1
|
Wen S, Lv X, Chu S, Wang R, Qin D. Associations between prenatal dioxin-like polychlorinated biphenyls exposure and glucocorticoid and androgenic hormones in umbilical cord blood. ENVIRONMENTAL RESEARCH 2025; 270:120927. [PMID: 39848526 DOI: 10.1016/j.envres.2025.120927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 01/25/2025]
Abstract
This study investigates the association between prenatal exposure to dioxin-like polychlorinated biphenyls (DL-PCBs) and glucocorticoid and androgenic hormone levels in cord blood. We analyzed cord blood samples from 500 mother-infant pairs from China (2022-2023), focusing on hormones including cortisol, cortisone, dehydroepiandrosterone (DHEA), and androstenedione. The main analysis revealed significant reductions in cortisol levels with increased exposure to PCB-77 (β = -3.37, 95% CI: -6.41, -0.32), PCB-81 (β = -2.90, 95% CI: -5.40, -0.39), and PCB-105 (β = -2.27, 95% CI: -4.45, -0.09). Cortisone levels also decreased with PCB-77 (β = -6.00, 95% CI: -10.98, -1.02), while DHEA increased with PCB-81 exposure (β = 0.19, 95% CI: 0.08, 0.31). Furthermore, the cortisol/DHEA ratio decreased significantly with PCB-77 (β = -2.45, 95% CI: -4.01, -0.88), indicating a disruption in the balance of glucocorticoid and androgenic hormones. Stratified analyses revealed significant sex-specific associations. Among boys, PCB189 was most strongly associated with reduced cortisol levels, while PCB169 exhibited the largest negative effect in girls. Mixture analysis using quantile g-computation demonstrated that each quartile increase in combined DL-PCB exposure was associated with a significant decrease in cortisol (β = -2.85, 95% CI: -4.73, -0.97) and cortisone (β = -4.78, 95% CI: -7.85, -1.71), alongside a significant increase in DHEA (β = 0.27, 95% CI: 0.11, 0.43) and androstenedione (β = 0.02, 95% 0.01, 0.04). The cortisol/DHEA and glucocorticoid/androgenic ratios also showed a significant reduction. Generalized weighted quantile sum (gWQS) analysis corroborated the negative associations for cortisol and cortisone but did not detect significant effects for DHEA and androstenedione. These results underscore the complex interactions of DL-PCB exposure with glucocorticoid and androgenic hormones, highlighting the importance of sex-specific and mixture-based approaches to understanding endocrine disruption in prenatal development.
Collapse
Affiliation(s)
- Shuang Wen
- Reproductive Center, Department of Obstetrics and Gynecology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Xuefeng Lv
- Department of Laboratory Medicine, The Third Affiliated Hospital of Zhengzhou University. Zhengzhou, Henan, China; Zhengzhou Key Laboratory for in Vitro Diagnosis of Hypertensive Disorders of Pregnancy. Zhengzhou, Henan, China
| | - Shuhui Chu
- Department of Obstetrics, The Third Affiliated Hospital of Zhengzhou University. Zhengzhou, Henan, China
| | - RuiXin Wang
- The Second Clinical College of Henan University of Traditional Chinese Medicine. Zhengzhou, Henan, China
| | - Dongchun Qin
- Department of Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University. Zhengzhou, Henan, China.
| |
Collapse
|
2
|
Yusuf JA, Akanbi ST, Olorunlowu DR, Opoola EK, Ogunlade EE, Kayode EA, Adejobi EO, Sulaiman YO, Odemakinde DI, Aworeni EO, Abdulmalik NI, Oluyemi DP, Isaac AE, Aromose OI, Adewale OM, Ogunrinde V, Adeleke TA, Adeleye OO. Molecular mechanism underlying stress response and adaptation. PROGRESS IN BRAIN RESEARCH 2025; 291:81-108. [PMID: 40222793 DOI: 10.1016/bs.pbr.2025.01.005] [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: 04/15/2025]
Abstract
Stress, a common life experience, impacts both mental and physical health, contributing to conditions such as anxiety and cardiovascular disease. It triggers physiological and psychological responses, primarily through the Hypothalamic-Pituitary-Adrenal (HPA) and Sympathetic-Adrenal-Medullary (SAM) axes, which are coordinated by the autonomic nervous system. Dysregulation of the glucocorticoid system, mediated by mineralocorticoid and glucocorticoid receptors, plays a critical role in neurodegenerative disorders like Alzheimer's disease. Cellular pathways like PI3K/Akt, NF-κB, and AP-1 transcription factors maintain homeostasis during stress and are targets for therapeutic research. Epigenetic influences and genomic modifications highlight the long-lasting effects of stress on gene expression. Adaptive responses, such as allostasis, allow the body to maintain stability amid stress. However, excessive stress leads to allostatic load, negatively impacting the immune, endocrine, and nervous systems. Current treatments include pharmacological and lifestyle interventions, with emerging approaches such as psychobiotics and precision medicine offering future potential.
