1
|
Li S, Zhang S, Rensen PCN, Meijer OC, Kooijman S, Kroon J. Out-of-phase treatment with the synthetic glucocorticoid betamethasone disturbs glucose metabolism in mice. Life Sci 2024; 357:123080. [PMID: 39332486 DOI: 10.1016/j.lfs.2024.123080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVE Endogenous glucocorticoid levels display a strong circadian rhythm, which is often not considered when synthetic glucocorticoids are prescribed as anti-inflammatory drugs. In this study we evaluated the effect timing of glucocorticoid administration, i.e. in-phase (administered when endogenous glucocorticoid levels are high) versus out-of-phase (administered when endogenous glucocorticoid levels are low). We investigated the synthetic glucocorticoid betamethasone - which is extensively used in the clinic - and monitored the development of common metabolic side effects in mice upon prolonged treatment, with a particular focus on glucose metabolism. METHODS Male and female C57BL/6J mice were treated with the synthetic glucocorticoid betamethasone in-phase and out-of-phase, and the development of metabolic side effects was monitored. RESULTS We observed that, compared with in-phase treatment, out-of-phase treatment with betamethasone results in hyperinsulinemia in both male and female C57BL/6J mice. We additionally found that out-of-phase betamethasone treatment strongly reduced insulin sensitivity as compared to in-phase administration during morning measurements. Our study shows that the adverse effects of betamethasone are dependent on the time of treatment with generally less side effects on glucose metabolism with in-phase treatment. CONCLUSIONS This study highlights differences in glucocorticoid outcome based on the time of measurement, advocating that potential circadian variation should be taken into account when studying glucocorticoid biology.
Collapse
Affiliation(s)
- Sheng Li
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sen Zhang
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan Kroon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
2
|
Zhou L, Pereiro MT, Li Y, Derigs M, Kuenne C, Hielscher T, Huang W, Kränzlin B, Tian G, Kobayashi K, Lu GHN, Roedl K, Schmidt C, Günther S, Looso M, Huber J, Xu Y, Wiech T, Sperhake JP, Wichmann D, Gröne HJ, Worzfeld T. Glucocorticoids induce a maladaptive epithelial stress response to aggravate acute kidney injury. Sci Transl Med 2024; 16:eadk5005. [PMID: 39356748 DOI: 10.1126/scitranslmed.adk5005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 05/26/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Acute kidney injury (AKI) is a frequent and challenging clinical condition associated with high morbidity and mortality and represents a common complication in critically ill patients with COVID-19. In AKI, renal tubular epithelial cells (TECs) are a primary site of damage, and recovery from AKI depends on TEC plasticity. However, the molecular mechanisms underlying adaptation and maladaptation of TECs in AKI remain largely unclear. Here, our study of an autopsy cohort of patients with COVID-19 provided evidence that injury of TECs by myoglobin, released as a consequence of rhabdomyolysis, is a major pathophysiological mechanism for AKI in severe COVID-19. Analyses of human kidney biopsies, mouse models of myoglobinuric and gentamicin-induced AKI, and mouse kidney tubuloids showed that TEC injury resulted in activation of the glucocorticoid receptor by endogenous glucocorticoids, which aggravated tubular damage. The detrimental effect of endogenous glucocorticoids on injured TECs was exacerbated by the administration of a widely clinically used synthetic glucocorticoid, dexamethasone, as indicated by experiments in mouse models of myoglobinuric- and folic acid-induced AKI, human and mouse kidney tubuloids, and human kidney slice cultures. Mechanistically, studies in mouse models of AKI, mouse tubuloids, and human kidney slice cultures demonstrated that glucocorticoid receptor signaling in injured TECs orchestrated a maladaptive transcriptional program to hinder DNA repair, amplify injury-induced DNA double-strand break formation, and dampen mTOR activity and mitochondrial bioenergetics. This study identifies glucocorticoid receptor activation as a mechanism of epithelial maladaptation, which is functionally important for AKI.
Collapse
Affiliation(s)
- Luping Zhou
- Institute of Pharmacology, University of Marburg, Karl-von-Frisch-Straße 2, Marburg 35043, Germany
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Taiping Street 25, Luzhou 646000, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Taiping Street 25, Luzhou 646000, China
| | - Marc Torres Pereiro
- Institute of Pharmacology, University of Marburg, Karl-von-Frisch-Straße 2, Marburg 35043, Germany
| | - Yanqun Li
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Taiping Street 25, Luzhou 646000, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Taiping Street 25, Luzhou 646000, China
| | - Marcus Derigs
- Department of Urology, University Hospital, University of Marburg, Baldingerstraße, Marburg 35043, Germany
| | - Carsten Kuenne
- Bioinformatics, Max Planck Institute for Heart and Lung Research, Ludwigstraße 43, Bad Nauheim 61231, Germany
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Wei Huang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Taiping Street 25, Luzhou 646000, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Taiping Street 25, Luzhou 646000, China
| | - Bettina Kränzlin
- Core Facility Preclinical Models, Medical Faculty Mannheim, University of Heidelberg, Ludolf-Krehl-Straße 13-17, Mannheim 68167, Germany
| | - Gang Tian
- Department of Laboratory Medicine, Affiliated Hospital of Southwest Medical University, Taiping Street 25, Luzhou 646000, China
| | - Kazuhiro Kobayashi
- Institute of Pharmacology, University of Marburg, Karl-von-Frisch-Straße 2, Marburg 35043, Germany
| | - Gia-Hue Natalie Lu
- Institute of Pharmacology, University of Marburg, Karl-von-Frisch-Straße 2, Marburg 35043, Germany
| | - Kevin Roedl
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Claudia Schmidt
- Light Microscopy Facility, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Stefan Günther
- Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Ludwigstraße 43, Bad Nauheim 61231, Germany
| | - Mario Looso
- Bioinformatics, Max Planck Institute for Heart and Lung Research, Ludwigstraße 43, Bad Nauheim 61231, Germany
| | - Johannes Huber
- Department of Urology, University Hospital, University of Marburg, Baldingerstraße, Marburg 35043, Germany
| | - Yong Xu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Taiping Street 25, Luzhou 646000, China
- Metabolic Vascular Diseases Key Laboratory of Sichuan Province, Taiping Street 25, Luzhou 646000, China
| | - Thorsten Wiech
- Institute of Pathology, Nephropathology Section, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Hamburg 20246, Germany
| | - Jan-Peter Sperhake
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Butenfeld 34, Hamburg 22529, Germany
| | - Dominic Wichmann
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, Hamburg 20246, Germany
| | - Hermann-Josef Gröne
- Institute of Pharmacology, University of Marburg, Karl-von-Frisch-Straße 2, Marburg 35043, Germany
- Medical Faculty, University of Heidelberg, Heidelberg 69120, Germany
| | - Thomas Worzfeld
- Institute of Pharmacology, University of Marburg, Karl-von-Frisch-Straße 2, Marburg 35043, Germany
| |
Collapse
|
3
|
Ngema M, Xulu ND, Ngubane PS, Khathi A. Pregestational Prediabetes Induces Maternal Hypothalamic-Pituitary-Adrenal (HPA) Axis Dysregulation and Results in Adverse Foetal Outcomes. Int J Mol Sci 2024; 25:5431. [PMID: 38791468 PMCID: PMC11122116 DOI: 10.3390/ijms25105431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Maternal type 2 diabetes mellitus (T2DM) has been shown to result in foetal programming of the hypothalamic-pituitary-adrenal (HPA) axis, leading to adverse foetal outcomes. T2DM is preceded by prediabetes and shares similar pathophysiological complications. However, no studies have investigated the effects of maternal prediabetes on foetal HPA axis function and postnatal offspring development. Hence, this study investigated the effects of pregestational prediabetes on maternal HPA axis function and postnatal offspring development. Pre-diabetic (PD) and non-pre-diabetic (NPD) female Sprague Dawley rats were mated with non-prediabetic males. After gestation, male pups born from the PD and NPD groups were collected. Markers of HPA axis function, adrenocorticotropin hormone (ACTH) and corticosterone, were measured in all dams and pups. Glucose tolerance, insulin and gene expressions of mineralocorticoid (MR) and glucocorticoid (GR) receptors were further measured in all pups at birth and their developmental milestones. The results demonstrated increased basal concentrations of ACTH and corticosterone in the dams from the PD group by comparison to NPD. Furthermore, the results show an increase basal ACTH and corticosterone concentrations, disturbed MR and GR gene expression, glucose intolerance and insulin resistance assessed via the Homeostasis Model Assessment (HOMA) indices in the pups born from the PD group compared to NPD group at all developmental milestones. These observations reveal that pregestational prediabetes is associated with maternal dysregulation of the HPA axis, impacting offspring HPA axis development along with impaired glucose handling.
Collapse
Affiliation(s)
| | | | | | - Andile Khathi
- School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Westville, Private Bag X54001, Durban 4041, KwaZulu Natal, South Africa; (M.N.); (N.D.X.); (P.S.N.)
| |
Collapse
|
4
|
Ignatz EH, Rise ML, Gamperl AK. Impact of stress phenotype, elevated temperature, and bacterin exposure on male Atlantic salmon ( Salmo salar) growth, stress, and immune biomarker gene expression. Physiol Genomics 2023; 55:587-605. [PMID: 37746713 DOI: 10.1152/physiolgenomics.00055.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/30/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023] Open
Abstract
In this study, postsmolt male Atlantic salmon, previously identified as low responders (LRs) or high responders (HRs) based on poststress cortisol levels, had their head kidney and liver sampled at 12°C and 20°C before injection (time 0) and after injection (i.e., at 12- and 24-h postinjection, respectively) with either Forte Micro (a multivalent vaccine containing bacterin, to capture peak antibacterial responses) or an equal volume of PBS. Quantitative real-time PCR (qPCR) was then used to measure the expression of 15 biomarker genes in the head kidney and 12 genes in the liver at each temperature/sampling point. Target transcripts were chosen that were related to growth, stress, and innate antibacterial immune responses. Many temperature, phenotype, and injection effects were found for individual genes within these three broad categories, and multivariate statistical analyses (i.e., principal component analysis and permutational multivariate analysis of variance) were used to look for overall patterns in transcript expression. These analyses revealed that HR salmon at 20°C mounted a more robust response (P < 0.05) for the 10 head kidney immune-related transcripts when injected with Forte Micro than LR salmon. In contrast, the seven liver stress-related transcripts displayed a greater response (P = 0.057) in LR versus HR fish with Forte Micro at 12°C. Overall, although this research did find some differences between LR and HR fish, it does not provide strong (conclusive) evidence that the selection of a particular phenotype would have major implications for the health of salmon over the temperature range examined.NEW & NOTEWORTHY This is the first paper to describe the impact of both temperature and bacterial stimulation on head kidney and liver transcript expression in Atlantic salmon characterized as LRs versus HRs. Notably, we found that HR salmon at 20°C mounted a more robust innate antibacterial immune response than LR salmon. In addition, LR fish at 12°C may (P = 0.057) exhibit higher expression of stress-related transcripts in response to vaccine injection relative to HR fish.
Collapse
Affiliation(s)
- Eric H Ignatz
- Department of Ocean Sciences, Memorial University, St. John's, Newfoundland and Labrador, Canada
| | - Matthew L Rise
- Department of Ocean Sciences, Memorial University, St. John's, Newfoundland and Labrador, Canada
| | - A Kurt Gamperl
- Department of Ocean Sciences, Memorial University, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
5
|
Paes-Leme B, Monteiro LDRN, Gholami K, Hoe SZ, Ferguson AV, Murphy D, Antunes-Rodrigues J, Rorato R, Reis LC, Mecawi AS. Fasting increases circulating angiotensin levels and brain Agtr1a expression in male rats. J Neuroendocrinol 2023; 35:e13334. [PMID: 37667574 DOI: 10.1111/jne.13334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 09/06/2023]
Abstract
In addition to being recognised for involvement in cardiovascular control and hydromineral balance, the renin-angiotensin system (RAS) has also been associated with the neuroendocrine control of energy balance. One of the main brain sites for angiotensin II (ANG II)/type 1 receptor (AT1 R) signalling is the subfornical organ (SFO), a circumventricular organ related to the control of autonomic functions, motivated behaviours and energy metabolism. Thus, we hypothesised that circulating ANG II may act on the SFO AT1 R receptors to integrate metabolic and hydromineral balance. We evaluated whether food deprivation can modulate systemic RAS activity and Agrt1a brain expression, and if ANG II/AT1 R signalling influences the hypothalamic expression of mRNAs encoding neuropeptides and food and water ingestion in fed and fasted Wistar rats. We found a significant increase in both ANG I and ANG II plasma levels after 24 and 48 h of fasting. Expression of Agrt1a mRNA in the SFO and paraventricular nucleus (PVN) also increased after food deprivation for 48 h. Treatment of fasted rats with low doses of losartan in drinking water attenuated the decrease in glycemia and meal-associated water intake without changing the expression in PVN or arcuate nucleus of mRNAs encoding selected neuropeptides related to energy homeostasis control. These findings point to a possible role of peripheral ANG II/SFO-AT1 R signalling in the control of refeeding-induced thirst. On the other hand, intracerebroventricular losartan treatment decreased food and water intake over dark time in fed but not in fasted rats.
