Public Health Threat. Front Endocrinol (Lausanne) 2019; 10:39. [PMID: 30792693 PMCID: PMC6374316 DOI: 10.3389/fendo.2019.00039] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/17/2019] [Indexed: 01/29/2023] Open
Abstract
Obesity and metabolic disorders are of great societal concern and generate substantial human health care costs globally. Interventions have resulted in only minimal impacts on disrupting this worsening health trend, increasing attention on putative environmental contributors. Exposure to numerous environmental contaminants have, over decades, been demonstrated to result in increased metabolic dysfunction and/or weight gain in cell and animal models, and in some cases, even in humans. There are numerous mechanisms through which environmental contaminants may contribute to metabolic dysfunction, though certain mechanisms, such as activation of the peroxisome proliferator activated receptor gamma or the retinoid x receptor, have received considerably more attention than less-studied mechanisms such as antagonism of the thyroid receptor, androgen receptor, or mitochondrial toxicity. As such, research on putative metabolic disruptors is growing rapidly, as is our understanding of molecular mechanisms underlying these effects. Concurrent with these advances, new research has evaluated current models of adipogenesis, and new models have been proposed. Only in the last several years have studies really begun to address complex mixtures of contaminants and how these mixtures may disrupt metabolic health in environmentally relevant exposure scenarios. Several studies have begun to assess environmental mixtures from various environments and study the mechanisms underlying their putative metabolic dysfunction; these studies hold real promise in highlighting crucial mechanisms driving observed organismal effects. In addition, high-throughput toxicity databases (ToxCast, etc.) may provide future benefits in prioritizing chemicals for in vivo testing, particularly once the causative molecular mechanisms promoting dysfunction are better understood and expert critiques are used to hone the databases. In this review, we will review the available literature linking metabolic disruption to endocrine-mediated molecular mechanisms, discuss the novel application of environmental mixtures and implications for in vivo metabolic health, and discuss the putative utility of applying high-throughput toxicity databases to answering complex organismal health outcome questions.
Collapse
|
75
|
Franco LM, Gadkari M, Howe KN, Sun J, Kardava L, Kumar P, Kumari S, Hu Z, Fraser IDC, Moir S, Tsang JS, Germain RN. Immune regulation by glucocorticoids can be linked to cell type-dependent transcriptional responses. J Exp Med 2019; 216:384-406. [PMID: 30674564 PMCID: PMC6363437 DOI: 10.1084/jem.20180595] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 10/22/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022] Open
Abstract
A functional genomics approach uncovers previously undescribed cell type–dependent responses that can be linked to the immunoregulatory actions of glucocorticoids in humans. Glucocorticoids remain the most widely used immunosuppressive and anti-inflammatory drugs, yet substantial gaps exist in our understanding of glucocorticoid-mediated immunoregulation. To address this, we generated a pathway-level map of the transcriptional effects of glucocorticoids on nine primary human cell types. This analysis revealed that the response to glucocorticoids is highly cell type dependent, in terms of the individual genes and pathways affected, as well as the magnitude and direction of transcriptional regulation. Based on these data and given their importance in autoimmunity, we conducted functional studies with B cells. We found that glucocorticoids impair upstream B cell receptor and Toll-like receptor 7 signaling, reduce transcriptional output from the three immunoglobulin loci, and promote significant up-regulation of the genes encoding the immunomodulatory cytokine IL-10 and the terminal-differentiation factor BLIMP-1. These findings provide new mechanistic understanding of glucocorticoid action and emphasize the multifactorial, cell-specific effects of these drugs, with potential implications for designing more selective immunoregulatory therapies.
Collapse
Affiliation(s)
- Luis M Franco
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Manasi Gadkari
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Katherine N Howe
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Jing Sun
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Parag Kumar
- Clinical Pharmacokinetics Research Unit, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Sangeeta Kumari
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Zonghui Hu
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Iain D C Fraser
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - John S Tsang
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,Center for Human Immunology, National Institutes of Health, Bethesda, MD
| | - Ronald N Germain
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
76
|
Sluzalska KD, Liebisch G, Ishaque B, Schmitz G, Rickert M, Steinmeyer J. The Effect of Dexamethasone, Adrenergic and Cholinergic Receptor Agonists on Phospholipid Metabolism in Human Osteoarthritic Synoviocytes. Int J Mol Sci 2019; 20:ijms20020342. [PMID: 30650648 PMCID: PMC6359197 DOI: 10.3390/ijms20020342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/09/2019] [Accepted: 01/11/2019] [Indexed: 01/07/2023] Open
Abstract
Phospholipids (PLs) possess the unique ability to contribute to synovial joint lubrication. The aim of our study was to determine for the first time the effect of dexamethasone and some adrenergic and cholinergic agonists on the biosynthesis and release of PLs from human fibroblast-like synoviocytes (FLS). Osteoarthritic human knee FLS were treated with dexamethasone, terbutaline, epinephrine, carbachol, and pilocarpine, or the glucocorticoid receptor antagonist RU 486. Simultaneously PL biosynthesis was determined through the incorporation of stable isotope-labeled precursors into PLs. Radioactive isotope-labeled precursors were used to radiolabel PLs for the subsequent quantification of their release into nutrient media. Lipids were extracted and quantified using electrospray ionization tandem mass spectrometry or liquid scintillation counting. Dexamethasone significantly decreased the biosynthesis of phosphatidylcholine, phosphatidylethanolamine (PE), PE-based plasmalogen, and sphingomyelin. The addition of RU 486 abolished these effects. A release of PLs from FLS into nutrient media was not recognized by any of the tested agents. None of the adrenergic or cholinergic receptor agonists modulated the PL biosynthesis. We demonstrate for the first time an inhibitory effect of dexamethasone on the PL biosynthesis of FLS from human knees. Moreover, our study indicates that the PL metabolism of synovial joints and lungs are differently regulated.
Collapse
Affiliation(s)
- Katarzyna D Sluzalska
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany.
| | - Gerhard Liebisch
- Department of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany.
| | - Bernd Ishaque
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany.
| | - Gerd Schmitz
- Department of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany.
| | - Markus Rickert
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany.
| | - Juergen Steinmeyer
- Laboratory for Experimental Orthopaedics, Department of Orthopaedics, Justus Liebig University Giessen, 35392 Giessen, Germany.
| |
Collapse
|
77
|
Olczak E, Kuryłowicz A, Wicik Z, Kołodziej P, Cąkała-Jakimowicz M, Buyanovskaya O, Ślusarczyk P, Mossakowska M, Puzianowska-Kuźnicka M. Glucocorticoid receptor (NR3C1) gene polymorphisms are associated with age and blood parameters in Polish Caucasian nonagenarians and centenarians. Exp Gerontol 2018; 116:20-24. [PMID: 30553025 DOI: 10.1016/j.exger.2018.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 11/23/2018] [Accepted: 12/04/2018] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Polymorphism of the glucocorticoid receptor gene (NR3C1) may modify protein abundance or function and therefore disturb human homeostasis. METHODS Variant frequencies of the three NR3C1 polymorphisms, rs2963154, rs10515522 and rs2918418, selected in silico as associated with longevity, was analyzed in 552 DNA samples from 95 to 106-year-old individuals and in 284 samples of cord blood DNA from newborns. RESULTS Frequencies of the TT genotypes of rs2963154 and rs10515522, and of the rs291841 CC genotype, were higher in the long-lived study subjects (p = 0.002, p = 0.016 and p = 0.028, respectively). In the long-lived cohort, the rs2963154 CC genotype was associated with higher concentrations of total (p = 0.007) and high-density cholesterol (p = 0.039). The rs10515522 CC genotype was associated with a higher concentration of total cholesterol (p = 0.049). The rs2918418 GG genotype was associated with higher concentrations of total (p = 0.03) and low-density cholesterol (p = 0.03). None of the polymorphisms was associated with fasting glucose, C-reactive protein levels and white blood count, prevalence of diabetes, stroke, myocardial infarction, or cognitive function. However, carriers of the rs10515522 minor allele had significantly better survival rates than carriers of other genotypes. CONCLUSION NR3C1 polymorphisms modify cholesterol levels, and may affect the survival rates of individuals in their tenth and eleventh decades of life.