Collapse
Affiliation(s)
- Joshua Ayodele Yusuf
- Molecular Biology and Biotechnology Division, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria; Neuroscience Unit, Department of Veterinary Anatomy, University of Ibadan, Ibadan, Oyo State, Nigeria.
| | - Stephen Tunmise Akanbi
- Central Research Laboratory, Ilorin, Kwara State, Nigeria; Gen'Omics Research Hub, Ogbomosho, Oyo State, Nigeria
| | - Darasimi Racheal Olorunlowu
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology (LAUTECH), Ogbomoso, Oyo State, Nigeria
| | - Elizabeth Kehinde Opoola
- Department of Anatomy, Faculty of Basic Medical Science, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Eniola Elizabeth Ogunlade
- Molecular Biology and Biotechnology Division, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Emmanuel Adebayo Kayode
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology (LAUTECH), Ogbomoso, Oyo State, Nigeria; LAUTECH Neuroscience Group (LNG), Oyo State, Nigeria
| | - Emmanuel Oluwagbenga Adejobi
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology (LAUTECH), Ogbomoso, Oyo State, Nigeria; LAUTECH Neuroscience Group (LNG), Oyo State, Nigeria
| | - Yasar Olalekan Sulaiman
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology (LAUTECH), Ogbomoso, Oyo State, Nigeria
| | - Dorcas Ifeoluwa Odemakinde
- Design and Development of Rapid Diagnostic Assay Division, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Esther Opeyemi Aworeni
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology (LAUTECH), Ogbomoso, Oyo State, Nigeria; Drosophila Research and Training Centre, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Nurat Ize Abdulmalik
- LAUTECH Neuroscience Group (LNG), Oyo State, Nigeria; Faculty of Nursing Sciences, Ladoke Akintola University of Technology (LAUTECH), Ogbomoso, Oyo State, Nigeria
| | - Dolapo Priscilla Oluyemi
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology (LAUTECH), Ogbomoso, Oyo State, Nigeria
| | - Ayomide Esther Isaac
- Neuroscience Unit, Department of Veterinary Anatomy, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Oluwaferanmi Israel Aromose
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology (LAUTECH), Ogbomoso, Oyo State, Nigeria; Genome Science Division, Helix Biogen Institute, Ogbomoso, Oyo State, Nigeria
| | - Oyindamola Munirat Adewale
- Humboldt Research Hub-Center for Emerging and Re-emerging Infectious Diseases (HRH-CERID), LAUTECH, Ogbomoso, Oyo State, Nigeria
| | - Victor Ogunrinde
- Drosophila Research and Training Centre, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Tijesunimi Ayomide Adeleke
- Molecular Biology and Biotechnology Division, Nigerian Institute of Medical Research, Yaba, Lagos, Nigeria
| | - Olufunto Omodele Adeleye
- Department of Anatomy, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology (LAUTECH), Ogbomoso, Oyo State, Nigeria; LAUTECH Neuroscience Group (LNG), Oyo State, Nigeria
| |
Collapse
|
3
|
Khan S, Livingstone DEW, Zielinska A, Doig CL, Cobice DF, Esteves CL, Man JTY, Homer NZM, Seckl JR, MacKay CL, Webster SP, Lavery GG, Chapman KE, Walker BR, Andrew R. Contribution of local regeneration of glucocorticoids to tissue steroid pools. J Endocrinol 2023; 258:e230034. [PMID: 37343234 PMCID: PMC10448579 DOI: 10.1530/joe-23-0034] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/20/2022] [Indexed: 06/23/2023]
Abstract
11β-Hydroxysteroid dehydrogenase 1 (11βHSD1) is a drug target to attenuate adverse effects of chronic glucocorticoid excess. It catalyses intracellular regeneration of active glucocorticoids in tissues including brain, liver and adipose tissue (coupled to hexose-6-phosphate dehydrogenase, H6PDH). 11βHSD1 activity in individual tissues is thought to contribute significantly to glucocorticoid levels at those sites, but its local contribution vs glucocorticoid delivery via the circulation is unknown. Here, we hypothesised that hepatic 11βHSD1 would contribute significantly to the circulating pool. This was studied in mice with Cre-mediated disruption of Hsd11b1 in liver (Alac-Cre) vs adipose tissue (aP2-Cre) or whole-body disruption of H6pdh. Regeneration of [9,12,12-2H3]-cortisol (d3F) from [9,12,12-2H3]-cortisone (d3E), measuring 11βHSD1 reductase activity was assessed at steady state following infusion of [9,11,12,12-2H4]-cortisol (d4F) in male mice. Concentrations of steroids in plasma and amounts in liver, adipose tissue and brain were measured using mass spectrometry interfaced with matrix-assisted laser desorption ionisation or liquid chromatography. Amounts of d3F were higher in liver, compared with brain and adipose tissue. Rates of appearance of d3F were ~6-fold slower in H6pdh-/- mice, showing the importance for whole-body 11βHSD1 reductase activity. Disruption of liver 11βHSD1 reduced the amounts of d3F in liver (by ~36%), without changes elsewhere. In contrast disruption of 11βHSD1 in adipose tissue reduced rates of appearance of circulating d3F (by ~67%) and also reduced regenerated of d3F in liver and brain (both by ~30%). Thus, the contribution of hepatic 11βHSD1 to circulating glucocorticoid levels and amounts in other tissues is less than that of adipose tissue.