Collapse
Affiliation(s)
- Bruno Paes-Leme
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Lívia da Rocha Natalino Monteiro
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - Khadijeh Gholami
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - See Ziau Hoe
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Alastair Victor Ferguson
- Department of Biomedical and Molecular Sciences and Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - David Murphy
- Molecular Neuroendocrinology Research Group, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - José Antunes-Rodrigues
- Department of Physiology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rodrigo Rorato
- Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Luis Carlos Reis
- Department of Physiological Sciences, Institute of Biological and Health Sciences, Federal Rural University of Rio de Janeiro, Seropédica, Rio de Janeiro, Brazil
| | - André Souza Mecawi
- Department of Biophysics, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Toledo-Corral CM, Ding L, Morales JC, Chapman TM, Romero MB, Weigensberg MJ. Morning Serum Cortisol Is Uniquely Associated with Cardiometabolic Risk Independent of Body Composition in Latino Adolescents. Metab Syndr Relat Disord 2023; 21:214-221. [PMID: 37042653 PMCID: PMC10181798 DOI: 10.1089/met.2022.0091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2023] Open
Abstract
Background: Alterations in morning serum cortisol (MSC) have been associated with higher cardiometabolic risk. This finding has been documented primarily in populations with overweight or obesity; however, it has not been clearly established if obesity plays a requisite role in this relationship. This study seeks to extend earlier findings by examining whether body composition measures alter the relationship between MSC with glucose and insulin markers, blood pressure, and lipid parameters in Latino youth in middle adolescence. Methods: This cross-sectional study included 196 healthy adolescents (130F/66M; mean age: 16.4 ± 0.6 years; 95% Latino; mean body mass index, BMI: 24.3 ± 5.7) from Los Angeles, California. Morning cortisol, glucose, insulin, glycated hemoglobin, and lipids (triglycerides and high-density lipoprotein cholesterol) were assessed from a fasting blood sample. Sitting systolic and diastolic blood pressure was averaged from duplicate measures. Body composition measures included BMI and waist circumference, which were used as proxies for total body and abdominal adiposity, respectively. Triplicate measurements of weight and height were averaged for calculation of BMI; age- and sex-specific BMI z-score was used to classify into normal BMI or overweight/obese BMI status. Waist circumference was measured in duplicate and the average was used to classify participants into two strata: normal/healthy waist circumference (<90th percentile for age, sex, and ethnicity) and high waist circumference (≥90th percentile). Results: The primary findings were that higher MSC was associated with higher fasting glucose and systolic blood pressure after adjusting for age, sex, and BMI z-score (and/or waist circumference). BMI status or waist circumference status did not alter these relationships. Main Conclusion: Our results suggest that the relationships between hypothalamic-pituitary-adrenal axis function and certain cardiometabolic risk factors may be independent of adiposity. Future research is warranted to discover the contributors and underlying mechanisms of these relationships in adolescent populations. ClinicalTrials.gov identifier: NCT02088294.
Collapse
Affiliation(s)
- Claudia M. Toledo-Corral
- Department of Health Sciences & Health Equity and Research Education (HERE) Center, California State University Northridge, Northridge, California, USA
| | - Li Ding
- Department of Population and Public Health Sciences and University of Southern California, Los Angeles, California, USA
| | - Jeremy C. Morales
- Department of Population and Public Health Sciences and University of Southern California, Los Angeles, California, USA
| | - Tiffany M. Chapman
- Department of Population and Public Health Sciences and University of Southern California, Los Angeles, California, USA
| | - Melyssa B. Romero
- Department of Population and Public Health Sciences and University of Southern California, Los Angeles, California, USA
| | - Marc J. Weigensberg
- Department of Pediatrics, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
7
|
Nagarajan G, Aruna A, Chang YM, Alkhamis YA, Mathew RT, Chang CF. Effects of Osmotic Stress on the mRNA Expression of prl, prlr, gr, gh, and ghr in the Pituitary and Osmoregulatory Organs of Black Porgy, Acanthopagrus schlegelii. Int J Mol Sci 2023; 24:ijms24065318. [PMID: 36982391 PMCID: PMC10049143 DOI: 10.3390/ijms24065318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
In euryhaline teleost black porgy, Acanthopagrus schlegelii, the glucocorticoid receptor (gr), growth hormone receptor (ghr), prolactin (prl)-receptor (prlr), and sodium–potassium ATPase alpha subunit (α-nka) play essential physiological roles in the osmoregulatory organs, including the gill, kidney, and intestine, during osmotic stress. The present study aimed to investigate the impact of pituitary hormones and hormone receptors in the osmoregulatory organs during the transfer from freshwater (FW) to 4 ppt and seawater (SW) and vice versa in black porgy. Quantitative real-time PCR (Q-PCR) was carried out to analyze the transcript levels during salinity and osmoregulatory stress. Increased salinity resulted in decreased transcripts of prl in the pituitary, α-nka and prlr in the gill, and α-nka and prlr in the kidney. Increased salinity caused the increased transcripts of gr in the gill and α-nka in the intestine. Decreased salinity resulted in increased pituitary prl, and increases in α-nka and prlr in the gill, and α-nka, prlr, and ghr in the kidney. Taken together, the present results highlight the involvement of prl, prlr, gh, and ghr in the osmoregulation and osmotic stress in the osmoregulatory organs (gill, intestine, and kidney). Pituitary prl, and gill and intestine prlr are consistently downregulated during the increased salinity stress and vice versa. It is suggested that prl plays a more significant role in osmoregulation than gh in the euryhaline black porgy. Furthermore, the present results highlighted that the gill gr transcript’s role was solely to balance the homeostasis in the black porgy during salinity stress.
Collapse
Affiliation(s)
- Ganesan Nagarajan
- Department of Basic Sciences, PYD, King Faisal University, Al Ahsa 31982, Saudi Arabia
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan
- Department of Aquaculture, National Taiwan Ocean University, Keelung 20224, Taiwan
- Correspondence: (G.N.); (C.-F.C.); Tel.: +966-0135896810 (G.N.); +886-2-2462-2192 (ext. 5209) (C.-F.C.)
| | - Adimoolam Aruna
- Department of Aquaculture, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Yu-Ming Chang
- Department of Aquaculture, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Yousef Ahmed Alkhamis
- Animal and Fish Production Department, College of Agricultural and Food Sciences, King Faisal University, Hofuf-420, Al-Asha 31982, Saudi Arabia
- Fish Resources Research Center, King Faisal University, Hofuf-420, Al-Asha 31982, Saudi Arabia
| | - Roshmon Thomas Mathew
- Fish Resources Research Center, King Faisal University, Hofuf-420, Al-Asha 31982, Saudi Arabia
| | - Ching-Fong Chang
- Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 20224, Taiwan
- Department of Aquaculture, National Taiwan Ocean University, Keelung 20224, Taiwan
- Correspondence: (G.N.); (C.-F.C.); Tel.: +966-0135896810 (G.N.); +886-2-2462-2192 (ext. 5209) (C.-F.C.)
| |
Collapse
|
8
|
Mitsis T, Papageorgiou L, Papakonstantinou E, Diakou I, Pierouli K, Dragoumani K, Bacopoulou F, Kino T, Chrousos GP, Eliopoulos E, Vlachakis D. A Genomic Study of the Japanese Population Focusing on the Glucocorticoid Receptor Interactome Highlights Distinct Genetic Characteristics Associated with Stress Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1423:101-113. [PMID: 37525035 DOI: 10.1007/978-3-031-31978-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
All living organisms have been programmed to maintain a complex inner equilibrium called homeostasis, despite numerous adversities during their lifespan. Any threatening or perceived as such stimuli for homeostasis is termed a stressor, and a highly conserved response system called the stress response system has been developed to cope with these stimuli and maintain or reinstate homeostasis. The glucocorticoid receptor, a transcription factor belonging to the nuclear receptors protein superfamily, has a major role in the stress response system, and research on its interactome may provide novel information regarding the mechanisms underlying homeostasis maintenance. A list of 149 autosomal genes that have an essential role in GR function or are prime examples of GRE-containing genes was composed in order to gain a comprehensive view of the GR interactome. A search for SNPs on those particular genes was conducted on a dataset of 3554 Japanese individuals, with mentioned polymorphisms being annotated with relevant information from the ClinVar, LitVar, and dbSNP databases. Forty-two SNPs of interest and their genomic locations were identified. These SNPs have been associated with drug metabolism and neuropsychiatric, metabolic, and immune system disorders, while most of them were located in intronic regions. The frequencies of those SNPs were later compared with a dataset consisting of 1465 Korean individuals in order to find population-specific characteristics based on some of the identified SNPs of interest. The results highlighted.that rs1043618 frequencies were different in the two populations, with mentioned polymorphism having a potential role in chronic obstructive pulmonary disease in response to environmental stressors. This SNP is located in the HSPA1A gene, which codes for an essential GR co-chaperone, and such information showcases that similar gene may be novel genomic targets for managing or combatting stress-related pathologies.
Collapse
Affiliation(s)
- Thanasis Mitsis
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Louis Papageorgiou
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Eleni Papakonstantinou
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Io Diakou
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Katerina Pierouli
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Konstantina Dragoumani
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health & Precision Medicine, Athens, Greece
- National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Tomoshige Kino
- Department of Human Genetics, Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, Athens, Greece
- National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Elias Eliopoulos
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Dimitrios Vlachakis
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece.
- University Research Institute of Maternal and Child Health & Precision Medicine, Athens, Greece.
- National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece.
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
9
|
Kupczyk D, Studzińska R, Kołodziejska R, Baumgart S, Modrzejewska M, Woźniak A. 11β-Hydroxysteroid Dehydrogenase Type 1 as a Potential Treatment Target in Cardiovascular Diseases. J Clin Med 2022; 11:jcm11206190. [PMID: 36294507 PMCID: PMC9605099 DOI: 10.3390/jcm11206190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/27/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022] Open
Abstract
Glucocorticoids (GCs) belong to the group of steroid hormones. Their representative in humans is cortisol. GCs are involved in most physiological processes of the body and play a significant role in important biological processes, including reproduction, growth, immune responses, metabolism, maintenance of water and electrolyte balance, functioning of the central nervous system and the cardiovascular system. The availability of cortisol to the glucocorticoid receptor is locally controlled by the enzyme 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1). Evidence of changes in intracellular GC metabolism in the pathogenesis of obesity, metabolic syndrome (MetS) and cardiovascular complications highlights the role of selective 11β-HSD1 inhibition in the pharmacotherapy of these diseases. This paper discusses the role of 11β-HSD1 in MetS and its cardiovascular complications and the importance of selective inhibition of 11β-HSD1.
Collapse
Affiliation(s)
- Daria Kupczyk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
- Correspondence: (D.K.); (R.S.)
| | - Renata Studzińska
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089 Bydgoszcz, Poland
- Correspondence: (D.K.); (R.S.)
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
| | - Szymon Baumgart
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Jurasza 2, 85-089 Bydgoszcz, Poland
| | - Martyna Modrzejewska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland
| |
Collapse
|
10
|
Earhart ML, Blanchard TS, Strowbridge N, Bugg WS, Schulte PM. Gene expression and latitudinal variation in the stress response in Fundulus heteroclitus. Comp Biochem Physiol A Mol Integr Physiol 2022; 268:111188. [PMID: 35304270 DOI: 10.1016/j.cbpa.2022.111188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 10/18/2022]
Abstract
Atlantic killifish, Fundulus heteroclitus, are intertidal marsh fish found along the east coast of North America. Associated with the thermal gradient along this coast, northern and southern killifish populations are known to differ in morphology, behavior, and physiology, including in their cortisol stress response. Our goal was to explore population differences in the stress response and identify underlying molecular mechanisms. We measured responses to both acute and repeated stress in plasma cortisol, stress axis mRNA expression, and body condition in northern and southern killifish. Following an acute stressor, the southern population had higher cortisol levels than the northern population but there was no difference between populations following repeated stress. In the brain, both corticotropin releasing factor and its binding protein had higher expression in the southern than the northern population, but the northern population showed more changes in mRNA levels following a stressor. In the head kidney, Melanocortin 2 Receptor and steroidogenic acute regulatory protein mRNA levels were higher in the southern population suggesting a larger capacity for cortisol synthesis than in the northern fish. Lastly, the glucocorticoid receptor GR1 mRNA levels were greater in the liver of southern fish, suggesting a greater capacity to respond to cortisol, and GR2 had differential expression in the head kidney, suggesting an interpopulation difference in stress axis negative feedback loops. Southern, but not northern, fish were able to maintain body condition following stress, suggesting that these differences in the stress response may be important for adaptation across latitudes.
Collapse
Affiliation(s)
- Madison L Earhart
- Department of Zoology, University of British Columba, Vancouver, Canada.
| | - Tessa S Blanchard
- Department of Zoology, University of British Columba, Vancouver, Canada
| | - Nicholas Strowbridge
- Department of Zoology, University of British Columba, Vancouver, Canada; Institute of Biodiversity, Animal Health & Comparative Medicine, College of Medical, Veterinary & Life Sciences, University of Glasglow, Glasglow, UK
| | - William S Bugg
- Department of Zoology, University of British Columba, Vancouver, Canada; Department of Biological Sciences, University of Manitoba, Winnipeg, Canada
| | | |
Collapse
|
11
|
Cossin-Sevrin N, Hsu BY, Marciau C, Viblanc VA, Ruuskanen S, Stier A. Effect of prenatal glucocorticoids and thyroid hormones on developmental plasticity of mitochondrial aerobic metabolism, growth and survival: an experimental test in wild great tits. J Exp Biol 2022; 225:jeb243414. [PMID: 35420125 PMCID: PMC10216743 DOI: 10.1242/jeb.243414] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 04/11/2022] [Indexed: 11/20/2022]
Abstract
Developmental plasticity is partly mediated by transgenerational effects, including those mediated by the maternal endocrine system. Glucocorticoid and thyroid hormones may play central roles in developmental programming through their action on metabolism and growth. However, the mechanisms by which they affect growth and development remain understudied. One hypothesis is that maternal hormones directly affect the production and availability of energy-carrying molecules (e.g. ATP) by their action on mitochondrial function. To test this hypothesis, we experimentally increased glucocorticoid and thyroid hormones in wild great tit eggs (Parus major) to investigate their impact on offspring mitochondrial aerobic metabolism (measured in blood cells), and subsequent growth and survival. We show that prenatal glucocorticoid supplementation affected offspring cellular aerobic metabolism by decreasing mitochondrial density, maximal mitochondrial respiration and oxidative phosphorylation, while increasing the proportion of the maximum capacity being used under endogenous conditions. Prenatal glucocorticoid supplementation only had mild effects on offspring body mass, size and condition during the rearing period, but led to a sex-specific (females only) decrease in body mass a few months after fledging. Contrary to our expectations, thyroid hormone supplementation did not affect offspring growth or mitochondrial metabolism. Recapture probability as juveniles or adults was not significantly affected by prenatal hormonal treatment. Our results demonstrate that prenatal glucocorticoids can affect post-natal mitochondrial density and aerobic metabolism. The weak effects on growth and apparent survival suggest that nestlings were mostly able to compensate for the transient decrease in mitochondrial aerobic metabolism induced by prenatal glucocorticoids.