Collapse
Affiliation(s)
- Elżbieta Olczak
- Warsaw University of Life Sciences, Nowoursynowska 166, 02-776 Warsaw, Poland
| | - Alina Kuryłowicz
- Mossakowski Medical Research Centre, PAS, Pawinskiego 5, 02-106 Warsaw, Poland.
| | - Zofia Wicik
- Mossakowski Medical Research Centre, PAS, Pawinskiego 5, 02-106 Warsaw, Poland.
| | - Paulina Kołodziej
- Medical Centre of Postgraduate Education, Marymoncka 99/103, 01-813 Warsaw, Poland
| | | | - Olga Buyanovskaya
- Mossakowski Medical Research Centre, PAS, Pawinskiego 5, 02-106 Warsaw, Poland.
| | - Przemyslaw Ślusarczyk
- International Institute of Molecular and Cell Biology, Księcia Trojdena 4, 02-109 Warsaw, Poland..
| | - Malgorzata Mossakowska
- International Institute of Molecular and Cell Biology, Księcia Trojdena 4, 02-109 Warsaw, Poland..
| | - Monika Puzianowska-Kuźnicka
- Mossakowski Medical Research Centre, PAS, Pawinskiego 5, 02-106 Warsaw, Poland; Medical Centre of Postgraduate Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
| |
Collapse
|
78
|
Cigarette Smoke During Breastfeeding in Rats Changes Glucocorticoid and Vitamin D Status in Obese Adult Offspring. Int J Mol Sci 2018; 19:ijms19103084. [PMID: 30304827 PMCID: PMC6213898 DOI: 10.3390/ijms19103084] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 09/28/2018] [Accepted: 10/05/2018] [Indexed: 12/11/2022] Open
Abstract
Maternal smoking increases obesogenesis in the progeny. Obesity is associated with several hormonal dysfunctions. In a rat model of postnatal tobacco smoke exposure, we previously reported increased central fat depot and disruption of some hormonal systems in the adult offspring. As both glucocorticoids and vitamin D alter lipogenesis and adipogenesis, here we evaluated the metabolism of these two hormones in visceral adipose tissue (VAT) and liver by Western blotting, and possible associations with lipogenesis biomarkers in adult rats that were exposed to tobacco smoke during their suckling period. At postnatal day (PN) 3, dams and offspring of both sexes were exposed (S group) or not (C group) to tobacco smoke, 4 × 1 h/day. At PN180, corticosteronemia was lower in S male and higher in S female offspring, without alterations in peripheral glucocorticoid metabolism and receptor. Adrenal ACTH receptor (MC2R) was higher in both sexes of S group. Despite unchanged serum vitamin D, liver 25-hydroxylase was higher in both sexes of S group. Male S offspring had higher 1α-hydroxylase, acetyl-CoA carboxylase (ACC), and fatty acid synthase (FAS) in VAT. Both sexes showed increased ACC protein content and reduced sirtuin mRNA in liver. Male S offspring had lower liver peroxisome proliferator-activated receptor-α. Tobacco exposure during lactation induced abdominal obesity in both sexes via distinct mechanisms. Males and females seem to develop HPA-axis dysfunction instead of changes in glucocorticoid metabolism and action. Lipogenesis in VAT and liver, as well as vitamin D status, are more influenced by postnatal smoke exposure in male than in female adult rat offspring.
Collapse
|
79
|
Cystathionine beta synthase-hydrogen sulfide system in paraventricular nucleus reduced high fatty diet induced obesity and insulin resistance by brain-adipose axis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3281-3291. [DOI: 10.1016/j.bbadis.2018.07.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 12/28/2022]
|
80
|
Veiga-Lopez A, Pu Y, Gingrich J, Padmanabhan V. Obesogenic Endocrine Disrupting Chemicals: Identifying Knowledge Gaps. Trends Endocrinol Metab 2018; 29:607-625. [PMID: 30017741 PMCID: PMC6098722 DOI: 10.1016/j.tem.2018.06.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/11/2018] [Accepted: 06/14/2018] [Indexed: 02/07/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are compounds that are part of everyday consumer products and industrial manufacturing processes. EDCs can interfere with the endocrine system, including the adipose tissue. Accumulating evidence from epidemiological, animal, and in vitro studies demonstrates that EDCs can alter body weight, adipose tissue expansion, circulating lipid profile, and adipogenesis, with some resulting in transgenerational effects. These outcomes appear to be mediated through multiple mechanisms, from nuclear receptor binding to epigenetic modifications. A better understanding of the signaling pathways via which these EDCs contribute to an obesogenic phenotype, the interaction amongst complex mixtures of obesogenic EDCs, and the risks they pose relative to the obesity epidemic are still needed for risk assessment and development of prevention strategies.
Collapse
Affiliation(s)
- Almudena Veiga-Lopez
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA.
| | - Yong Pu
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA
| | - Jeremy Gingrich
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, MI 48824, USA; Department of Pharmacology and Toxicology, College of Natural Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
81
|
Freitas FV, Barbosa WM, Silva LAA, Garozi MJDO, Pinheiro JDA, Borçoi AR, Conti CL, Arpini JK, de Paula H, de Oliveira MM, Archanjo AB, de Freitas ÉAS, de Oliveira DR, Borloti EB, Louro ID, Alvares-da-Silva AM. Psychosocial stress and central adiposity: A Brazilian study with a representative sample of the public health system users. PLoS One 2018; 13:e0197699. [PMID: 30063700 PMCID: PMC6067710 DOI: 10.1371/journal.pone.0197699] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/17/2018] [Indexed: 11/18/2022] Open
Abstract
Objective To assess the association between indicators of psychosocial stress and central adiposity in adult users of the Unified Health System (SUS) from Southeast of Brazil. Methods This cross-sectional study was conducted with 384 adults (20 to 59 years old) from the city of Alegre, Southeastern Brazil. The simple random sample represented the population using the public health system of the municipality. The prevalence of obesity was based on the Body Mass Index, and central adiposity (dependent variable) was measured by waist circumference in centimeters. The independent variables were the following indicators of psychosocial stress: food and nutrition insecurity (yes/no), serum cortisol (μg/dL), symptoms suggestive of depression using the Beck Depression Inventory-II ≥ 17 (yes/no), and altered blood pressure ≥ 130/85 mmHg (yes/no). Univariate linear regression was performed between central adiposity and each stress indicator, and later the models were adjusted for socioeconomic, health, and lifestyle variables. All analyses were made separately by rural and urban location. Results The prevalence of weight excess, by the classification of the Body Mass Index ≥ 25.0 kg/m2, was 68.3% and, by waist circumference, 71.5% of individuals presented an increased risk for metabolic complications related to central adiposity. Mean waist circumference scores for the rural and urban population were 89.3 ± 12.7 cm and 92.9 ± 14.7 cm, respectively (p = 0.012). Indicators of stress that were associated with central adiposity were: cortisol in the rural population (β = -0.60; 95% CI = -1.09;-0.11) and altered blood pressure in the urban population (β = 6.66; 95% CI = 2.14;11.18). This occurred both in the raw analysis and in the models adjusted for confounding factors. Conclusion Central adiposity was inversely associated with cortisol in the rural population and directly associated with higher arterial blood pressure in the urban population, suggesting a local influence on how individuals react to stress.