Collapse
Affiliation(s)
- Nina Cossin-Sevrin
- Department of Biology, University of Turku, FI-20014 Turku, Finland
- Université de Strasbourg, Centre National de la Recherche Scientifique, Institut Pluridisciplinaire Hubert Curien, UMR 7178, 67087 Strasbourg, France
| | - Bin-Yan Hsu
- Department of Biology, University of Turku, FI-20014 Turku, Finland
| | - Coline Marciau
- Department of Biology, University of Turku, FI-20014 Turku, Finland
- Institute for Marine and Antarctic Studies, University of Tasmania, Battery Point, TAS 7004, Australia
| | - Vincent A. Viblanc
- Université de Strasbourg, Centre National de la Recherche Scientifique, Institut Pluridisciplinaire Hubert Curien, UMR 7178, 67087 Strasbourg, France
| | - Suvi Ruuskanen
- Department of Biological and Environmental Sciences, University of Jyväskylä, FI-40014 Jyväskylä, Finland
| | - Antoine Stier
- Department of Biology, University of Turku, FI-20014 Turku, Finland
- Université Claude Bernard Lyon 1, CNRS, ENTPE, UMR 5023 LEHNA, F-69622 Villeurbanne, France
| |
Collapse
|
12
|
Häussler S, Sadri H, Ghaffari MH, Sauerwein H. Symposium review: Adipose tissue endocrinology in the periparturient period of dairy cows. J Dairy Sci 2022; 105:3648-3669. [PMID: 35181138 DOI: 10.3168/jds.2021-21220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/05/2022] [Indexed: 12/17/2022]
Abstract
The involvement of adipose tissue (AT) in metabolism is not limited to energy storage but turned out to be much more complex. We now know that in addition to lipid metabolism, AT is important in glucose homeostasis and AA metabolism and also has a role in inflammatory processes. With the discovery of leptin in 1994, the concept of AT being able to secrete messenger molecules collectively termed as adipokines, and acting in an endo-, para-, and autocrine manner emerged. Moreover, based on its asset of receptors, many stimuli from other tissues reaching AT via the bloodstream can also elicit distinct responses and thus integrate AT as a control element in the regulatory circuits of the whole body's functions. The protein secretome of human differentiated adipocytes was described to comprise more than 400 different proteins. However, in dairy cows, the characterization of the physiological time course of adipokines in AT during the transition from pregnancy to lactation is largely limited to the mRNA level; for the protein level, the analytical methods are limited and available assays often lack sound validation. In addition to proteinaceous adipokines, small compounds such as steroids can also be secreted from AT. Due to the lipophilic nature of steroids, they are stored in AT, but during the past years, AT became also known as being able to metabolize and even to generate steroid hormones de novo. In high-yielding dairy cows, AT is substantially mobilized due to increased energy requirements related to lactation. As to whether the steroidogenic system in AT is affected and may change during the common loss of body fat is largely unknown. Moreover, most research about AT in transition dairy cows is based on subcutaneous AT, whereas other depots have scarcely been investigated. This contribution aims to review the changes in adipokine mRNA and-where available-protein expression with time relative to calving in high-yielding dairy cows at different conditions, including parity, body condition, diet, specific feed supplements, and health disorders. In addition, the review provides insights into steroidogenic pathways in dairy cows AT, and addresses differences between fat depots where possible.
Collapse
Affiliation(s)
- Susanne Häussler
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany.
| | - Hassan Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, 516616471 Tabriz, Iran
| | - Morteza H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| |
Collapse
|
13
|
Vanderhaeghen T, Timmermans S, Watts D, Paakinaho V, Eggermont M, Vandewalle J, Wallaeys C, Van Wyngene L, Van Looveren K, Nuyttens L, Dewaele S, Vanden Berghe J, Lemeire K, De Backer J, Dirkx L, Vanden Berghe W, Caljon G, Ghesquière B, De Bosscher K, Wielockx B, Palvimo JJ, Beyaert R, Libert C. Reprogramming of glucocorticoid receptor function by hypoxia. EMBO Rep 2022; 23:e53083. [PMID: 34699114 PMCID: PMC8728616 DOI: 10.15252/embr.202153083] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 01/07/2023] Open
Abstract
Here, we investigate the impact of hypoxia on the hepatic response of glucocorticoid receptor (GR) to dexamethasone (DEX) in mice via RNA-sequencing. Hypoxia causes three types of reprogramming of GR: (i) much weaker induction of classical GR-responsive genes by DEX in hypoxia, (ii) a number of genes is induced by DEX specifically in hypoxia, and (iii) hypoxia induces a group of genes via activation of the hypothalamic-pituitary-adrenal (HPA) axis. Transcriptional profiles are reflected by changed GR DNA-binding as measured by ChIP sequencing. The HPA axis is induced by hypothalamic HIF1α and HIF2α activation and leads to GR-dependent lipolysis and ketogenesis. Acute inflammation, induced by lipopolysaccharide, is prevented by DEX in normoxia but not during hypoxia, and this is attributed to HPA axis activation by hypoxia. We unfold new physiological pathways that have consequences for patients suffering from GC resistance.
Collapse
|
14
|
Medger K, Prins A, Lutermann H, Ganswindt A, Ganswindt SB, Bennett NC. Repeatability of daily profiles of baseline glucocorticoid metabolites measured in the urine and faeces of eastern rock sengis (Elephantulus myurus). Gen Comp Endocrinol 2021; 312:113857. [PMID: 34284023 DOI: 10.1016/j.ygcen.2021.113857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/24/2021] [Accepted: 07/14/2021] [Indexed: 10/20/2022]
Abstract
Repeatability of hormone concentrations is of great interest for studies investigating the evolution of hormonal traits. Particularly the repeatability of glucocorticoids (GCs) in response to a stressor is frequently investigated, but often only point (initial and/or response value), or single measures are used. A new method takes into account the entire individual hormone profile and generates an individual profile repeatability (PR) score. The method was developed for response profiles, but it may also be valuable for baseline values in species with diurnal changes in hormone concentrations. GCs are determined in a variety of matrices, and repeatability can vary considerably depending on the matrix. We investigated the repeatability of baseline GC metabolite (GCM) concentrations measured in urine (uGCM) and faeces (fGCM) of captive eastern rock sengis (Elephantulus myurus) using the more traditional linear mixed model approach and the PR method. GCMs were assessed over 24 h and measurements were repeated twice with two weeks between replicates. A diurnal rhythm in GCM concentrations associated with the activity period of the sengis was found in urine, but not in the faeces. Urinary GCM concentrations exhibited a moderate repeatability, whereas the repeatability of fGCM concentrations was low. Urinary GCM concentrations and their repeatability differed between the sexes; with higher concentrations and lower PR scores in females. No such sex differences were apparent for fGCM concentrations and the PR score was not able to characterise repeatability of fGCM concentrations, which were lacking a distinct profile. The PR score enabled a successful quantification of the repeatability of the diurnal uGCM profiles. Hormone profile, sex and sample matrix can affect hormonal traits considerably and the results may be obscured if these factors are not carefully considered.
Collapse
Affiliation(s)
- Katarina Medger
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa.
| | - André Prins
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Heike Lutermann
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - André Ganswindt
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Stefanie B Ganswindt
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Nigel C Bennett
- Mammal Research Institute, Department of Zoology and Entomology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa; SARChI Chair of Mammal Behavioural Ecology and Physiology, Department of Zoology and Entomology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| |
Collapse
|
15
|
Kelly KP, Ellacott KLJ, Chen H, McGuinness OP, Johnson CH. Time-optimized feeding is beneficial without enforced fasting. Open Biol 2021; 11:210183. [PMID: 34610264 PMCID: PMC8492177 DOI: 10.1098/rsob.210183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/15/2021] [Indexed: 01/04/2023] Open
Abstract
Time-restricted feeding (TRF) studies underscore that when food is consumed during the daily cycle is important for weight gain/loss because the circadian clock rhythmically modulates metabolism. However, the interpretation of previous TRF studies has been confounded by study designs that introduced an extended period of enforced fasting. We introduce a novel time-optimized feeding (TOF) regimen that disentangles the effects of phase-dependent feeding from the effects of enforced fasting in mice, as well as providing a laboratory feeding protocol that more closely reflects the eating patterns of humans who usually have 24 hour access to food. Moreover, we test whether a sudden switch from ad libitum food access to TRF evokes a corticosterone (stress) response. Our data indicate that the timing of high-fat feeding under TOF allows most of the benefit of TRF without obligatory fasting or evoking a stress response. This benefit occurs through stable temporal coupling of carbohydrate/lipid oxidation with feeding. These results highlight that timing the ingestion of calorically dense foods to optimized daily phases will enhance lipid oxidation and thereby limit fat accumulation.
Collapse
Affiliation(s)
- Kevin P. Kelly
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
| | - Kate L. J. Ellacott
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Owen P. McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Carl Hirschie Johnson
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
16
|
Liu C, Ding J, Gao X, Du C, Hou C, Wu X, Shen W, Zhu J. Effects of acute low temperature stress on the hormones and gene expression of glucocorticoid receptor of large yellow croaker Larimichthys crocea. J Therm Biol 2021; 99:103018. [PMID: 34420651 DOI: 10.1016/j.jtherbio.2021.103018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/12/2021] [Accepted: 05/30/2021] [Indexed: 11/28/2022]
Abstract
The neuroendocrine system of fish responds to low temperature via regulating hormones. To explore the adaptability of Larimichthys crocea to low temperature, the levels of the plasma cortisol, thyroid stimulating hormone (TSH), triiodothyronine (T3), thyroxine (T4), total cholesterol (TC), and glucose were determined after exposure to low temperature and during subsequent rewarming. Furthermore, the mRNA expression of the glucocorticoid receptor (GR) gene was analyzed under the stress. We found that the levels of the plasma cortisol, TSH, T3, glucose, and TC increased under the low temperature stress, suggesting that elevated hormones may be conducive to promoting the mobilization of the glucose and lipid in L. crocea exposed to low temperature. During the rewarming period, the plasma cortisol level decreased, whereas the T3 level was still significantly higher than that in the control group. Notably, the plasma T4 level was unaffected by the temperature changes. Furthermore, the sequence alignment and phylogenetic tree analysis revealed that the GR protein of L. crocea had high homology and a similar protein structure with those from other teleosts. Under the low temperature stress, the GR mRNA expression increased in the brain and head kidney, whereas it basically returned to the control level following rewarming. These findings revealed the changes of the hormones and the potential function of the GR gene in L. crocea following exposure to low temperature, providing some insights into breeding low temperature-resistant varieties of L. crocea.
Collapse
Affiliation(s)
- Cheng Liu
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, China; Ningbo Academy of Oceanology and Fishery, Ningbo, 315012, Zhejiang, China
| | - Jie Ding
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, China; Ningbo Academy of Oceanology and Fishery, Ningbo, 315012, Zhejiang, China
| | - Xinming Gao
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Chen Du
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Congcong Hou
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Xiongfei Wu
- Ningbo Academy of Oceanology and Fishery, Ningbo, 315012, Zhejiang, China
| | - Weiliang Shen
- Ningbo Academy of Oceanology and Fishery, Ningbo, 315012, Zhejiang, China.
| | - Junquan Zhu
- Key Laboratory of Applied Marine Biotechnology of Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
17
|
Abstract
Learning to respond appropriately to one's surrounding environment is fundamental to survival. Importantly, however, individuals vary in how they respond to cues in the environment and this variation may be a key determinant of psychopathology. The ability of seemingly neutral cues to promote maladaptive behavior is a hallmark of several psychiatric disorders including, substance use disorder, post-traumatic stress disorder, eating disorders and obsessive-compulsive disorder. Thus, it is important to uncover the neural mechanisms by which such cues are able to attain inordinate control and promote psychopathological behavior. Here, we suggest that glucocorticoids play a critical role in this process. Glucocorticoids are primarily recognized as the main hormone secreted in response to stress but are known to exert their effects across the body and the brain, and to affect learning and memory, cognition and reward-related behaviors, among other things. Here we speculate that glucocorticoids act to facilitate a dopamine-dependent form of cue-reward learning that appears to be relevant to a number of psychiatric conditions. Specifically, we propose to utilize the sign-tracker/goal-tracker animal model as a means to capture individual variation in stimulus-reward learning and to isolate the role of glucocorticoid-dopamine interactions in mediating these individual differences. It is hoped that this framework will lead to the discovery of novel mechanisms that contribute to complex neuropsychiatric disorders and their comorbidity.
Collapse
Affiliation(s)
- Sofia A. Lopez
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Shelly B. Flagel
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
18
|
Okun JG, Rusu PM, Chan AY, Wu Y, Yap YW, Sharkie T, Schumacher J, Schmidt KV, Roberts-Thomson KM, Russell RD, Zota A, Hille S, Jungmann A, Maggi L, Lee Y, Blüher M, Herzig S, Keske MA, Heikenwalder M, Müller OJ, Rose AJ. Liver alanine catabolism promotes skeletal muscle atrophy and hyperglycaemia in type 2 diabetes. Nat Metab 2021; 3:394-409. [PMID: 33758419 DOI: 10.1038/s42255-021-00369-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 02/18/2021] [Indexed: 01/31/2023]
Abstract
Both obesity and sarcopenia are frequently associated in ageing, and together may promote the progression of related conditions such as diabetes and frailty. However, little is known about the pathophysiological mechanisms underpinning this association. Here we show that systemic alanine metabolism is linked to glycaemic control. We find that expression of alanine aminotransferases is increased in the liver in mice with obesity and diabetes, as well as in humans with type 2 diabetes. Hepatocyte-selective silencing of both alanine aminotransferase enzymes in mice with obesity and diabetes retards hyperglycaemia and reverses skeletal muscle atrophy through restoration of skeletal muscle protein synthesis. Mechanistically, liver alanine catabolism driven by chronic glucocorticoid and glucagon signalling promotes hyperglycaemia and skeletal muscle wasting. We further provide evidence for amino acid-induced metabolic cross-talk between the liver and skeletal muscle in ex vivo experiments. Taken together, we reveal a metabolic inter-tissue cross-talk that links skeletal muscle atrophy and hyperglycaemia in type 2 diabetes.
Collapse
Affiliation(s)
- Jürgen G Okun
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Patricia M Rusu
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Andrea Y Chan
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Yuqin Wu
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Yann W Yap
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Thomas Sharkie
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Jonas Schumacher
- Division of Molecular Metabolic Control, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kathrin V Schmidt
- Division of Inherited Metabolic Diseases, University Children's Hospital, Heidelberg, Germany
| | - Katherine M Roberts-Thomson
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Ryan D Russell
- Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Annika Zota
- Division of Molecular Metabolic Control, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital and Chair Molecular Metabolic Control, Technical University Munich, Neuherberg, Germany
| | - Susanne Hille
- Department of Internal Medicine III, University of Kiel, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Heidelberg and Kiel sites, Germany
| | - Andreas Jungmann
- German Center for Cardiovascular Research (DZHK), Heidelberg and Kiel sites, Germany
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany
| | - Ludovico Maggi
- Division of Molecular Metabolic Control, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Young Lee
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig, Leipzig, Germany
| | - Stephan Herzig
- Division of Molecular Metabolic Control, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine I, Heidelberg University Hospital and Chair Molecular Metabolic Control, Technical University Munich, Neuherberg, Germany
| | - Michelle A Keske
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), Heidelberg and Kiel sites, Germany
| | - Adam J Rose
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.
- Division of Molecular Metabolic Control, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
19
|
Patt M, Gysi J, Faresse N, Cidlowski JA, Odermatt A. Protein phosphatase 1 alpha enhances glucocorticoid receptor activity by a mechanism involving phosphorylation of serine-211. Mol Cell Endocrinol 2020; 518:110873. [PMID: 32585168 PMCID: PMC7606615 DOI: 10.1016/j.mce.2020.110873] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 04/17/2020] [Accepted: 05/18/2020] [Indexed: 10/24/2022]
Abstract
By acting as a ligand-dependent transcription factor the glucocorticoid receptor (GR) mediates the actions of glucocorticoids and regulates many physiological processes. An impaired regulation of glucocorticoid action has been associated with numerous disorders. Thus, the elucidation of underlying signaling pathways is essential to understand mechanisms of disrupted glucocorticoid function and contribution to diseases. This study found increased GR transcriptional activity upon overexpression of protein phosphatase 1 alpha (PP1α) in HEK-293 cells and decreased expression levels of GR-responsive genes following PP1α knockdown in the endogenous A549 cell model. Mechanistic investigations revealed reduced phosphorylation of GR-Ser211 following PP1α silencing and provided a first indication for an involvement of glycogen synthase kinase 3 (GSK-3). Thus, the present study identified PP1α as a novel post-translational activator of GR signaling, suggesting that disruption of PP1α function could lead to impaired glucocorticoid action and thereby contribute to diseases.
Collapse
Affiliation(s)
- Melanie Patt
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Missionsstrasse 64, 4055, Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| | - Joël Gysi
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| | | | - John A Cidlowski
- Signal Transduction Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, 27709, USA.
| | - Alex Odermatt
- Swiss Centre for Applied Human Toxicology (SCAHT), University of Basel, Missionsstrasse 64, 4055, Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
20
|
Fructose Consumption Affects Glucocorticoid Signaling in the Liver of Young Female Rats. Nutrients 2020; 12:nu12113470. [PMID: 33198224 PMCID: PMC7698302 DOI: 10.3390/nu12113470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/28/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
The effects of early-life fructose consumption on hepatic signaling pathways and their relation to the development of metabolic disorders in later life are not fully understood. To investigate whether fructose overconsumption at a young age induces alterations in glucocorticoid signaling that might contribute to development of metabolic disturbances, we analysed glucocorticoid receptor hormone-binding parameters and expression of its target genes involved in gluconeogenesis (phosphoenolpyruvate carboxykinase and glucose-6-phosphatase) and lipid metabolism (lipin-1), as well as redox and inflammatory status in the liver of female rats subjected to a fructose-rich diet immediately after weaning. The fructose diet increased hepatic corticosterone concentration, 11β-hydroxysteroid dehydrogenase type 1 level, glucocorticoid receptor protein level and hormone-binding activity, as well as lipin-1 level. The expression of glucose-6-phosphatase was reduced in fructose-fed rats, while phosphoenolpyruvate carboxykinase remained unaltered. The fructose-rich diet increased the level of fructose transporter GLUT2, while the expression of fructolytic enzymes fructokinase and aldolase B remained unaltered. The diet also affected pro-inflammatory pathways, but had no effect on the antioxidant defence system. In conclusion, a fructose-rich diet applied immediately after weaning promoted lipogenesis and enhanced hepatic glucocorticoid signaling, possibly to protect against inflammatory damage, but without an effect on gluconeogenesis and antioxidant enzymes. Yet, prolonged treatment might ultimately lead to more pronounced metabolic disturbances.
Collapse
|
21
|
Diz-Chaves Y, Herrera-Pérez S, González-Matías LC, Lamas JA, Mallo F. Glucagon-Like Peptide-1 (GLP-1) in the Integration of Neural and Endocrine Responses to Stress. Nutrients 2020; 12:nu12113304. [PMID: 33126672 PMCID: PMC7692797 DOI: 10.3390/nu12113304] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Glucagon like-peptide 1 (GLP-1) within the brain is produced by a population of preproglucagon neurons located in the caudal nucleus of the solitary tract. These neurons project to the hypothalamus and another forebrain, hindbrain, and mesolimbic brain areas control the autonomic function, feeding, and the motivation to feed or regulate the stress response and the hypothalamic-pituitary-adrenal axis. GLP-1 receptor (GLP-1R) controls both food intake and feeding behavior (hunger-driven feeding, the hedonic value of food, and food motivation). The activation of GLP-1 receptors involves second messenger pathways and ionic events in the autonomic nervous system, which are very relevant to explain the essential central actions of GLP-1 as neuromodulator coordinating food intake in response to a physiological and stress-related stimulus to maintain homeostasis. Alterations in GLP-1 signaling associated with obesity or chronic stress induce the dysregulation of eating behavior. This review summarized the experimental shreds of evidence from studies using GLP-1R agonists to describe the neural and endocrine integration of stress responses and feeding behavior.
Collapse
Affiliation(s)
- Yolanda Diz-Chaves
- CINBIO, Universidade de Vigo, Grupo FB3A, Laboratorio de Endocrinología, 36310 Vigo, Spain;
- Correspondence: (Y.D.-C.); (F.M.); Tel.: +34-(986)-130226 (Y.D.-C.); +34-(986)-812393 (F.M.)
| | - Salvador Herrera-Pérez
- CINBIO, Universidade de Vigo, Grupo FB3B, Laboratorio de Neurociencia, 36310 Vigo, Spain; (S.H.-P.); (J.A.L.)
| | | | - José Antonio Lamas
- CINBIO, Universidade de Vigo, Grupo FB3B, Laboratorio de Neurociencia, 36310 Vigo, Spain; (S.H.-P.); (J.A.L.)
| | - Federico Mallo
- CINBIO, Universidade de Vigo, Grupo FB3A, Laboratorio de Endocrinología, 36310 Vigo, Spain;
- Correspondence: (Y.D.-C.); (F.M.); Tel.: +34-(986)-130226 (Y.D.-C.); +34-(986)-812393 (F.M.)
| |
Collapse
|
22
|
Præstholm SM, Correia CM, Grøntved L. Multifaceted Control of GR Signaling and Its Impact on Hepatic Transcriptional Networks and Metabolism. Front Endocrinol (Lausanne) 2020; 11:572981. [PMID: 33133019 PMCID: PMC7578419 DOI: 10.3389/fendo.2020.572981] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022] Open
Abstract
Glucocorticoids (GCs) and the glucocorticoid receptor (GR) are important regulators of development, inflammation, stress response and metabolism, demonstrated in various diseases including Addison's disease, Cushing's syndrome and by the many side effects of prolonged clinical administration of GCs. These conditions include severe metabolic challenges in key metabolic organs like the liver. In the liver, GR is known to regulate the transcription of key enzymes in glucose and lipid metabolism and contribute to the regulation of circadian-expressed genes. Insights to the modes of GR regulation and the underlying functional mechanisms are key for understanding diseases and for the development of improved clinical uses of GCs. The activity and function of GR is regulated at numerous levels including ligand availability, interaction with heat shock protein (HSP) complexes, expression of GR isoforms and posttranslational modifications. Moreover, recent genomics studies show functional interaction with multiple transcription factors (TF) and coregulators in complex transcriptional networks controlling cell type-specific gene expression by GCs. In this review we describe the different regulatory steps important for GR activity and discuss how different TF interaction partners of GR selectively control hepatic gene transcription and metabolism.
Collapse
Affiliation(s)
| | | | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
23
|
de Guia RM. Stress, glucocorticoid signaling pathway, and metabolic disorders. Diabetes Metab Syndr 2020; 14:1273-1280. [PMID: 32755820 DOI: 10.1016/j.dsx.2020.06.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Glucocorticoids and the GR serve as an essential molecular mediator of stress and different physiologic processes. This review summarizes main findings from studies on the role of the GC/GR signaling in the modulation of genes for nutrient processing by the different organs involved in metabolic diseases. METHODS Descriptive review of relevant papers known to the author was conducted. RESULTS Several high-throughput screenings in the past 15 years have identified potential GR DNA-binding regions in different cell types with genes that are annotated to be important for the control of metabolism. Transcriptional regulation of these GC-responsive genes provides links between the hypothalamic-pituitary-adrenal axis (HPA) and systemic energy homeostasis in both physiological and pathophysiological states. Future studies must reconsider the use of agonist, the utilization of animal models of stress and metabolic disorders, and validation in humans. CONCLUSION This review recapitulates the significant role of the GC/GR signaling in molecular metabolic control and metabolic disorders. Potential future research focus and optimizations have also been identified.
Collapse
Affiliation(s)
- Roldan M de Guia
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; Keio Global Research Institute (KGRI) and Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan; Czech Centre for Phenogenomics (CCP), Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic.
| |
Collapse
|
24
|
Ruddick-Collins LC, Morgan PJ, Johnstone AM. Mealtime: A circadian disruptor and determinant of energy balance? J Neuroendocrinol 2020; 32:e12886. [PMID: 32662577 DOI: 10.1111/jne.12886] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/24/2020] [Accepted: 06/14/2020] [Indexed: 12/21/2022]
Abstract
Circadian rhythms play a critical role in the physiological processes involved in energy metabolism and energy balance (EB). A large array of metabolic processes, including the expression of many energy-regulating endocrine hormones, display temporal rhythms that are driven by both the circadian clock and food intake. Mealtime has been shown to be a compelling zeitgeber in peripheral tissue rhythms. Inconsistent signalling to the periphery, because of mismatched input from the central clock vs time of eating, results in circadian disruption in which central and/or peripheral rhythms are asynchronously time shifted or their amplitudes reduced. A growing body of evidence supports the negative health effects of circadian disruption, with strong evidence in murine models that mealtime-induced circadian disruption results in various metabolic consequences, including energy imbalance and weight gain. Increased weight gain has been reported to occur even without differences in energy intake, indicating an effect of circadian disruption on energy expenditure. However, the translation of these findings to humans is not well established because the ability to undertake rigorously controlled dietary studies that explore the chronic effects on energy regulation is challenging. Establishing the neuroendocrine changes in response to both acute and chronic variations in mealtime, along with observations in populations with routinely abnormal mealtimes, may provide greater insight into underlying mechanisms that influence long-term weight management under different meal patterns. Human studies should explore mechanisms through relevant biomarkers; for example, cortisol, leptin, ghrelin and other energy-regulating neuroendocrine factors. Mistiming between aggregate hormonal signals, or between hormones with their receptors, may cause reduced signalling intensity and hormonal resistance. Understanding how mealtimes may impact on the coordination of endocrine factors is essential for untangling the complex regulation of EB. Here a review is provided on current evidence of the impacts of mealtime on energy metabolism and the underlying neuroendocrine mechanisms, with a specific focus on human research.
Collapse
Affiliation(s)
| | - Peter J Morgan
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|
25
|
Comparison of high-intensity interval training and moderate-intensity continuous training in their effects on behavioral functions and CORT levels in streptozotocin-induced diabetic mice. SPORT SCIENCES FOR HEALTH 2020. [DOI: 10.1007/s11332-020-00661-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
26
|
MLL2 regulates glucocorticoid receptor-mediated transcription of ENACα in human retinal pigment epithelial cells. Biochem Biophys Res Commun 2020; 525:675-680. [PMID: 32139118 DOI: 10.1016/j.bbrc.2020.02.046] [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] [Received: 01/26/2020] [Accepted: 02/07/2020] [Indexed: 11/24/2022]
Abstract
Glucocorticoids require the glucocorticoid receptor (GR), a type of ligand-dependent nuclear receptor to transmit their downstream effects. Upon glucocorticoid binding, GR associates with glucocorticoid response elements (GREs) and recruits other transcriptional coregulators to activate or repress target gene transcription. Many SET-domain family proteins have been demonstrated to contribute to GR-mediated transcriptional activity. However, whether histone H3K4-specific methyltransferase plays a cell-type-specific role in GR transcriptional regulation remains poorly understood. In this report, we examined MLL2 (KMT2D), a histone-lysine methyltransferase that catalyzes histone H3 lysine 4 methylation (H3K4me). Furthermore, we demonstrated that MLL2 specifically regulates the transcription of some GR target genes (e.g., ENACα and FLJ20371) in ARPE-19 cells, but has no effect in A549 cells. Mechanistically, co-immunoprecipitation assays revealed that MLL2 is associated with GR in a ligand-independent manner in APRE-19 cells. Moreover, chromatin immunoprecipitation analyses demonstrated that MLL2 could co-occupy glucocorticoid response elements (GREs) of GR target genes along with GR following Dex stimulation. Finally, the FAIRE-qPCR results illustrated that MLL2 is pivotal in establishing chromatin structure accessibility at the GREs of ARPE-19 specific genes in the presence of Dex. Taken together, our study determined that MLL2 regulates GR-mediated transcription in a cell-type-specific manner, and we provide a molecular mechanism to explain the specific role of MLL2 in regulating GR target gene expression in ARPE-19 cells.