Collapse
Affiliation(s)
- Flávia Vitorino Freitas
- Biotechnology/Renorbio Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil
- Pharmacy and Nutrition Department, Federal University of Espirito Santo, Alegre, ES, Brazil
- * E-mail:
| | - Wagner Miranda Barbosa
- Pharmacy and Nutrition Department, Federal University of Espirito Santo, Alegre, ES, Brazil
| | | | | | - Júlia de Assis Pinheiro
- Biotechnology/Renorbio Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Aline Ribeiro Borçoi
- Biotechnology/Renorbio Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Catarine Lima Conti
- Biotechnology/Renorbio Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | - Heberth de Paula
- Pharmacy and Nutrition Department, Federal University of Espirito Santo, Alegre, ES, Brazil
| | - Mayara Mota de Oliveira
- Biotechnology/Renorbio Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Anderson Barros Archanjo
- Biotechnology/Renorbio Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | | | | | - Iuri Drumond Louro
- Biotechnology/Renorbio Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Adriana Madeira Alvares-da-Silva
- Biotechnology/Renorbio Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil
- Department of Biology, Federal University of Espirito Santo, Alegre, ES, Brazil
| |
Collapse
|
82
|
Noyin K, Akpinar E, Cadirci E, Cinar I, Aydin P. THE EFFECTS OF VARIOUS DOSES OF PREDNISOLONE ADMINISTRATION ON SERUM VASPIN LEVELS IN RATS. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2018; 14:320-323. [PMID: 31149278 PMCID: PMC6525765 DOI: 10.4183/aeb.2018.320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
AIM The aim of this study was to investigate the dose-dependent effects of prednisolone administration on serum vaspin levels and correlate this with changes in the BMI and lipogenesis in rats. MATERIALS AND METHODS Twenty-four albino Wistar male rats weighing between 190-240 g were divided into four groups, three experimental (5 mg/kg, 10 mg/kg, and 20 mg/kg prednisolone) and one control. The prednisolone groups were given once-daily doses for 30 days, orally. In addition, the rats were weighed, and their height and waist circumferences were measured once a week. At the end of 30 days, vaspin and glucose levels were measured from blood samples. RESULTS In the prednisolone groups, the vaspin levels significantly increased when compared with the control group. The control group has a serum vaspin level of 155 ± 20.99 pg/mL and this level has been increased by prednisolone administration in a dose dependent manner. In the prednisolone groups, especially the 10 mg/kg and 20 mg/kg groups, the glucose levels increased in a dose dependent fashion. CONCLUSION Prednisolone administration significantly increased serum glucose and vaspin levels in a dose dependent manner, indicating that the increase in the serum vaspin levels could be related to the increase in the serum glucose concentration. Vaspin can be a molecule that is released in response to increased glucose and can be a rebound defense mechanism to modulate the blood glucose concentration. We suggest vaspin as a potential target for the treatment and diagnosis of diabetes mellitus and other metabolic disorders.
Collapse
Affiliation(s)
- K. Noyin
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - E. Akpinar
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - E. Cadirci
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - I. Cinar
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - P. Aydin
- Erzurum Regional Training and Research Hospital, Department of Anesthesiology and Reanimation, Erzurum, Turkey
| |
Collapse
|
83
|
Hu Y, Sun Q, Hu Y, Hou Z, Zong Y, Omer NA, Abobaker H, Zhao R. Corticosterone-Induced Lipogenesis Activation and Lipophagy Inhibition in Chicken Liver Are Alleviated by Maternal Betaine Supplementation. J Nutr 2018; 148:316-325. [PMID: 29546310 DOI: 10.1093/jn/nxx073] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Background We have shown previously that in ovo betaine injection can prevent nonalcoholic fatty liver induced by glucocorticoid exposure in chickens; yet it remains unknown whether feeding betaine to laying hens may exert similar effects in their progeny. Objective In this study, we fed laying hens a betaine-supplemented diet, and the progeny were later exposed chronically to corticosterone (CORT) to test hepatoprotective effects and further elucidate underlying mechanisms. Methods Rugao yellow-feathered laying hens (n = 120) were fed a basal (control, C) diet or a 0.5% betaine-supplemented (B) diet for 28 d before their eggs were collected for incubation. At 49 d of age, male chickens selected from each group were daily injected subcutaneously with solvent (15% ethanol; vehicle, VEH) or CORT (4.0 mg/kg body mass) for 7 d to establish a fatty liver model. Chickens in the 4 groups (C-VEH, C-CORT, B-VEH, and B-CORT) were killed at day 57. Plasma and hepatic triglyceride (TG) concentrations, as well as the hepatic expression of genes involved in lipogenesis and lipophagy, were determined. Results CORT induced a 1.6-fold increase in the plasma TG concentration (P < 0.05) and a 1.8-fold increment in the hepatic TG concentration (P < 0.05), associated with activation of lipogenic genes (70-780%). In contrast, lipophagy and mitochondrial β-oxidation genes were inhibited by 30-60% (P < 0.05) in CORT-treated chickens. These CORT-induced changes were completely normalized by maternal betaine supplementation or were partially normalized to intermediate values that were significantly different from those in the C-VEH and C-CORT groups. These effects were accompanied by modifications in CpG methylation and glucocorticoid receptor binding to the promoters of major lipogenic and lipophagic genes (P < 0.05). Conclusions These results indicate that maternal betaine supplementation protects male juvenile chickens from CORT-induced TG accumulation in the liver via epigenetic modulation of lipogenic and lipophagic genes.
Collapse
Affiliation(s)
- Yun Hu
- MOE Joint International Research Laboratory of Animal Health & Food Safety
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Qinwei Sun
- MOE Joint International Research Laboratory of Animal Health & Food Safety
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Yan Hu
- Poultry Institute, Chinese Academy of Agriculture Science, Yangzhou, Jiangsu, China
| | - Zhen Hou
- MOE Joint International Research Laboratory of Animal Health & Food Safety
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Yibo Zong
- MOE Joint International Research Laboratory of Animal Health & Food Safety
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Nagmeldin A Omer
- MOE Joint International Research Laboratory of Animal Health & Food Safety
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Halima Abobaker
- MOE Joint International Research Laboratory of Animal Health & Food Safety
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| |
Collapse
|
84
|
Chijimatsu R, Kobayashi M, Ebina K, Iwahashi T, Okuno Y, Hirao M, Fukuhara A, Nakamura N, Yoshikawa H. Impact of dexamethasone concentration on cartilage tissue formation from human synovial derived stem cells in vitro. Cytotechnology 2018; 70:819-829. [PMID: 29352392 DOI: 10.1007/s10616-018-0191-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 01/11/2018] [Indexed: 02/05/2023] Open
Abstract
Human synovial mesenchymal stem cells (hSMSCs) are a promising cell source for cartilage regeneration because of their superior chondrogenic potential in vitro. This study aimed to further optimize the conditions for inducing chondrogenesis of hSMSCs, focusing on the dose of dexamethasone in combination with transforming growth factor-β3 (TGFβ3) and/or bone morphogenetic protein-2 (BMP2). When hSMSCs-derived aggregates were cultured with TGFβ3, dexamethasone up to 10 nM promoted chondrogenesis, but attenuated it with heterogeneous tissue formation when used at concentrations over than 100 nM. On the other hands, BMP2-induced chondrogenesis was remarkably disturbed in the presence of more than 10 nM dexamethasone along with unexpected adipogenic differentiation. In the presence of both TGFβ3 and BMP2, dexamethasone dose dependently promoted cartilaginous tissue formation as judged by tissue volume, proteoglycan content, and type 2 collagen expression, whereas few adipocytes were detected in the formed tissue when cultures were supplemented with over 100 nM dexamethasone. Even in chondrogenic conditions, dexamethasone thus affected hSMSCs differentiation not only toward chondrocytes, but also towards adipocytes dependent on the dose and combined growth factor. These findings have important implications regarding the use of glucocorticoids in in vitro tissue engineering for cartilage regeneration using hSMSCs.