Collapse
|
27
|
Medger K, Bennett NC, Prins A, Lutermann H, Ganswindt A. Sex and dose-dependent responses of urinary and faecal glucocorticoid metabolite concentrations following an ACTH challenge in eastern rock sengis (Elephantulus myurus). Comp Biochem Physiol A Mol Integr Physiol 2020; 245:110696. [PMID: 32268194 DOI: 10.1016/j.cbpa.2020.110696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 01/18/2023]
Abstract
Non-invasive methods for measuring glucocorticoids and their metabolites are frequently used in ecological, behavioural and physiological studies of mammals. Using faeces, urine and other matrices for such a measurement has considerable advantages in comparison to more traditional methods, but also requires thorough validation of the methods used. Eastern rock sengis (Elephantulus myurus) are fascinating African mammals and the non-invasive monitoring of the adrenocortical activity opens up new opportunities to study their biology. We were able to validate two assays for measuring urinary (uGCM) and faecal glucocorticoid metabolite (fGCM) concentrations in this species using a dose-dependent challenge with adrenocorticotropic hormone (ACTH). A higher concentration of ACTH elicited higher uGCM and fGCM concentrations in both males and females. Interestingly, uGCM and fGCM concentrations and the responses to ACTH were higher in females than in males and small changes in faecal glucocorticoid metabolites could not be reliably detected in males. In contrast to ACTH, a saline injection did not result in an increase in uGCM or fGCM concentrations. The study also provided insight into when responses to a stressor are likely to be detected in the urine and faeces of sengis and opens up new opportunities to study the stress physiology of this and other sengi species. It further emphasises the importance of thoroughly validating non-invasive methods for measuring hormones in both sexes of a species and for incorporating dose-dependent approaches.
Collapse
Affiliation(s)
- Katarina Medger
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa.
| | - Nigel C Bennett
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa; Department of Zoology and Entomology, SARChI Chair of Mammal Behavioural Ecology and Physiology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - André Prins
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Heike Lutermann
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Andre Ganswindt
- Department of Zoology and Entomology, Mammal Research Institute, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa; Endocrine Research Laboratory, Department of Anatomy and Physiology, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort 0110, South Africa
| |
Collapse
|
28
|
Shokraneh M, Sadeghi AA, Mousavi SN, Esmaeilkhanian S, Chamani M. Effects of in ovo injection of nano-selenium and nano-zinc oxide and high eggshell temperature during late incubation on antioxidant activity, thyroid and glucocorticoid hormones and some blood metabolites in broiler hatchlings. ACTA ACUST UNITED AC 2020. [DOI: 10.4025/actascianimsci.v42i1.46029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
This experiment was conducted to evaluate the effects of in ovo injection of nano-selenium (Nano-Se) and nano-zinc oxide (Nano-ZnO) and high eggshell temperature (EST) during late incubation on blood parameters of broiler hatchlings. A total of 750 fertile eggs, were weighed and randomly distributed among 5 treatment groups on each of 5 replicate tray levels. The injection was performed on 17 d of incubation. Treatments included of: 1) Eggs not injected and incubated at normal EST (control); 2) Eggs not injected and incubated at high EST; 3) Eggs injected NaCl solution and incubated at high EST (sham); 4) Eggs injected NaCl solution containing 40 µg Nano-Se and incubated at high EST; 5) Eggs injected NaCl solution containing 500 µg Nano-ZnO and incubated at high EST. EST of 37.8ºC (normal) or 38.9ºC (high) was applied from d 19 to 21 of incubation. In ovo injection of Nano-Se and Nano-ZnO significantly increased activity of GSH-Px and SOD and total protein, but decreased the levels of corticosterone, cortisol, T4 and T3 at high EST. Injection of Nano-Se and Nano-ZnO had a significant role in alleviating the negative effects of high temperature incubation and heat stress by increased antioxidant activity and reduced oxidative stress.
Collapse
|
29
|
Manou-Stathopoulou V, Korbonits M, Ackland GL. Redefining the perioperative stress response: a narrative review. Br J Anaesth 2019; 123:570-583. [PMID: 31547969 DOI: 10.1016/j.bja.2019.08.011] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/21/2019] [Accepted: 08/11/2019] [Indexed: 12/13/2022] Open
Abstract
The systemic stress response triggered by surgical trauma is characterised by sterile inflammation preceding metabolic and neuroendocrine dysregulation. However, the relevance of the classically described 'stress response' is now highly questionable in an era where profound physiological deconditioning is common in older, frail surgical patients. Commonly used assessment techniques do not accurately reflect hypothalamic-pituitary-adrenal axis integrity after major surgery. Clinical interpretation of plasma concentrations of cortisol, the prototypical stress hormone, is rarely accurate, because of study heterogeneity, the inherently dynamic characteristics of cortisol production, and assay variability. Before surgery, chronic psychosocial stress and common cardiorespiratory co-morbidities are clinically relevant modifiers of neuroendocrine activation to acute stress/inflammation. The frequent development of multi-morbidity after major surgery further clouds the compartmentalised, discrete model of neuroendocrine activation after initial tissue injury. Starvation, impaired mobility, and sepsis after surgery generate distinct neuroendocrine profiles that challenge the conventional model of neuroendocrine activation. Basic science studies suggest that high circulating levels of cortisol may directly cause organ injury. Conversely, randomised controlled clinical trials investigating glucocorticoid supplementation have delivered contrasting results, with some suggesting a protective effect in the perioperative period. Here, we consider many of the confounding factors that have emerged to challenge the conventional model of the surgical stress response, and suggest that a more nuanced understanding of changes in hypothalamic-pituitary-adrenal axis physiology is warranted to advance perioperative medicine. Re-examining the perioperative stress response presents opportunities for improving outcomes through enhancing the understanding of the neuroendocrine aspects of preparation for and recovery from surgery.
Collapse
Affiliation(s)
- Vasiliki Manou-Stathopoulou
- Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gareth L Ackland
- Translational Medicine and Therapeutics, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
30
|
Kozłowska L, Janasik B, Nowicka K, Wąsowicz W. A urinary metabolomics study of a Polish subpopulation environmentally exposed to arsenic. J Trace Elem Med Biol 2019; 54:44-54. [PMID: 31109620 DOI: 10.1016/j.jtemb.2019.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/12/2019] [Accepted: 03/31/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Almost every organ in the human body can be affected by arsenic (As) exposure associated with various industrial processes, as well as with contaminated food, drinking water and polluted air. Much is known about high exposure to inorganic As but there is little data on the metabolic changes connected to a low exposure e.g. in people living in smelter areas. OBJECTIVES The objectives of the study were: (1) characterise urinary concentration of total arsenic (AsT) in Polish inhabitants of the vicinity of a copper smelter area, (2) speciation analysis of various forms of arsenic in girls (GL), boys (BL), women (WL) and men (ML) with a slightly elevated AsT concentration and age/sex matched groups with a substantially higher AsT concentration, (GH, BH, WH and MH - respectively), (3) comparison of metabolomics profiles of urine between the age/sex matched people with low and high AsT concentrations. METHODS Urine samples were analysed for total arsenic and its chemical forms (AsIII; AsV, methylarsonic acid, dimethylarsinic acid, arsenobetaine) using HPLC-ICP-MS. Untargeted metabolomics analysis of the urine samples was performed using UPLC system connected to Q-TOF-MS equipped with an electrospray source. The XCMS Online program was applied for feature detection, retention time correction, alignment, statistics, annotation and identification. Potentially identified compounds were fragmented and resulting spectra were compared to the spectra in the Human Metabolome Database. RESULTS Urine concentration of AsT was, as follows: GL 16.40 ± 0.83; GH 115.23 ± 50.52; BL 16.48 ± 0.83; BH 95.00 ± 50.03; WL 16.93 ± 1.21; WH 170.13 ± 96.47; ML 16.91 ± 1.20; MH 151.71 ± 84.31 μg/l and percentage of arsenobetaine in AsT was, as follows: GL 65.5 ± 13.8%, GH 87.2 ± 4.7%, BL 59.8 ± 12.5%, BH 90.5 ± 2.4%, WL 50.8 ± 14.1%, WH 90.4 ± 3.5%, ML 53.3 ± 10.0%, MH 74.6 ± 20.2%. In the people with low and high AsT concentrations there were significant differences in the intensity of signal (is.) from numerous compounds being metabolites of neurotransmitters, nicotine and hormones transformation (serotonin in the girls and women; catecholamines in the girls, boys and women; mineralocorticoids and glucocorticoids in the boys, androgens in the women and men and nicotine in the boys, women and men). These changes might have been associated with higher is. from metabolites of leucine, tryptophan, purine degradation (in the GH, WH), urea cycle (in the WH and MH), glycolysis (in the WH) and with lower is. from metabolites of tricarboxylic acid cycle (in the BH) in comparison with low AsT matched groups. In the MH vs. ML higher is. from metabolite of lipid peroxidation (4-hydroxy-2-nonenal) was observed. Additionally, the presence of significant differences was reported in is. from food components metabolites, which might have modulated the negative effects of As (vitamin C in the girls, boys and men, vitamin B6 in the girls, boys and women as well as phenolic compounds in the boys and girls). We hypothesize that the observed higher is. from metabolites of sulphate (in MH) and glucoronate degradation (in BH, WH and MH) than in the matched low AsT groups may be related to the impaired glucuronidation and sulfonation and higher is. from catecholamines, nicotine and hormones. CONCLUSION Our results indicated that even a low exposure to As is associated with metabolic changes and that urine metabolomics studies could be a good tool to reflect their wide spectrum connected to specific environmental exposure to As, e.g. in smelter areas.
Collapse
Affiliation(s)
- Lucyna Kozłowska
- Department of Dietetics, Faculty of Human Nutrition and Consumer Sciences, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776, Warsaw, Poland.
| | - Beata Janasik
- Departament of Biological and Environmental Monitoring, Nofer Institute of Occupational Medicine, Św. Teresy od Dzieciątka Jezus 8, 91-348, Łódź, Poland.
| | - Katarzyna Nowicka
- Department of Dietetics, Faculty of Human Nutrition and Consumer Sciences, Warsaw University of Life Sciences, Nowoursynowska 159c, 02-776, Warsaw, Poland.
| | - Wojciech Wąsowicz
- Departament of Biological and Environmental Monitoring, Nofer Institute of Occupational Medicine, Św. Teresy od Dzieciątka Jezus 8, 91-348, Łódź, Poland.
| |
Collapse
|
31
|
La Noce M, Mele L, Laino L, Iolascon G, Pieretti G, Papaccio G, Desiderio V, Tirino V, Paino F. Cytoplasmic Interactions between the Glucocorticoid Receptor and HDAC2 Regulate Osteocalcin Expression in VPA-Treated MSCs. Cells 2019; 8:cells8030217. [PMID: 30841579 PMCID: PMC6468918 DOI: 10.3390/cells8030217] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/26/2019] [Accepted: 02/28/2019] [Indexed: 12/19/2022] Open
Abstract
Epigenetic regulation has been considered an important mechanism for influencing stem cell differentiation. In particular, histone deacetylases (HDACs) have been shown to play a role in the osteoblast differentiation of mesenchymal stem cells (MSCs). In this study, the effect of the HDAC inhibitor, valproic acid (VPA), on bone formation in vivo by MSCs was determined. Surprisingly, VPA treatment, unlike other HDAC inhibitors, produced a well-organized lamellar bone tissue when MSCs–collagen sponge constructs were implanted subcutaneously into nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice, although a decrease of osteocalcin (OC) expression was observed. Consequently, we decided to investigate the molecular mechanisms by which VPA exerts such effects on MSCs. We identified the glucocorticoid receptor (GR) as being responsible for that downregulation, and suggested a correlation between GR and HDAC2 inhibition after VPA treatment, as evidenced by HDAC2 knockdown. Furthermore, using co-immunoprecipitation analysis, we showed for the first time in the cytoplasm, binding between GR and HDAC2. Additionally, chromatin immunoprecipitation (ChIP) assays confirmed the role of GR in OC downregulation, showing recruitment of GR to the nGRE element in the OC promoter. In conclusion, our results highlight the existence of a cross-talk between GR and HDAC2, providing a mechanistic explanation for the influence of the HDAC inhibitor (namely VPA) on osteogenic differentiation in MSCs. Our findings open new directions in targeted therapies, and offer new insights into the regulation of MSC fate determination.
Collapse
Affiliation(s)
- Marcella La Noce
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Luigi Mele
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Luigi Laino
- Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania, "Luigi Vanvitelli", 80121 Naples, Italy.
| | - Giovanni Iolascon
- Department of Medical and Surgical Specialties and Dentistry, University of Campania "Luigi Vanvitelli", 80121 Naples, Italy.
| | - Gorizio Pieretti
- Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania, "Luigi Vanvitelli", 80121 Naples, Italy.
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Virginia Tirino
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Francesca Paino
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy.
| |
Collapse
|
32
|
Hu X, Wang Y, Sheikhahmadi A, Li X, Buyse J, Lin H, Song Z. Effects of glucocorticoids on lipid metabolism and AMPK in broiler chickens' liver. Comp Biochem Physiol B Biochem Mol Biol 2019; 232:23-30. [PMID: 30790719 DOI: 10.1016/j.cbpb.2019.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 02/06/2019] [Indexed: 12/21/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) plays a pivotal role in the regulation of carbohydrate, lipid, and protein metabolism in animals. In this study, we examined whether any cross talk exists between glucocorticoids and AMPK in the regulation of the liver bile acid biosynthesis pathway. Dexamethasone treatment decreased the growth performance of broiler chickens. The liver mRNA levels of fatty acid transport protein (FATP-1), farnesoid X receptor (FXR), AMPK alpha 1 subunit (AMPKα1), and glucocorticoid receptor were significantly upregulated in DEX-treated broilers; the gene expression of liver cholesterol 7 alpha-hydroxylase (CYP7A1) was significantly downregulated. The protein level of liver CYP7A1 was significantly decreased by DEX treatment at both 24 and 72 h, while the protein level of p-AMPK/ t-AMPK stayed unchanged. In the in vitro cultured hepatocytes, compound C pretreatment blocked the increase in CYP7A1 protein level by DEX and significantly suppressed FATP-1, SREBP-1c, FXR, and CYP7A1 gene expression stimulated by DEX. Compound C treatment significantly reduces the protein level of p-AMPK, and the combination of compound C and DEX significantly reduces the protein level of t-AMPK. Thus, glucocorticoids affected liver AMPK and the bile acid synthesis signal pathway, and AMPK might be involved in the glucocorticoid effect of liver bile acid synthesis.
Collapse
Affiliation(s)
- Xiyi Hu
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Yufeng Wang
- Division Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Kasteelpark Arenberg 30, 3001 Leuven, Belgium
| | - Ardashir Sheikhahmadi
- Department of Animal Science, Faculty of Agriculture, University of Kurdistan, Sanandaj 66177-15175, Iran
| | - Xianlei Li
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Johan Buyse
- Division Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Kasteelpark Arenberg 30, 3001 Leuven, Belgium
| | - Hai Lin
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Zhigang Song
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong 271018, China.
| |
Collapse
|
33
|
Zhang X, Yang S, Chen J, Su Z. Unraveling the Regulation of Hepatic Gluconeogenesis. Front Endocrinol (Lausanne) 2019; 9:802. [PMID: 30733709 PMCID: PMC6353800 DOI: 10.3389/fendo.2018.00802] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/20/2018] [Indexed: 02/05/2023] Open
Abstract
Hepatic gluconeogenesis, de novo glucose synthesis from available precursors, plays a crucial role in maintaining glucose homeostasis to meet energy demands during prolonged starvation in animals. The abnormally increased rate of hepatic gluconeogenesis contributes to hyperglycemia in diabetes. Gluconeogenesis is regulated on multiple levels, such as hormonal secretion, gene transcription, and posttranslational modification. We review here the molecular mechanisms underlying the transcriptional regulation of gluconeogenesis in response to nutritional and hormonal changes. The nutrient state determines the hormone release, which instigates the signaling cascades in the liver to modulate the activities of various transcriptional factors through various post-translational modifications like phosphorylation, methylation, and acetylation. AMP-activated protein kinase (AMPK) can mediate the activities of some transcription factors, however its role in the regulation of gluconeogenesis remains uncertain. Metformin, a primary hypoglycemic agent of type 2 diabetes, ameliorates hyperglycemia predominantly through suppression of hepatic gluconeogenesis. Several molecular mechanisms have been proposed to be metformin's mode of action.
Collapse
Affiliation(s)
| | | | | | - Zhiguang Su
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Theiler-Schwetz V, Zaufel A, Schlager H, Obermayer-Pietsch B, Fickert P, Zollner G. Bile acids and glucocorticoid metabolism in health and disease. Biochim Biophys Acta Mol Basis Dis 2019; 1865:243-251. [DOI: 10.1016/j.bbadis.2018.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/18/2018] [Accepted: 08/01/2018] [Indexed: 12/12/2022]
|
35
|
Bursać B, Djordjevic A, Veličković N, Milutinović DV, Petrović S, Teofilović A, Gligorovska L, Preitner F, Tappy L, Matić G. Involvement of glucocorticoid prereceptor metabolism and signaling in rat visceral adipose tissue lipid metabolism after chronic stress combined with high-fructose diet. Mol Cell Endocrinol 2018; 476:110-118. [PMID: 29729371 DOI: 10.1016/j.mce.2018.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/04/2018] [Accepted: 04/29/2018] [Indexed: 12/28/2022]
Abstract
Both fructose overconsumption and increased glucocorticoids secondary to chronic stress may contribute to overall dyslipidemia. In this study we specifically assessed the effects and interactions of dietary fructose and chronic stress on lipid metabolism in the visceral adipose tissue (VAT) of male Wistar rats. We analyzed the effects of 9-week 20% high fructose diet and 4-week chronic unpredictable stress, separately and in combination, on VAT histology, glucocorticoid prereceptor metabolism, glucocorticoid receptor subcellular redistribution and expression of major metabolic genes. Blood triglycerides and fatty acid composition were also measured to assess hepatic Δ9 desaturase activity. The results showed that fructose diet increased blood triglycerides and Δ9 desaturase activity. On the other hand, stress led to corticosterone elevation, glucocorticoid receptor activation and decrease in adipocyte size, while phosphoenolpyruvate carboxykinase, adipose tissue triglyceride lipase, FAT/CD36 and sterol regulatory element binding protein-1c (SREBP-1c) were increased, pointing to VAT lipolysis and glyceroneogenesis. The combination of stress and fructose diet was associated with marked stimulation of fatty acid synthase and acetyl-CoA carboxylase mRNA level and with increased 11β-hydroxysteroid dehydrogenase type 1 and hexose-6-phosphate dehydrogenase protein levels, suggesting a coordinated increase in hexose monophosphate shunt and de novo lipogenesis. It however did not influence the level of peroxisome proliferator-activated receptor-gamma, SREBP-1c and carbohydrate responsive element-binding protein. In conclusion, our results showed that only combination of dietary fructose and stress increase glucocorticoid prereceptor metabolism and stimulates lipogenic enzyme expression suggesting that interaction between stress and fructose may be instrumental in promoting VAT expansion and dysfunction.
Collapse
Affiliation(s)
- Biljana Bursać
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11000, Belgrade, Serbia
| | - Ana Djordjevic
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11000, Belgrade, Serbia.
| | - Nataša Veličković
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11000, Belgrade, Serbia
| | - Danijela Vojnović Milutinović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11000, Belgrade, Serbia
| | - Snježana Petrović
- Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Tadeuša Košćuška 1, Belgrade, 11129, Serbia
| | - Ana Teofilović
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11000, Belgrade, Serbia
| | - Ljupka Gligorovska
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11000, Belgrade, Serbia
| | - Frederic Preitner
- Mouse Metabolic Facility (MEF), Center for Integrative genomics, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Luc Tappy
- Department of Physiology, University of Lausanne, UNIL-CHUV, Rue du Bugnon 7, CH-1005, Lausanne, Switzerland
| | - Gordana Matić
- Department of Biochemistry, Institute for Biological Research "Siniša Stanković", University of Belgrade, 142 Despot Stefan Blvd., 11000, Belgrade, Serbia
| |
Collapse
|
36
|
Lamri A, Pigeyre M, Garver WS, Meyre D. The Extending Spectrum of NPC1-Related Human Disorders: From Niemann-Pick C1 Disease to Obesity. Endocr Rev 2018; 39:192-220. [PMID: 29325023 PMCID: PMC5888214 DOI: 10.1210/er.2017-00176] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 01/02/2018] [Indexed: 12/22/2022]
Abstract
The Niemann-Pick type C1 (NPC1) protein regulates the transport of cholesterol and fatty acids from late endosomes/lysosomes and has a central role in maintaining lipid homeostasis. NPC1 loss-of-function mutations in humans cause NPC1 disease, a rare autosomal-recessive lipid-storage disorder characterized by progressive and lethal neurodegeneration, as well as liver and lung failure, due to cholesterol infiltration. In humans, genome-wide association studies and post-genome-wide association studies highlight the implication of common variants in NPC1 in adult-onset obesity, body fat mass, and type 2 diabetes. Heterozygous human carriers of rare loss-of-function coding variants in NPC1 display an increased risk of morbid adult obesity. These associations have been confirmed in mice models, showing an important interaction with high-fat diet. In this review, we describe the current state of knowledge for NPC1 variants in relationship to pleiotropic effects on metabolism. We provide evidence that NPC1 gene variations may predispose to common metabolic diseases by modulating steroid hormone synthesis and/or lipid homeostasis. We also propose several important directions of research to further define the complex roles of NPC1 in metabolism. This review emphasizes the contribution of NPC1 to obesity and its metabolic complications.
Collapse
Affiliation(s)
- Amel Lamri
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Marie Pigeyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.,INSERM 1190, European Genomics Institute for Diabetes, University of Lille, CHRU Lille, Lille, France
| | - William S Garver
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
37
|
Abstract
BACKGROUND An increased prevalence of arterial stiffness in systemic lupus erythematosus (SLE) patients has been established, but the mechanisms of progression of arterial stiffness with increasing age have not been fully explored. OBJECTIVES To investigate age-related progression of arterial stiffness among SLE patients relative to healthy controls. METHODS A total of 161 female SLE patients who were enrolled in the Chinese SLE Treatment and Research group (CSTAR) registry and 135 age-matched healthy control subjects participated in this cross-sectional investigation. Traditional cardiovascular risk factors and SLE-related parameters were assessed on the day that brachial-ankle pulse wave velocity (baPWV) was examined. SAS 9.3 was used to perform all statistical analyses in this study. Linear regression and curvilinear regression models were utilized to analyze the association between age and arterial stiffness. RESULTS Arterial stiffness based on baPWV significantly differed between the SLE patients and controls in the different age groups, and within the SLE group. The baPWV increments for each age group (<25, 25-34, 35-45, and >45) were 30 cm/s, 52 cm/s, and 121 cm/s for the controls and 61 cm/s, 132 cm/s, and 155 cm/s for the SLE patients, respectively. Curvilinear regression and linear regression revealed various trends of increased arterial stiffness among the SLE patients compared with the healthy controls. The correlation coefficients between age and arterial stiffness significantly differed among the SLE patients relative to the healthy controls (correlation coefficients of 0.46478 and 0.52612, respectively; t = 2.05; P = 0.0409). Multivariate analysis revealed that age, mean blood pressure (BP) (P < 0.0001), erythrocyte sedimentation rate (ESR) (P = 0.0073), prednisone course (P = 0.0144), and SLE disease activity (P = 0.0405) were associated with arterial stiffness among the SLE patients. Further, these patients exhibited earlier exposure to and higher frequencies of several risk factors compared with the controls in each age group (<25 years: OR = 6.3253; 25-34 years: OR = 3.1043; 35-45 years: OR = 3.1316; >45 years: OR = 3.6020). CONCLUSIONS The mechanisms of the age-related progression of arterial stiffness differed among the SLE patients relative to the healthy controls. Furthermore, accelerated arterial stiffness was observed among the SLE patients relative to the healthy controls with advancing age.
Collapse
|
38
|
Scheideler M, Herzig S, Georgiadi A. Endocrine and autocrine/paracrine modulators of brown adipose tissue mass and activity as novel therapeutic strategies against obesity and type 2 diabetes. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0043/hmbci-2017-0043.xml. [PMID: 28850545 DOI: 10.1515/hmbci-2017-0043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 07/28/2017] [Indexed: 12/17/2022]
Abstract
The dramatically increasing world-wide prevalence of obesity is recognized as a risk factor for the development of various diseases. The growing research on the role of adipose tissue in controlling energy homeostasis and insulin sensitivity has revealed that the promotion of brown adipose tissue (BAT) activity and the browning of white adipose tissue (WAT) leads to multiple health benefits and prevents obesity and type 2 diabetes (T2D). Inducible thermogenic adipocytes do exist in adult humans and are linked with increased energy combustion and lower body fat mass. Thus brown adipocytes are currently placed at the center of attention for novel therapeutic strategies against metabolic diseases such as obesity and diabetes. Besides the classical, norepinephrine-mediated sympathetic recruitment and activation of thermogenic adipocytes, a number of novel circulating factors have been recently identified to have a positive or negative impact on thermogenic adipocyte formation and activity. In this review their mechanism of action and the plausible therapeutic applications will be summarized and discussed.
Collapse
|
39
|
Effects of high-intensity interval versus mild-intensity endurance training on metabolic phenotype and corticosterone response in rats fed a high-fat or control diet. PLoS One 2017; 12:e0181684. [PMID: 28727846 PMCID: PMC5519214 DOI: 10.1371/journal.pone.0181684] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 07/04/2017] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to compare the effects of high-intensity interval training (HI) to mild-intensity endurance training (ME), combined with a high-fat diet (HFD) or control diet (CD) on metabolic phenotype and corticosterone levels in rats. Fifty-three rats were randomized to 6 groups according to diet and training regimen as follows: CD and sedentary (CS, n = 11), CD and ME (CME, n = 8), CD and HI (CHI, n = 8), HFD and sedentary (HS, n = 10), HFD and ME (HME, n = 8), and HFD and HI (HHI, n = 8). All exercise groups were trained for 10 weeks and had matched running distances. Dietary intake, body composition, blood metabolites, and corticosterone levels were measured. Histological lipid droplets were observed in the livers. The HFD led to hyperglycemia, hyperlipidemia and higher body fat (all, P < 0.01, η2 > 0.06), as well as higher corticosterone levels (P < 0.01, η2 = 0.09) compared with the CD groups. Exercise training improved fat weight, glucose, and lipid profiles, and reduced corticosterone levels (P < 0.01, η2 = 0.123). Furthermore, body and fat weight, serum glucose and triglycerides, lipid content in the liver, and corticosterone levels (P < 0.05) were lower with HI training compared to ME training. Reductions in HFD-induced body weight gain, blood glucose and lipid profiles, and corticosterone levels, as well as improvements in QUICKI were better with HHI compared to HME. Correlation analyses revealed that corticosterone levels were significantly associated with phenotype variables (P < 0.01). Corticosterone level was inversely correlated with QUICKI (r = −0.38, P < 0.01). Altogether, these results indicate that HFD may elicit an exacerbated basal serum corticosterone level and thus producing a metabolic imbalance. Compared with ME training, HI training contributes to greater improvements in metabolic and corticosterone responses, leading to a greater reduction in susceptibility to HFD-induced disorders.