Collapse
Affiliation(s)
- Ryota Chijimatsu
- Graduate School of Medicine, Orthopaedic Surgery, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Masato Kobayashi
- Graduate School of Medicine, Orthopaedic Surgery, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Kosuke Ebina
- Graduate School of Medicine, Orthopaedic Surgery, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan.
| | - Toru Iwahashi
- Graduate School of Medicine, Orthopaedic Surgery, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Yosuke Okuno
- Graduate School of Medicine, Metabolic Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Makoto Hirao
- Graduate School of Medicine, Orthopaedic Surgery, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Atsunori Fukuhara
- Graduate School of Medicine, Metabolic Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| | - Norimasa Nakamura
- Graduate School of Medicine, Orthopaedic Surgery, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
- Institute for Medical Science in Sports, Osaka Health Science University, 1-9-27 Kita-ku Tenma, Osaka, Osaka, Japan
- Center for Advanced Medical Engineering and Informatics, Osaka University, 1-1 Yamadaoka, Suita, Osaka, Japan
| | - Hideki Yoshikawa
- Graduate School of Medicine, Orthopaedic Surgery, Osaka University, 2-2 Yamadaoka, Suita, Osaka, Japan
| |
Collapse
|
85
|
Krupková M, Liška F, Kazdová L, Šedová L, Kábelová A, Křenová D, Křen V, Šeda O. Single-Gene Congenic Strain Reveals the Effect of Zbtb16 on Dexamethasone-Induced Insulin Resistance. Front Endocrinol (Lausanne) 2018; 9:185. [PMID: 29731739 PMCID: PMC5919955 DOI: 10.3389/fendo.2018.00185] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/05/2018] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Glucocorticoids (GCs) are potent therapeutic agents frequently used for treatment of number of conditions, including hematologic, inflammatory, and allergic diseases. Both their therapeutic and adverse effects display significant interindividual variation, partially attributable to genetic factors. We have previously isolated a seven-gene region of rat chromosome 8 sensitizing to dexamethasone (DEX)-induced dyslipidemia and insulin resistance (IR) of skeletal muscle. Using two newly derived congenic strains, we aimed to investigate the effect of one of the prime candidates for this pharmacogenetic interaction, the Zbtb16 gene. METHODS Adult male rats of SHR-Lx.PD5PD-Zbtb16 (n = 9) and SHR-Lx.PD5SHR-Zbtb16 (n = 8) were fed standard diet (STD) and subsequently treated with DEX in drinking water (2.6 µg/ml) for 3 days. The morphometric and metabolic profiles of both strains including oral glucose tolerance test, triacylglycerols (TGs), free fatty acids, insulin, and C-reactive protein levels were assessed before and after the DEX treatment. Insulin sensitivity of skeletal muscle and visceral adipose tissue was determined by incorporation of radioactively labeled glucose. RESULTS The differential segment of SHR-Lx.PD5SHR-Zbtb16 rat strain spans 563 kb and contains six genes: Htr3a, Htr3b, Usp28, Zw10, Tmprss5, and part of Drd2. The SHR-Lx.PD5PD-Zbtb16 minimal congenic strain contains only Zbtb16 gene on SHR genomic background and its differential segment spans 254 kb. Total body weight was significantly increased in SHR-Lx.PD5PD-Zbtb16 strain compared with SHR-Lx.PD5SHR-Zbtb16 , however, no differences in the weights of adipose tissue depots were observed. While STD-fed rats of both strains did not show major differences in their metabolic profiles, after DEX treatment the SHR-Lx.PD5PD-Zbtb16 congenic strain showed increased levels of TGs, glucose, and blunted inhibition of lipolysis by insulin. Both basal and insulin-stimulated incorporation of radioactively labeled glucose into skeletal muscle glycogen were significantly reduced in SHR-Lx.PD5PD-Zbtb16 strain, but the insulin sensitivity of adipose tissue was comparable between the two strains. CONCLUSION The metabolic disturbances including impaired glucose tolerance, dyslipidemia, and IR of skeletal muscle observed after DEX treatment in the congenic SHR-Lx.PD5PD-Zbtb16 reveal the Zbtb16 locus as a possible sensitizing factor for side effects of GC therapy.
Collapse
Affiliation(s)
- Michaela Krupková
- The First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, The General Teaching Hospital, Prague, Czechia
| | - František Liška
- The First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, The General Teaching Hospital, Prague, Czechia
| | - Ludmila Kazdová
- Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Lucie Šedová
- The First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, The General Teaching Hospital, Prague, Czechia
- Laboratory of Transgenic Models of Diseases, Division BIOCEV, Institute of Molecular Genetics of the Czech Academy of Sciences, v.v.i., Vestec, Prague, Czechia
| | - Adéla Kábelová
- The First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, The General Teaching Hospital, Prague, Czechia
| | - Drahomíra Křenová
- The First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, The General Teaching Hospital, Prague, Czechia
| | - Vladimír Křen
- The First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, The General Teaching Hospital, Prague, Czechia
| | - Ondřej Šeda
- The First Faculty of Medicine, Institute of Biology and Medical Genetics, Charles University, The General Teaching Hospital, Prague, Czechia
- *Correspondence: Ondřej Šeda,
| |
Collapse
|
86
|
de Kloet AD, Herman JP. Fat-brain connections: Adipocyte glucocorticoid control of stress and metabolism. Front Neuroendocrinol 2018; 48:50-57. [PMID: 29042142 DOI: 10.1016/j.yfrne.2017.10.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 01/08/2023]
Abstract
Glucocorticoids act via multiple mechanisms to mobilize energy for maintenance and restoration of homeostasis. In adipose tissue, glucocorticoids can promote lipolysis and facilitate adipocyte differentiation/growth, serving both energy-mobilizing and restorative processes during negative energy balance. Recent data suggest that adipose-dependent feedback may also be involved in regulation of stress responses. Adipocyte glucocorticoid receptor (GR) deletion causes increased HPA axis stress reactivity, due to a loss of negative feedback signals into the CNS. The fat-to-brain signal may be mediated by neuronal mechanisms, release of adipokines or increased lipolysis. The ability of adipose GRs to inhibit psychogenic as well as metabolic stress responses suggests that (1) feedback regulation of the HPA axis occurs across multiple bodily compartments, and (2) fat tissue integrates psychogenic stress signals. These studies support a link between stress biology and energy metabolism, a connection that has clear relevance for numerous disease states and their comorbidities.
Collapse
Affiliation(s)
- Annette D de Kloet
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL 32611, United States
| | - James P Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States.
| |
Collapse
|
87
|
Bartko J, Derhaschnig U, Neels T, Nabozny GH, Harcken C, Leuschner J, De Vries F, Jilma B. Selective glucocorticoid receptor modulation inhibits cytokine responses in a canine model of mild endotoxemia. Pharmacol Res 2017; 125:215-223. [PMID: 28923543 DOI: 10.1016/j.phrs.2017.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/07/2017] [Accepted: 09/12/2017] [Indexed: 12/24/2022]
Abstract
Selective glucocorticoid receptor modulators (GRMs) promise to reduce adverse events of glucocorticoids while maintaining anti-inflammatory potency. The present study tested the anti-inflammatory activity of two novel non-steroidal GRMs (GRM1: BI 607812 BS, GRM2: BI 653048 BS*H3PO4) in comparison to prednisolone in a canine model of low dose endotoxemia. This study compared the anti-inflammatory and pharmacokinetic profile of escalating daily oral doses of GRM1 (1, 2.5, 5 and 10mg/kg) and GRM2 (0.1, 0.25 and 1mg/kg) with prednisolone (0.25 and 0.5mg/kg) and placebo after intravenous infusion of endotoxin (0.1μg/kg) to Beagle dogs. This was followed by a 14-day evaluation study of safety and pharmacokinetics. Endotoxin challenge increased TNF-α ∼2000-fold and interleukin-6 (IL-6) 100-fold. Prednisolone and both GRMs suppressed peak TNF-α and IL-6 by 71-82% as compared with placebo. The highest doses of GRM1 and GRM2 reduced the mean body temperature increase by ∼30%. The endotoxin-induced rise in plasma cortisol was strongly suppressed in all treatment groups. Pharmacokinetics of both GRMs were non-linear. Adverse effects of endotoxemia such as vomiting were mitigated by GRM2 and prednisolone, indicating an antiemetic effect. During the 14-day treatment period, the adverse event profile of both GRMs appeared to be similar to prednisolone. Both GRMs had anti-inflammatory effects comparable to prednisolone and showed good safety profiles. Compounds targeting the glucocorticoid receptor selectively may provide an alternative to traditional glucocorticoids in the treatment of inflammatory disease.
Collapse
Affiliation(s)
- Johann Bartko
- Department of Clinical Pharmacology, Medical University of Vienna, Austria; Ludwig Boltzmann Institute of Osteology at the Hanusch Hospital of WGKK and AUVA Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria
| | - Ulla Derhaschnig
- Department of Clinical Pharmacology, Medical University of Vienna, Austria; Department of Emergency Medicine, Medical University of Vienna, Austria
| | - Tania Neels
- Department of Clinical Pharmacology, Medical University of Vienna, Austria
| | | | | | - Jost Leuschner
- LPT Laboratory of Pharmacology and Toxicology GmbH & Co. KG, Germany
| | | | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, Austria.
| |
Collapse
|
88
|
Decreased comfort food intake and allostatic load in adolescents carrying the A3669G variant of the glucocorticoid receptor gene. Appetite 2017; 116:21-28. [DOI: 10.1016/j.appet.2017.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 11/18/2022]
|
89
|
Sengupta N, Reardon DC, Gerard PD, Baldwin WS. Exchange of polar lipids from adults to neonates in Daphnia magna: Perturbations in sphingomyelin allocation by dietary lipids and environmental toxicants. PLoS One 2017; 12:e0178131. [PMID: 28542405 PMCID: PMC5443554 DOI: 10.1371/journal.pone.0178131] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/09/2017] [Indexed: 11/18/2022] Open
Abstract
Because xenosensing nuclear receptors are also lipid sensors that regulate lipid allocation, we hypothesized that toxicant-induced modulation of HR96 activity would alter lipid profiles and the balance between adult survival and neonate production following exposure in Daphnia magna. Adult daphnids were exposed to unsaturated fatty acid- and toxicant- activators or inhibitors of HR96 and later starved to test whether chemical exposure altered allocation toward survival or reproduction. The HR96 activators, linoleic acid and atrazine, decreased reproduction as expected with concomitant changes in the expression of HR96 regulated genes such as magro. The HR96 inhibitors, docosahexaenoic acid (DHA) and triclosan, increased reproduction or neonate starvation survival, respectively. However, pre-exposure to triclosan increased in neonate survival at the expense of reproductive maturation. Lipidomic analysis revealed that sphingomyelins (SM) are predominantly found in neonates and therefore we propose are important in development. DHA and triclosan increased neonatal SM, consistent with HR96’s regulation of Niemann-Pick genes. While DHA altered expression of magro, Niemann-Pick 1b, mannosidase, and other HR96-regulated genes as expected, triclosan primarily perturbed sphingomyelinase and mannosidase expression indicating different but potentially overlapping mechanisms for perturbing SM. Overall, SM appears to be a key lipid in Daphnia maturation and further support was provided by carmofur, which inhibits sphingomyelin/ceramide metabolism and in turn severely represses Daphnia maturation and initial brood production. In conclusion, toxicants can perturb lipid allocation and in turn impair development and reproduction.
Collapse
Affiliation(s)
- Namrata Sengupta
- Environmental Toxicology Program, Clemson University, Clemson, SC, United States of America
| | - Delaney C. Reardon
- Biological Sciences, Clemson University, Clemson, SC, United States of America
| | - Patrick D. Gerard
- Mathematical Sciences, Clemson University, Clemson, SC, United States of America
| | - William S. Baldwin
- Environmental Toxicology Program, Clemson University, Clemson, SC, United States of America
- Biological Sciences, Clemson University, Clemson, SC, United States of America
- * E-mail:
| |
Collapse
|
90
|
Chadwick JA, Hauck JS, Gomez-Sanchez CE, Gomez-Sanchez EP, Rafael-Fortney JA. Gene expression effects of glucocorticoid and mineralocorticoid receptor agonists and antagonists on normal human skeletal muscle. Physiol Genomics 2017; 49:277-286. [PMID: 28432191 DOI: 10.1152/physiolgenomics.00128.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/10/2017] [Accepted: 04/17/2017] [Indexed: 12/28/2022] Open
Abstract
Mineralocorticoid and glucocorticoid receptors are closely related steroid hormone receptors that regulate gene expression through many of the same hormone response elements. However, their transcriptional activities and effects in skeletal muscles are largely unknown. We recently identified mineralocorticoid receptors (MR) in skeletal muscles after finding that combined treatment with the angiotensin-converting enzyme inhibitor lisinopril and MR antagonist spironolactone was therapeutic in Duchenne muscular dystrophy mouse models. The glucocorticoid receptor (GR) agonist prednisolone is the current standard-of-care treatment for Duchenne muscular dystrophy because it prolongs ambulation, likely due to its anti-inflammatory effects. However, data on whether glucocorticoids have a beneficial or detrimental direct effect on skeletal muscle are controversial. Here, we begin to define the gene expression profiles in normal differentiated human skeletal muscle myotubes treated with MR and GR agonists and antagonists. The MR agonist aldosterone and GR agonist prednisolone had highly overlapping gene expression profiles, supporting the notion that prednisolone acts as both a GR and MR agonist that may have detrimental effects on skeletal muscles. Co-incubations with aldosterone plus either nonspecific or selective MR antagonists, spironolactone or eplerenone, resulted in similar numbers of gene expression changes, suggesting that both drugs can block MR activation to a similar extent. Eplerenone treatment alone decreased a number of important muscle-specific genes. This information may be used to develop biomarkers to monitor clinical efficacy of MR antagonists or GR agonists in muscular dystrophy, develop a temporally coordinated treatment with both drugs, or identify novel therapeutics with more specific downstream targets.