Collapse
|
40
|
Pošćić N, Montanari T, D’Andrea M, Licastro D, Pilla F, Ajmone-Marsan P, Minuti A, Sgorlon S. Breed and adaptive response modulate bovine peripheral blood cells' transcriptome. J Anim Sci Biotechnol 2017; 8:11. [PMID: 28149510 PMCID: PMC5264304 DOI: 10.1186/s40104-017-0143-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 01/07/2017] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Adaptive response includes a variety of physiological modifications to face changes in external or internal conditions and adapt to a new situation. The acute phase proteins (APPs) are reactants synthesized against environmental stimuli like stress, infection, inflammation. METHODS To delineate the differences in molecular constituents of adaptive response to the environment we performed the whole-blood transcriptome analysis in Italian Holstein (IH) and Italian Simmental (IS) breeds. For this, 663 IH and IS cows from six commercial farms were clustered according to the blood level of APPs. Ten extreme individuals (five APP+ and APP- variants) from each farm were selected for the RNA-seq using the Illumina sequencing technology. Differentially expressed (DE) genes were analyzed using dynamic impact approach (DIA) and DAVID annotation clustering. Milk production data were statistically elaborated to assess the association of APP+ and APP- gene expression patterns with variations in milk parameters. RESULTS The overall de novo assembly of cDNA sequence data generated 13,665 genes expressed in bovine blood cells. Comparative genomic analysis revealed 1,152 DE genes in the comparison of all APP+ vs. all APP- variants; 531 and 217 DE genes specific for IH and IS comparison respectively. In all comparisons overexpressed genes were more represented than underexpressed ones. DAVID analysis revealed 369 DE genes across breeds, 173 and 73 DE genes in IH and IS comparison respectively. Among the most impacted pathways for both breeds were vitamin B6 metabolism, folate biosynthesis, nitrogen metabolism and linoleic acid metabolism. CONCLUSIONS Both DIA and DAVID approaches produced a high number of significantly impacted genes and pathways with a narrow connection to adaptive response in cows with high level of blood APPs. A similar variation in gene expression and impacted pathways between APP+ and APP- variants was found between two studied breeds. Such similarity was also confirmed by annotation clustering of the DE genes. However, IH breed showed higher and more differentiated impacts compared to IS breed and such particular features in the IH adaptive response could be explained by its higher metabolic activity. Variations of milk production data were significantly associated with APP+ and APP- gene expression patterns.
Collapse
Affiliation(s)
- Nataliya Pošćić
- Department of Agriculture, Food, Environment and Animal Science (DI4A), University of Udine, via delle Scienze 206, 33100 Udine, Italy
| | - Tommaso Montanari
- Department of Agriculture, Food, Environment and Animal Science (DI4A), University of Udine, via delle Scienze 206, 33100 Udine, Italy
| | - Mariasilvia D’Andrea
- Department of Agricultural, Environmental and Food Sciences, University of Molise, via F. De Sanctis snc, 86100 Campobasso, Italy
| | - Danilo Licastro
- CBM S.c.r.l, SS 14 – km 163.5 AREA Science Park, 34149 Basovizza, TS Italy
| | - Fabio Pilla
- Department of Agricultural, Environmental and Food Sciences, University of Molise, via F. De Sanctis snc, 86100 Campobasso, Italy
| | - Paolo Ajmone-Marsan
- Institute of Zootechnics, Catholic University of the Sacred Heart, via Emilia Parmense 84, 29133 Piacenza, Italy
| | - Andrea Minuti
- Institute of Zootechnics, Catholic University of the Sacred Heart, via Emilia Parmense 84, 29133 Piacenza, Italy
| | - Sandy Sgorlon
- Department of Agriculture, Food, Environment and Animal Science (DI4A), University of Udine, via delle Scienze 206, 33100 Udine, Italy
| |
Collapse
|
41
|
Dunford EC, Riddell MC. The Metabolic Implications of Glucocorticoids in a High-Fat Diet Setting and the Counter-Effects of Exercise. Metabolites 2016; 6:metabo6040044. [PMID: 27929385 PMCID: PMC5192450 DOI: 10.3390/metabo6040044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 11/25/2016] [Accepted: 11/30/2016] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoids (GCs) are steroid hormones, naturally produced by activation of the hypothalamic-pituitary-adrenal (HPA) axis, that mediate the immune and metabolic systems. Synthetic GCs are used to treat a number of inflammatory conditions and diseases including lupus and rheumatoid arthritis. Generally, chronic or high dose GC administration is associated with side effects such as steroid-induced skeletal muscle loss, visceral adiposity, and diabetes development. Patients who are taking exogenous GCs could also be more susceptible to poor food choices, but the effect that increasing fat consumption in combination with elevated exogenous GCs has only recently been investigated. Overall, these studies show that the damaging metabolic effects initiated through exogenous GC treatment are significantly amplified when combined with a high fat diet (HFD). Rodent studies of a HFD and elevated GCs demonstrate more glucose intolerance, hyperinsulinemia, visceral adiposity, and skeletal muscle lipid deposition when compared to rodents subjected to either treatment on its own. Exercise has recently been shown to be a viable therapeutic option for GC-treated, high-fat fed rodents, with the potential mechanisms still being examined. Clinically, these mechanistic studies underscore the importance of a low fat diet and increased physical activity levels when individuals are given a course of GC treatment.
Collapse
Affiliation(s)
- Emily C Dunford
- School of Kinesiology and Health Science, Faculty of Health, Muscle Health Research Center and Physical Activity and Chronic Disease Unit, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada.
| | - Michael C Riddell
- School of Kinesiology and Health Science, Faculty of Health, Muscle Health Research Center and Physical Activity and Chronic Disease Unit, York University, 4700 Keele Street, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
42
|
Roqueta-Rivera M, Esquejo RM, Phelan PE, Sandor K, Daniel B, Foufelle F, Ding J, Li X, Khorasanizadeh S, Osborne TF. SETDB2 Links Glucocorticoid to Lipid Metabolism through Insig2a Regulation. Cell Metab 2016; 24:474-484. [PMID: 27568546 PMCID: PMC5023502 DOI: 10.1016/j.cmet.2016.07.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/28/2016] [Accepted: 07/28/2016] [Indexed: 10/21/2022]
Abstract
Transcriptional and chromatin regulations mediate the liver response to nutrient availability. The role of chromatin factors involved in hormonal regulation in response to fasting is not fully understood. We have identified SETDB2, a glucocorticoid-induced putative epigenetic modifier, as a positive regulator of GR-mediated gene activation in liver. Insig2a increases during fasting to limit lipid synthesis, but the mechanism of induction is unknown. We show Insig2a induction is GR-SETDB2 dependent. SETDB2 facilitates GR chromatin enrichment and is key to glucocorticoid-dependent enhancer-promoter interactions. INSIG2 is a negative regulator of SREBP, and acute glucocorticoid treatment decreased active SREBP during refeeding or in livers of Ob/Ob mice, both systems of elevated SREBP-1c-driven lipogenesis. Knockdown of SETDB2 or INSIG2 reversed the inhibition of SREBP processing. Overall, these studies identify a GR-SETDB2 regulatory axis of hepatic transcriptional reprogramming and identify SETDB2 as a potential target for metabolic disorders with aberrant glucocorticoid actions.
Collapse
Affiliation(s)
- Manuel Roqueta-Rivera
- Sanford Burnham Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA
| | - Ryan M Esquejo
- Sanford Burnham Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA
| | - Peter E Phelan
- Sanford Burnham Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA
| | - Katalin Sandor
- Sanford Burnham Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA
| | - Bence Daniel
- Sanford Burnham Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA
| | - Fabienne Foufelle
- INSERM, UMR-S 872, Centre de Recherches des Cordeliers, 75006 Paris, France; Université Pierre et Marie Curie-Paris, 75005 Paris, France
| | - Jun Ding
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Boulevard, Orlando, FL 32827, USA
| | - Xiaoman Li
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Boulevard, Orlando, FL 32827, USA
| | - Sepideh Khorasanizadeh
- Sanford Burnham Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA
| | - Timothy F Osborne
- Sanford Burnham Prebys Medical Discovery Institute, 6400 Sanger Road, Orlando, FL 32827, USA.
| |
Collapse
|
43
|
Nagasawa K, Matsuura N, Takeshita Y, Ito S, Sano Y, Yamada Y, Uchinaka A, Murohara T, Nagata K. Attenuation of cold stress-induced exacerbation of cardiac and adipose tissue pathology and metabolic disorders in a rat model of metabolic syndrome by the glucocorticoid receptor antagonist RU486. Nutr Diabetes 2016; 6:e207. [PMID: 27110688 PMCID: PMC4855259 DOI: 10.1038/nutd.2016.14] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 03/07/2016] [Accepted: 03/16/2016] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Chronic stress affects the central nervous system as well as endocrine, metabolic and immune systems. However, the effects of cold stress on cardiovascular and metabolic disorders in metabolic syndrome (MetS) have remained unclear. We recently characterized DahlS.Z-Lepr(fa)/Lepr(fa) (DS/obese) rats, derived from a cross between Dahl salt-sensitive and Zucker rats, as a new animal model of MetS. We have now investigated the effects of chronic cold stress and glucocorticoid receptor (GR) blockade on cardiac and adipose tissue pathology as well as on metabolic parameters in this model. METHODS DS/obese rats were exposed to cold stress (immersion in ice-cold water to a depth of 1-2 cm for 2 h per day) with or without subcutaneous injection of the GR antagonist RU486 (2 mg kg(-1)day(-1)) for 4 weeks beginning at 9 weeks of age. Age-matched homozygous lean (DahlS.Z-Lepr(+)/Lepr(+)) littermates served as a control. RESULTS Chronic cold stress exacerbated hypertension as well as left ventricular (LV) hypertrophy, fibrosis and diastolic dysfunction in DS/obese rats in a manner sensitive to RU486 treatment. Cold stress with or without RU486 did not affect body weight or fat mass. In contrast, cold stress further increased cardiac oxidative stress as well as macrophage infiltration and proinflammatory gene expression in LV and visceral fat tissue, with all of these effects being attenuated by RU486. Cold stress also further increased GR and 11β-hydroxysteroid dehydrogenase type 1 mRNA and protein abundance in LV and visceral adipose tissue, and these effects were again inhibited by RU486. In addition, RU486 ameliorated the stress-induced aggravation of dyslipidemia, glucose intolerance and insulin resistance in DS/obese rats. CONCLUSIONS Our results implicate GR signaling in cold stress-induced exacerbation of cardiac and adipose tissue pathology as well as of abnormal glucose and lipid metabolism in a rat model of MetS.
Collapse
Affiliation(s)
- K Nagasawa
- Department of Pathophysiological Laboratory Sciences, Nagoya, Japan
| | - N Matsuura
- Department of Pathophysiological Laboratory Sciences, Nagoya, Japan
| | - Y Takeshita
- Department of Pathophysiological Laboratory Sciences, Nagoya, Japan
| | - S Ito
- Department of Pathophysiological Laboratory Sciences, Nagoya, Japan
| | - Y Sano
- Department of Pathophysiological Laboratory Sciences, Nagoya, Japan
| | - Y Yamada
- Department of Pathophysiological Laboratory Sciences, Nagoya, Japan
| | - A Uchinaka
- Department of Pathophysiological Laboratory Sciences, Nagoya, Japan
| | - T Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - K Nagata
- Department of Pathophysiological Laboratory Sciences, Nagoya, Japan
| |
Collapse
|
44
|
Mellor CL, Steinmetz FP, Cronin MTD. The identification of nuclear receptors associated with hepatic steatosis to develop and extend adverse outcome pathways. Crit Rev Toxicol 2015; 46:138-52. [PMID: 26451809 DOI: 10.3109/10408444.2015.1089471] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The development of adverse outcome pathways (AOPs) is becoming a key component of twenty-first century toxicology. AOPs provide a conceptual framework that links the molecular initiating event to an adverse outcome through organized toxicological knowledge, bridging the gap from chemistry to toxicological effect. As nuclear receptors (NRs) play essential roles for many physiological processes within the body, they are used regularly as drug targets for therapies to treat many diseases including diabetes, cancer and neurodegenerative diseases. Due to the heightened development of NR ligands, there is increased need for the identification of related AOPs to facilitate their risk assessment. Many NR ligands have been linked specifically to steatosis. This article reviews and summarizes the role of NR and their importance with links between NR examined to identify plausible putative AOPs. The following NRs are shown to induce hepatic steatosis upon ligand binding: aryl hydrocarbon receptor, constitutive androstane receptor, oestrogen receptor, glucocorticoid receptor, farnesoid X receptor, liver X receptor, peroxisome proliferator-activated receptor, pregnane X receptor and the retinoic acid receptor. A preliminary, putative AOP was formed for NR binding linked to hepatic steatosis as the adverse outcome.
Collapse
Affiliation(s)
- Claire L Mellor
- a School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , England
| | - Fabian P Steinmetz
- a School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , England
| | - Mark T D Cronin
- a School of Pharmacy and Biomolecular Sciences , Liverpool John Moores University , Liverpool , England
| |
Collapse
|
45
|
Li CC, Munitic I, Mittelstadt PR, Castro E, Ashwell JD. Suppression of Dendritic Cell-Derived IL-12 by Endogenous Glucocorticoids Is Protective in LPS-Induced Sepsis. PLoS Biol 2015; 13:e1002269. [PMID: 26440998 PMCID: PMC4595142 DOI: 10.1371/journal.pbio.1002269] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 08/28/2015] [Indexed: 01/16/2023] Open
Abstract
Sepsis, an exaggerated systemic inflammatory response, remains a major medical challenge. Both hyperinflammation and immunosuppression are implicated as causes of morbidity and mortality. Dendritic cell (DC) loss has been observed in septic patients and in experimental sepsis models, but the role of DCs in sepsis, and the mechanisms and significance of DC loss, are poorly understood. Here, we report that mice with selective deletion of the glucocorticoid receptor (GR) in DCs (GRCD11c-cre) were highly susceptible to LPS-induced septic shock, evidenced by elevated inflammatory cytokine production, hypothermia, and mortality. Neutralizing anti-IL-12 antibodies prevented hypothermia and death, demonstrating that endogenous GC-mediated suppression of IL-12 is protective. In LPS-challenged GRCD11c-cre mice, CD8+ DCs were identified as the major source of prolonged IL-12 production, which correlated with elevations of NK cell-derived IFN-γ. In addition, the loss of GR in CD11c+ cells rescued LPS-induced loss of CD8+ DCs but not other DC subsets. Unlike wild-type animals, exposure of GRCD11c-cre mice to low-dose LPS did not induce CD8+ DC loss or tolerance to subsequent challenge with high dose, but neutralization of IL-12 restored the ability of low-dose LPS to tolerize. Therefore, endogenous glucocorticoids blunt LPS-induced inflammation and promote tolerance by suppressing DC IL-12 production. Lipopolysaccharide (LPS) from bacteria causes the increased production of endogenous glucocorticoids, protecting mice from sepsis and contributing to LPS tolerance by suppressing production of interleukin-12 (IL-12) by dendritic cells and causing the death of the primary producers of IL-12. Read the Synopsis. Sepsis refers to life-threatening systemic inflammation, often caused by infection with bacteria that produce lipopolysaccharide (LPS). Glucocorticoids, immunosuppressive hormones produced by the adrenals, have been used to treat sepsis for over 50 y, but little is known about the role of endogenous (naturally occurring) glucocorticoids in systemic inflammation. Macrophages have been considered the primary source of inflammatory mediators (cytokines) and a target for glucocorticoid-mediated suppression. The possible role of another immune cell population, dendritic cells, has not been explored in detail. We created a mouse model in which the glucocorticoid receptor is selectively deleted in dendritic cells (DCs). We found that the elevation of glucocorticoids that accompanies sepsis protects mice from LPS-induced septic shock by suppressing DC production of IL-12, a cytokine that causes the secretion of other inflammatory mediators. In addition, LPS-induced glucocorticoids caused the death of a subset of DCs that are the primary producers of IL-12. Glucocorticoids were also found to be important for the phenomenon of "LPS tolerance", in which inoculation with low-dose LPS makes mice resistant to rechallenge with a high dose. This unexpected role of DC-produced IL-12 and its suppression by endogenous glucocorticoids may account, at least in part, for the known association of adrenal insufficiency and prolonged sepsis.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/therapeutic use
- CD11c Antigen/genetics
- CD11c Antigen/metabolism
- Cells, Cultured
- Crosses, Genetic
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/pathology
- Dose-Response Relationship, Drug
- Female
- Glucocorticoids/agonists
- Glucocorticoids/antagonists & inhibitors
- Glucocorticoids/blood
- Glucocorticoids/metabolism
- Immunity, Innate/drug effects
- Interleukin-12/antagonists & inhibitors
- Interleukin-12/blood
- Interleukin-12/metabolism
- Lipopolysaccharides/toxicity
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Glucocorticoid/agonists
- Receptors, Glucocorticoid/antagonists & inhibitors
- Receptors, Glucocorticoid/genetics
- Receptors, Glucocorticoid/metabolism
- Shock, Septic/immunology
- Shock, Septic/metabolism
- Shock, Septic/pathology
- Shock, Septic/prevention & control
- Signal Transduction/drug effects
- Specific Pathogen-Free Organisms
- Spleen/drug effects
- Spleen/immunology
- Spleen/metabolism
- Spleen/pathology
Collapse
Affiliation(s)
- Caiyi C. Li
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ivana Munitic
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Paul R. Mittelstadt
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ehydel Castro
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jonathan D. Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
46
|
Chen S, Qian J, Shi X, Gao T, Liang T, Liu C. Control of hepatic gluconeogenesis by the promyelocytic leukemia zinc finger protein. Mol Endocrinol 2015; 28:1987-98. [PMID: 25333514 DOI: 10.1210/me.2014-1164] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The promyelocytic leukemia zinc finger (PLZF) protein is involved in major biological processes including energy metabolism, although its role remains unknown. In this study, we demonstrated that hepatic PLZF expression was induced in fasted or diabetic mice. PLZF promoted gluconeogenic gene expression and hepatic glucose output, leading to hyperglycemia. In contrast, hepatic PLZF knockdown improved glucose homeostasis in db/db mice. Mechanistically, peroxisome proliferator-activated receptor γ coactivator 1α and the glucocorticoid receptor synergistically activated PLZF expression. We conclude that PLZF is a critical regulator of hepatic gluconeogenesis. PLZF manipulation may benefit the treatment of metabolic diseases associated with gluconeogenesis.
Collapse
Affiliation(s)
- Siyu Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology and College of Life Sciences (S.C., J.Q., X.S., T.G., T.L., C.L.), Nanjing Normal University, Nanjing, Jiangsu 210023, China; and State Key Laboratory of Natural Medicines (C.L.), China Pharmaceutical University, Nanjing, Jiangsu 210009, China
| | | | | | | | | | | |
Collapse
|
47
|
Okun JG, Conway S, Schmidt KV, Schumacher J, Wang X, de Guia R, Zota A, Klement J, Seibert O, Peters A, Maida A, Herzig S, Rose AJ. Molecular regulation of urea cycle function by the liver glucocorticoid receptor. Mol Metab 2015; 4:732-40. [PMID: 26500844 PMCID: PMC4588454 DOI: 10.1016/j.molmet.2015.07.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 07/20/2015] [Accepted: 07/23/2015] [Indexed: 11/21/2022] Open
Abstract
Objective One of the major side effects of glucocorticoid (GC) treatment is lean tissue wasting, indicating a prominent role in systemic amino acid metabolism. In order to uncover a novel aspect of GCs and their intracellular-receptor, the glucocorticoid receptor (GR), on metabolic control, we conducted amino acid and acylcarnitine profiling in human and mouse models of GC/GR gain- and loss-of-function. Methods Blood serum and tissue metabolite levels were determined in Human Addison's disease (AD) patients as well as in mouse models of systemic and liver-specific GR loss-of-function (AAV-miR-GR) with or without dexamethasone (DEX) treatments. Body composition and neuromuscular and metabolic function tests were conducted in vivo and ex vivo, the latter using precision cut liver slices. Results A serum metabolite signature of impaired urea cycle function (i.e. higher [ARG]:[ORN + CIT]) was observed in human (CTRL: 0.45 ± 0.03, AD: 1.29 ± 0.04; p < 0.001) and mouse (AAV-miR-NC: 0.97 ± 0.13, AAV-miR-GR: 2.20 ± 0.19; p < 0.001) GC/GR loss-of-function, with similar patterns also observed in liver. Serum urea levels were consistently affected by GC/GR gain- (∼+32%) and loss (∼−30%) -of-function. Combined liver-specific GR loss-of-function with DEX treatment revealed a tissue-autonomous role for the GR to coordinate an upregulation of liver urea production rate in vivo and ex vivo, and prevent hyperammonaemia and associated neuromuscular dysfunction in vivo. Liver mRNA expression profiling and GR-cistrome mining identified Arginase I (ARG1) a urea cycle gene targeted by the liver GR. Conclusions The liver GR controls systemic and liver urea cycle function by transcriptional regulation of ARG1 expression. Metabolite profiling revealed a role for the HPA-axis-liver GR in regulating urea cycle function in mouse and humans. The liver GR controls enhanced urea cycle function during chronic glucocorticoid exposure. Liver Arginase I is a key urea cycle transcript regulated by the GR.
Collapse
Affiliation(s)
- Jürgen G Okun
- Division of Neuropediatrics and Metabolic Medicine, University Children's Hospital, Heidelberg, Germany
| | - Sean Conway
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Kathrin V Schmidt
- Division of Neuropediatrics and Metabolic Medicine, University Children's Hospital, Heidelberg, Germany
| | - Jonas Schumacher
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Xiaoyue Wang
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Roldan de Guia
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Annika Zota
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany ; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, 85764 Neuherberg, Germany
| | - Johanna Klement
- Department of Internal Medicine, University of Lübeck, 23538 Lübeck, Germany
| | - Oksana Seibert
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Achim Peters
- Department of Internal Medicine, University of Lübeck, 23538 Lübeck, Germany
| | - Adriano Maida
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany ; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, 85764 Neuherberg, Germany
| | - Stephan Herzig
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany ; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, 85764 Neuherberg, Germany
| | - Adam J Rose
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| |
Collapse
|
48
|
Blockade of glucocorticoid receptors with RU486 attenuates cardiac damage and adipose tissue inflammation in a rat model of metabolic syndrome. Hypertens Res 2015; 38:741-50. [PMID: 26155752 DOI: 10.1038/hr.2015.77] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 05/07/2015] [Accepted: 06/04/2015] [Indexed: 12/16/2022]
Abstract
Glucocorticoids are stress hormones that modulate metabolic, inflammatory and cardiovascular processes. We recently characterized DahlS.Z-Lepr(fa)/Lepr(fa) (DS/obese) rats, derived from a cross between Dahl salt-sensitive (DS) and Zucker rats, as a new animal model of metabolic syndrome (MetS). We have now investigated the effects of glucocorticoid receptor (GR) blockade on cardiac and adipose tissue pathology and gene expression, as well as on glucose metabolism in this model. DS/obese rats were treated with the GR blocker RU486 (2 mg kg(-1) per day, subcutaneous) for 4 weeks beginning at 9 weeks of age. Age-matched homozygous lean (DahlS.Z-Lepr(+)/Lepr(+), or DS/lean) littermates of DS/obese rats served as controls. Treatment of DS/obese rats with RU486 attenuated left ventricular (LV) fibrosis and diastolic dysfunction, as well as cardiac oxidative stress and inflammation, without affecting hypertension or LV hypertrophy. Administration of RU486 to DS/obese rats also inhibited the upregulation of GR and 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) expression at the mRNA and protein levels in the heart; it attenuated adiposity and adipose tissue inflammation, as well as the upregulation of GR and 11β-HSD1 mRNA and protein expression in adipose tissue; it ameliorated fasting hyperinsulinemia as well as insulin resistance and glucose intolerance. Our results thus implicate the glucocorticoid-GR axis in the pathophysiology of MetS, and they suggest that GR blockade has therapeutic potential for the treatment of this condition.
Collapse
|
49
|
Matsuura N, Nagasawa K, Minagawa Y, Ito S, Sano Y, Yamada Y, Hattori T, Watanabe S, Murohara T, Nagata K. Restraint stress exacerbates cardiac and adipose tissue pathology via β-adrenergic signaling in rats with metabolic syndrome. Am J Physiol Heart Circ Physiol 2015; 308:H1275-86. [DOI: 10.1152/ajpheart.00906.2014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/10/2015] [Indexed: 11/22/2022]
Abstract
Restraint stress stimulates sympathetic nerve activity and can affect adiposity and metabolism. However, the effects of restraint stress on cardiovascular and metabolic disorders in metabolic syndrome (MetS) have remained unclear. We investigated the effects of chronic restraint stress and β-adrenergic receptor (β-AR) blockade on cardiac and adipose tissue pathology and metabolic disorders in a rat model of MetS. DahlS.Z- Leprfa/ Leprfa (DS/obese) rats, derived from a cross between Dahl salt-sensitive and Zucker rats. Rats were exposed to restraint stress (restraint cage, 2 h/day) for 4 wk from 9 wk of age with or without daily subcutaneous administration of the β-AR blocker propranolol (2 mg/kg). Age-matched homozygous lean littermates of DS/obese rats (DahlS.Z- Lepr+ /Lepr+ rats) served as control animals. Chronic restraint stress exacerbated hypertension as well as left ventricular hypertrophy, fibrosis, diastolic dysfunction, and oxidative stress in a manner sensitive to propranolol treatment. Restraint stress attenuated body weight gain in DS/obese rats, and this effect tended to be reversed by propranolol ( P = 0.0682). Restraint stress or propranolol did not affect visceral or subcutaneous fat mass. However, restraint stress potentiated cardiac and visceral adipose tissue inflammation in DS/obese rats, and these effects were ameliorated by propranolol. Restraint stress also exacerbated glucose intolerance, insulin resistance, and abnormal lipid metabolism in a manner sensitive to propranolol. In addition, restraint stress increased urinary norepinephrine excretion, and propranolol attenuated this effect. Our results thus implicate β-ARs in the exacerbation of cardiac and adipose tissue pathology and abnormal glucose and lipid metabolism induced by restraint stress in this model of MetS.
Collapse
Affiliation(s)
- Natsumi Matsuura
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kai Nagasawa
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuji Minagawa
- Department of Medical Technology, Nagoya University School of Health Sciences, Nagoya, Japan; and
| | - Shogo Ito
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yusuke Sano
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichiro Yamada
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takuya Hattori
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shogo Watanabe
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kohzo Nagata
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
50
|
Schäff C, Rohrbeck D, Steinhoff-Wagner J, Kanitz E, Sauerwein H, Bruckmaier R, Hammon H. Hepatic glucocorticoid and α1- and β2-adrenergic receptors in calves change during neonatal maturation and are related to energy regulation. J Dairy Sci 2015; 98:1046-56. [DOI: 10.3168/jds.2014-8636] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/07/2014] [Indexed: 11/19/2022]
|