Collapse
Affiliation(s)
- Jessica A Chadwick
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - J Spencer Hauck
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Celso E Gomez-Sanchez
- Department of Internal Medicine, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Elise P Gomez-Sanchez
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jill A Rafael-Fortney
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio;
| |
Collapse
|
91
|
Hinds TD, Hosick PA, Chen S, Tukey RH, Hankins MW, Nestor-Kalinoski A, Stec DE. Mice with hyperbilirubinemia due to Gilbert's syndrome polymorphism are resistant to hepatic steatosis by decreased serine 73 phosphorylation of PPARα. Am J Physiol Endocrinol Metab 2017; 312:E244-E252. [PMID: 28096081 PMCID: PMC5406988 DOI: 10.1152/ajpendo.00396.2016] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/06/2017] [Accepted: 01/07/2017] [Indexed: 01/07/2023]
Abstract
Gilbert's syndrome in humans is derived from a polymorphism (TA repeat) in the hepatic UGT1A1 gene that results in decreased conjugation and increased levels of unconjugated bilirubin. Recently, we have shown that bilirubin binds directly to the fat-burning nuclear peroxisome proliferator-activated receptor-α (PPARα). Additionally, we have shown that serine 73 phosphorylation [Ser(P)73] of PPARα decreases activity by reducing its protein levels and transcriptional activity. The aim of this study was to determine whether humanized mice with the Gilbert's polymorphism (HuUGT*28) have increased PPARα activation and reduced hepatic fat accumulation. To determine whether humanized mice with Gilbert's mutation (HuUGT*28) have reduced hepatic lipids, we placed them and C57BL/6J control mice on a high-fat (60%) diet for 36 wk. Body weights, fat and lean mass, and fasting blood glucose and insulin levels were measured every 6 wk throughout the investigation. At the end of the study, hepatic lipid content was measured and PPARα regulated genes as well as immunostaining of Ser(P)73 PPARα from liver sections. The HuUGT*28 mice had increased serum bilirubin, lean body mass, decreased fat mass, and hepatic lipid content as well as lower serum glucose and insulin levels. Also, the HuUGT*28 mice had reduced Ser(P)73 PPARα immunostaining in livers and increased PPARα transcriptional activity compared with controls. A chronic but mild endogenous increase in unconjugated hyperbiliubinemia protects against hepatic steatosis through a reduction in Ser(P)73 PPARα, causing an increase in PPARα transcriptional activity.
Collapse
Affiliation(s)
- Terry D Hinds
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio
| | - Peter A Hosick
- Department of Exercise Science and Physical Education, Montclair State University, Montclair, New Jersey
| | - Shujuan Chen
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California; and
| | - Robert H Tukey
- Laboratory of Environmental Toxicology, Department of Pharmacology, University of California San Diego, La Jolla, California; and
| | - Michael W Hankins
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi
| | - Andrea Nestor-Kalinoski
- Advanced Microscopy and Imaging Center, Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - David E Stec
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi;
| |
Collapse
|
92
|
Khudyakov JI, Champagne CD, Meneghetti LM, Crocker DE. Blubber transcriptome response to acute stress axis activation involves transient changes in adipogenesis and lipolysis in a fasting-adapted marine mammal. Sci Rep 2017; 7:42110. [PMID: 28186107 PMCID: PMC5301240 DOI: 10.1038/srep42110] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/05/2017] [Indexed: 01/04/2023] Open
Abstract
Stress can compromise an animal’s ability to conserve metabolic stores and participate in energy-demanding activities that are critical for fitness. Understanding how wild animals, especially those already experiencing physiological extremes (e.g. fasting), regulate stress responses is critical for evaluating the impacts of anthropogenic disturbance on physiology and fitness, key challenges for conservation. However, studies of stress in wildlife are often limited to baseline endocrine measurements and few have investigated stress effects in fasting-adapted species. We examined downstream molecular consequences of hypothalamic-pituitary-adrenal (HPA) axis activation by exogenous adrenocorticotropic hormone (ACTH) in blubber of northern elephant seals due to the ease of blubber sampling and its key role in metabolic regulation in marine mammals. We report the first phocid blubber transcriptome produced by RNAseq, containing over 140,000 annotated transcripts, including metabolic and adipocytokine genes of interest. The acute response of blubber to stress axis activation, measured 2 hours after ACTH administration, involved highly specific, transient (lasting <24 hours) induction of gene networks that promote lipolysis and adipogenesis in mammalian adipocytes. Differentially expressed genes included key adipogenesis factors which can be used as blubber-specific markers of acute stress in marine mammals of concern for which sampling of other tissues is not possible.
Collapse
Affiliation(s)
- J I Khudyakov
- Department of Biological Sciences, University of the Pacific, 3601 Pacific Avenue, Stockton, CA 95211, USA.,National Marine Mammal Foundation, 2240 Shelter Island Drive Suite 200, San Diego, CA 92106, USA
| | - C D Champagne
- National Marine Mammal Foundation, 2240 Shelter Island Drive Suite 200, San Diego, CA 92106, USA.,Department of Biological Sciences, Old Dominion University, 1 Monarch Way, Norfolk, VA 23529, USA
| | - L M Meneghetti
- Department of Biology, Sonoma State University, 1801 E Cotati Avenue, Rohnert Park, CA 94928, USA
| | - D E Crocker
- Department of Biology, Sonoma State University, 1801 E Cotati Avenue, Rohnert Park, CA 94928, USA
| |
Collapse
|
93
|
Bose SK, Hutson I, Harris CA. Hepatic Glucocorticoid Receptor Plays a Greater Role Than Adipose GR in Metabolic Syndrome Despite Renal Compensation. Endocrinology 2016; 157:4943-4960. [PMID: 27754788 PMCID: PMC5133352 DOI: 10.1210/en.2016-1615] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Exogenous glucocorticoid administration results in hyperglycemia, insulin resistance, hepatic dyslipidemia, and hypertension, a constellation of findings known as Cushing's syndrome. These effects are mediated by the glucocorticoid receptor (GR). Because GR activation in liver and adipose has been implicated in metabolic syndrome (MS), we wanted to determine the role of GR in these tissues in the development of MS. Because GR knockout (KO) mice (whole-body KO) exhibit perinatal lethality due to respiratory failure, we generated tissue-specific (liver or adipose) GRKO mice using cre-lox technology. Real-time PCR analysis of liver mRNA from dexamethasone-treated wildtype (WT) and liver GRKO mice indicated that hepatic GR regulates the expression of key genes involved in gluconeogenesis and glycogen metabolism. Interestingly, we have observed that liver-specific deletion of GR resulted in a significant increase in mRNA expression of key genes involved in gluconeogenesis and glycogen metabolism in kidney tissue, indicating a compensatory mechanism to maintain glucose homeostasis. We have also observed that GR plays an important role in regulating the mRNA expression of key genes involved in lipid metabolism. Liver GRKO mice demonstrated decreased fat mass and liver glycogen content compared with WT mice administered dexamethasone for 2 weeks. Adipose-specific deletion of GR did not alter glucose tolerance or insulin sensitivity of adipose GRKO mice compared with WT mice administrated dexamethasone. This indicates that liver GR might be more important in development of MS in dexamethasone-treated mice, whereas adipose GR plays a little role in these paradigms.
Collapse
Affiliation(s)
- Sandip K Bose
- Department of Internal Medicine (S.K.B., I.H., C.A.H.), Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri 63110; and John Cochran Division (C.A.H.), Veterans Affairs St. Louis Health Care System, St. Louis, Missouri 63106
| | - Irina Hutson
- Department of Internal Medicine (S.K.B., I.H., C.A.H.), Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri 63110; and John Cochran Division (C.A.H.), Veterans Affairs St. Louis Health Care System, St. Louis, Missouri 63106
| | - Charles A Harris
- Department of Internal Medicine (S.K.B., I.H., C.A.H.), Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, Missouri 63110; and John Cochran Division (C.A.H.), Veterans Affairs St. Louis Health Care System, St. Louis, Missouri 63106
| |
Collapse
|
94
|
Rodriguez JM, Monsalves-Alvarez M, Henriquez S, Llanos MN, Troncoso R. Glucocorticoid resistance in chronic diseases. Steroids 2016; 115:182-192. [PMID: 27643454 DOI: 10.1016/j.steroids.2016.09.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 12/25/2022]
Abstract
Glucocorticoids are involved in several responses triggered by a variety of environmental and physiological stimuli. These hormones have a wide-range of regulatory effects in organisms. Synthetic glucocorticoids are extensively used to suppress allergic, inflammatory, and immune disorders. Although glucocorticoids are highly effective for therapeutic purposes, some patients chronically treated with glucocorticoids can develop reduced glucocorticoid sensitivity or even resistance, increasing patient vulnerability to exaggerated inflammatory responses. Glucocorticoid resistance can occur in several chronic diseases, including asthma, major depression, and cardiovascular conditions. In this review, we discuss the complexity of the glucocorticoid receptor and the potential role of glucocorticoid resistance in the development of chronic diseases.
Collapse
Affiliation(s)
- Juan M Rodriguez
- Institute of Nutrition and Food Technology, University of Chile, Santiago 7830490, Chile
| | - Matías Monsalves-Alvarez
- Institute of Nutrition and Food Technology, University of Chile, Santiago 7830490, Chile; Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas, Universidad Andres Bello, Santiago, Chile
| | - Sandra Henriquez
- Institute of Nutrition and Food Technology, University of Chile, Santiago 7830490, Chile
| | - Miguel N Llanos
- Institute of Nutrition and Food Technology, University of Chile, Santiago 7830490, Chile
| | - Rodrigo Troncoso
- Institute of Nutrition and Food Technology, University of Chile, Santiago 7830490, Chile; Advanced Center for Chronic Disease, Faculty of Chemistry and Pharmacy, University of Chile, Santiago 8380492, Chile.
| |
Collapse
|
95
|
Marino JS, Stechschulte LA, Stec DE, Nestor-Kalinoski A, Coleman S, Hinds TD. Glucocorticoid Receptor β Induces Hepatic Steatosis by Augmenting Inflammation and Inhibition of the Peroxisome Proliferator-activated Receptor (PPAR) α. J Biol Chem 2016; 291:25776-25788. [PMID: 27784782 DOI: 10.1074/jbc.m116.752311] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/11/2016] [Indexed: 01/01/2023] Open
Abstract
Glucocorticoids (GCs) regulate energy supply in response to stress by increasing hepatic gluconeogenesis during fasting. Long-term GC treatment induces hepatic steatosis and weight gain. GC signaling is coordinated via the GC receptor (GR) GRα, as the GRβ isoform lacks a ligand-binding domain. The roles of the GR isoforms in the regulation of lipid accumulation is unknown. The purpose of this study was to determine whether GRβ inhibits the actions of GCs in the liver, or enhances hepatic lipid accumulation. We show that GRβ expression is increased in adipose and liver tissues in obese high-fat fed mice. Adenovirus-mediated delivery of hepatic GRβ overexpression (GRβ-Ad) resulted in suppression of gluconeogenic genes and hyperglycemia in mice on a regular diet. Furthermore, GRβ-Ad mice had increased hepatic lipid accumulation and serum triglyceride levels possibly due to the activation of NF-κB signaling and increased tumor necrosis factor α (TNFα) and inducible nitric-oxide synthase expression, indicative of enhanced M1 macrophages and the development of steatosis. Consequently, GRβ-Ad mice had increased glycogen synthase kinase 3β (GSK3β) activity and reduced hepatic PPARα and fibroblast growth factor 21 (FGF21) expression and lower serum FGF21 levels, which are two proteins known to increase during fasting to enhance the burning of fat by activating the β-oxidation pathway. In conclusion, GRβ antagonizes the GC-induced signaling during fasting via GRα and the PPARα-FGF21 axis that reduces fat burning. Furthermore, hepatic GRβ increases inflammation, which leads to hepatic lipid accumulation.
Collapse
Affiliation(s)
- Joseph S Marino
- From the Department of Kinesiology, Laboratory of Systems Physiology, University of North Carolina, Charlotte, North Carolina 28223
| | | | - David E Stec
- the Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular-Renal Research Center, University of Mississippi Medical Center, Jackson, Mississippi 39216, and
| | | | - Sydni Coleman
- the University of Cincinnati, College of Medicine, Cincinnati, Ohio 45220
| | - Terry D Hinds
- Center for Hypertension and Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine, Toledo, Ohio 43614,
| |
Collapse
|
96
|
Hinds TD, Burns KA, Hosick PA, McBeth L, Nestor-Kalinoski A, Drummond HA, AlAmodi AA, Hankins MW, Vanden Heuvel JP, Stec DE. Biliverdin Reductase A Attenuates Hepatic Steatosis by Inhibition of Glycogen Synthase Kinase (GSK) 3β Phosphorylation of Serine 73 of Peroxisome Proliferator-activated Receptor (PPAR) α. J Biol Chem 2016; 291:25179-25191. [PMID: 27738106 DOI: 10.1074/jbc.m116.731703] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/30/2016] [Indexed: 01/21/2023] Open
Abstract
Non-alcoholic fatty liver disease is the most rapidly growing form of liver disease and if left untreated can result in non-alcoholic steatohepatitis, ultimately resulting in liver cirrhosis and failure. Biliverdin reductase A (BVRA) is a multifunctioning protein primarily responsible for the reduction of biliverdin to bilirubin. Also, BVRA functions as a kinase and transcription factor, regulating several cellular functions. We report here that liver BVRA protects against hepatic steatosis by inhibiting glycogen synthase kinase 3β (GSK3β) by enhancing serine 9 phosphorylation, which inhibits its activity. We show that GSK3β phosphorylates serine 73 (Ser(P)73) of the peroxisome proliferator-activated receptor α (PPARα), which in turn increased ubiquitination and protein turnover, as well as decreased activity. Interestingly, liver-specific BVRA KO mice had increased GSK3β activity and Ser(P)73 of PPARα, which resulted in decreased PPARα protein and activity. Furthermore, the liver-specific BVRA KO mice exhibited increased plasma glucose and insulin levels and decreased glycogen storage, which may be due to the manifestation of hepatic steatosis observed in the mice. These findings reveal a novel BVRA-GSKβ-PPARα axis that regulates hepatic lipid metabolism and may provide unique targets for the treatment of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Terry D Hinds
- the Center for Hypertension and Personalized Medicine, Department of Physiology & Pharmacology,
| | - Katherine A Burns
- the Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania 16802, and.,the Department of Environmental Health, Division of Environmental Genetics and Molecular Toxicology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Peter A Hosick
- From the Department of Physiology & Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi 39216.,the Department of Exercise Science and Physical Education, Montclair State University, Montclair, New Jersey 07043
| | - Lucien McBeth
- the Center for Hypertension and Personalized Medicine, Department of Physiology & Pharmacology
| | - Andrea Nestor-Kalinoski
- Advanced Microscopy & Imaging Center, Department of Surgery, University of Toledo College of Medicine and Life Sciences, Toledo Ohio 43614
| | - Heather A Drummond
- From the Department of Physiology & Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Abdulhadi A AlAmodi
- From the Department of Physiology & Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Michael W Hankins
- From the Department of Physiology & Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - John P Vanden Heuvel
- the Department of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania 16802, and
| | - David E Stec
- From the Department of Physiology & Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, Mississippi 39216,
| |
Collapse
|
97
|
Multiple Sclerosis and Obesity: Possible Roles of Adipokines. Mediators Inflamm 2016; 2016:4036232. [PMID: 27721574 PMCID: PMC5046034 DOI: 10.1155/2016/4036232] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/22/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022] Open
Abstract
Multiple Sclerosis (MS) is an autoimmune disorder of the Central Nervous System that has been associated with several environmental factors, such as diet and obesity. The possible link between MS and obesity has become more interesting in recent years since the discovery of the remarkable properties of adipose tissue. Once MS is initiated, obesity can contribute to increased disease severity by negatively influencing disease progress and treatment response, but, also, obesity in early life is highly relevant as a susceptibility factor and causally related risk for late MS development. The aim of this review was to discuss recent evidence about the link between obesity, as a chronic inflammatory state, and the pathogenesis of MS as a chronic autoimmune and inflammatory disease. First, we describe the main cells involved in MS pathogenesis, both from neural tissue and from the immune system, and including a new participant, the adipocyte, focusing on their roles in MS. Second, we concentrate on the role of several adipokines that are able to participate in the mediation of the immune response in MS and on the possible cross talk between the latter. Finally, we explore recent therapy that involves the transplantation of adipocyte precursor cells for the treatment of MS.
Collapse
|
98
|
Conceição EPS, Moura EG, Manhães AC, Carvalho JC, Nobre JL, Oliveira E, Lisboa PC. Calcium reduces vitamin D and glucocorticoid receptors in the visceral fat of obese male rats. J Endocrinol 2016; 230:263-74. [PMID: 27325245 DOI: 10.1530/joe-16-0041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/20/2016] [Indexed: 01/04/2023]
Abstract
Rats overfed during lactation show higher visceral adipose tissue (VAT) mass and metabolic dysfunctions at adulthood. As both vitamin D and glucocorticoids change adipogenesis, parameters related to metabolism and action of these hormones in the adipocyte can be altered in rats raised in small litters (SL). We also studied the antiobesity effects of high calcium diet since it decreases visceral fat in obesity models. On postnatal day (PN) 3, litter size was adjusted to 3pups/dam (SL) to induce overfeeding. Control litters (NL) remained with 10pups/dam until weaning. From PN120 to PN180, half of the SL rats were fed standard chow (SL) and the other half was fed a calcium-supplemented chow (SL-Ca, 10g CaCO3/kg). Both SL groups were heavier and hyperphagic when compared with the NL group; however, SL-Ca rats ate less than SL. SL-Ca rats had decreased VAT mass and adipocyte size, associated with lower hypothalamic NPY content, VAT fat acid synthase content and leptinemia. At PN120, SL rats had increased plasma 25(OH)D3, Cyp27b1 mRNA and glucocorticoid receptor (GR-α) in the VAT, but lower vitamin D receptor (Vdr) mRNA. At PN180, Cyp27b1 and GR-α remained higher, while Vdr normalized in SL rats. SL-Ca rats had normal VAT Cyp27b1 and GR-α, but lower Vdr Thus, higher body mass and glucocorticoid receptors in the VAT of SL rats are normalized by calcium-enriched diet, and Vdr expression in this tissue is reduced, suggesting a possible role of glucocorticoids and vitamin D in calcium action in the adipocyte.
Collapse
Affiliation(s)
- E P S Conceição
- Laboratory of Endocrine PhysiologyDepartment of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - E G Moura
- Laboratory of Endocrine PhysiologyDepartment of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - A C Manhães
- Laboratory of NeurophysiologyDepartment of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J C Carvalho
- Laboratory of Endocrine PhysiologyDepartment of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - J L Nobre
- Laboratory of Endocrine PhysiologyDepartment of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - E Oliveira
- Laboratory of Endocrine PhysiologyDepartment of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - P C Lisboa
- Laboratory of Endocrine PhysiologyDepartment of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
99
|
Timcodar (VX-853) Is a Non-FKBP12 Binding Macrolide Derivative That Inhibits PPARγ and Suppresses Adipogenesis. PPAR Res 2016; 2016:6218637. [PMID: 27190501 PMCID: PMC4848453 DOI: 10.1155/2016/6218637] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/27/2016] [Indexed: 11/17/2022] Open
Abstract
Nutrient overload and genetic factors have led to a worldwide epidemic of obesity that is the underlying cause of diabetes, atherosclerosis, and cardiovascular disease. In this study, we used macrolide drugs such as FK506, rapamycin, and macrolide derived, timcodar (VX-853), to determine their effects on lipid accumulation during adipogenesis. Rapamycin and FK506 bind to FK506-binding proteins (FKBPs), such as FKBP12, which causes suppression of the immune system and inhibition of mTOR. Rapamycin has been previously reported to inhibit the adipogenic process and lipid accumulation. However, rapamycin treatment in rodents caused immune suppression and glucose resistance, even though the mice lost weight. Here we show that timcodar (1 μM), a non-FKBP12-binding drug, significantly (p < 0.001) inhibited lipid accumulation during adipogenesis. A comparison of the same concentration of timcodar (1 μM) and rapamycin (1 μM) showed that both are inhibitors of lipid accumulation during adipogenesis. Importantly, timcodar potently (p < 0.01) suppressed transcriptional regulators of adipogenesis, PPARγ and C/EBPα, resulting in the inhibition of genes involved in lipid accumulation. These studies set the stage for timcodar as a possible antiobesity therapy, which is rapidly emerging as a pandemic.
Collapse
|
100
|
Stec DE, John K, Trabbic CJ, Luniwal A, Hankins MW, Baum J, Hinds TD. Bilirubin Binding to PPARα Inhibits Lipid Accumulation. PLoS One 2016; 11:e0153427. [PMID: 27071062 PMCID: PMC4829185 DOI: 10.1371/journal.pone.0153427] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/29/2016] [Indexed: 01/31/2023] Open
Abstract
Numerous clinical and population studies have demonstrated that increased serum bilirubin levels protect against cardiovascular and metabolic diseases such as obesity and diabetes. Bilirubin is a potent antioxidant, and the beneficial actions of moderate increases in plasma bilirubin have been thought to be due to the antioxidant effects of this bile pigment. In the present study, we found that bilirubin has a new function as a ligand for PPARα. We show that bilirubin can bind directly to PPARα and increase transcriptional activity. When we compared biliverdin, the precursor to bilirubin, on PPARα transcriptional activation to known PPARα ligands, WY 14,643 and fenofibrate, it showed that fenofibrate and biliverdin have similar activation properties. Treatment of 3T3-L1 adipocytes with biliverdin suppressed lipid accumulation and upregulated PPARα target genes. We treated wild-type and PPARα KO mice on a high fat diet with fenofibrate or bilirubin for seven days and found that both signal through PPARα dependent mechanisms. Furthermore, the effect of bilirubin on lowering glucose and reducing body fat percentage was blunted in PPARα KO mice. These data demonstrate a new function for bilirubin as an agonist of PPARα, which mediates the protection from adiposity afforded by moderate increases in bilirubin.
Collapse
Affiliation(s)
- David E. Stec
- Cardiovascular-Renal Research Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State St, Jackson, Mississippi, 39216, United States of America
| | - Kezia John
- Center for Hypertension and Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, United States of America
| | - Christopher J. Trabbic
- Center for Drug Design and Development, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, 43614, United States of America
| | - Amarjit Luniwal
- Center for Drug Design and Development, University of Toledo College of Pharmacy and Pharmaceutical Sciences, Toledo, OH, 43614, United States of America
- North American Science Associates, Inc. (NAMSA), 6750 Wales Rd, Northwood, Ohio, 43619, United States of America
| | - Michael W. Hankins
- Cardiovascular-Renal Research Center, Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State St, Jackson, Mississippi, 39216, United States of America
| | - Justin Baum
- Center for Hypertension and Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, United States of America
| | - Terry D. Hinds
- Center for Hypertension and Personalized Medicine, Department of Physiology & Pharmacology, University of Toledo College of Medicine, Toledo, OH, 43614, United States of America
- * E-mail:
| |
Collapse
|