Collapse
Affiliation(s)
- S Khan
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - D E W Livingstone
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Science, University of Edinburgh, Hugh Robson Building, Edinburgh, UK
| | - A Zielinska
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - C L Doig
- Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham, UK
| | - D F Cobice
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - C L Esteves
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - J T Y Man
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - N Z M Homer
- Mass Spectrometry Core Laboratory, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - J R Seckl
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - C L MacKay
- SIRCAMS, School of Chemistry, University of Edinburgh, Joseph Black Building, King's Buildings, Edinburgh, UK
| | - S P Webster
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - G G Lavery
- Department of Biosciences, School of Science & Technology, Nottingham Trent University, Nottingham, UK
| | - K E Chapman
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - B R Walker
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Clinical & Translational Research Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, UK
| | - R Andrew
- Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Mass Spectrometry Core Laboratory, Edinburgh Clinical Research Facility, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
Bini J, Parikh L, Lacadie C, Hwang JJ, Shah S, Rosenberg SB, Seo D, Lam K, Hamza M, De Aguiar RB, Constable T, Sherwin RS, Sinha R, Jastreboff AM. Stress-level glucocorticoids increase fasting hunger and decrease cerebral blood flow in regions regulating eating. Neuroimage Clin 2022; 36:103202. [PMID: 36126514 PMCID: PMC9486604 DOI: 10.1016/j.nicl.2022.103202] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/24/2022] [Accepted: 09/16/2022] [Indexed: 12/14/2022]
Abstract
CONTEXT The neural regulation of appetite and energy homeostasis significantly overlaps with the neurobiology of stress. Frequent exposure to repeated acute stressors may cause increased allostatic load and subsequent dysregulation of the cortico-limbic striatal system leading to inefficient integration of postprandial homeostatic and hedonic signals. It is therefore important to understand the neural mechanisms by which stress generates alterations in appetite that may drive weight gain. OBJECTIVE To determine glucocorticoid effects on metabolic, neural and behavioral factors that may underlie the association between glucocorticoids, appetite and obesity risk. METHODS A randomized double-blind cross-over design of overnight infusion of hydrocortisone or saline followed by a fasting morning perfusion magnetic resonance imaging to assess regional cerebral blood flow (CBF) was completed. Visual Analog Scale (VAS) hunger, cortisol and metabolic hormones were also measured. RESULTS Hydrocortisone relative to saline significantly decreased whole brain voxel based CBF responses in the hypothalamus and related cortico-striatal-limbic regions. Hydrocortisone significantly increased hunger VAS pre-scan, insulin, glucose and leptin, but not other metabolic hormones versus saline CBF groups. Hydrocortisone related increases in hunger were predicted by less reduction of CBF (hydrocortisone minus saline) in the medial OFC, medial brainstem and thalamus, left primary sensory cortex and right superior and medial temporal gyrus. Hunger ratings were also positively associated with plasma insulin on hydrocortisone but not saline day. CONCLUSIONS Increased glucocorticoids at levels akin to those experienced during psychological stress, result in increased fasting hunger and decreased regional cerebral blood flow in a distinct brain network of prefrontal, emotional, reward, motivation, sensory and homeostatic regions that underlie control of food intake.
Collapse
Affiliation(s)
- Jason Bini
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States
| | - Lisa Parikh
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Cheryl Lacadie
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States
| | - Janice J Hwang
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Saloni Shah
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Samuel B Rosenberg
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Dongju Seo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Katherine Lam
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Muhammad Hamza
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Renata Belfort De Aguiar
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Todd Constable
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States
| | - Robert S Sherwin
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Rajita Sinha
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States.
| | - Ania M Jastreboff
- Division of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States; Division of Pediatric Endocrinology, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
5
|
Fehsel K, Christl J. Comorbidity of osteoporosis and Alzheimer's disease: Is `AKT `-ing on cellular glucose uptake the missing link? Ageing Res Rev 2022; 76:101592. [PMID: 35192961 DOI: 10.1016/j.arr.2022.101592] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 02/08/2023]
Abstract
Osteoporosis and Alzheimer's disease (AD) are both degenerative diseases. Osteoporosis often proceeds cognitive deficits, and multiple studies have revealed common triggers that lead to energy deficits in brain and bone. Risk factors for osteoporosis and AD, such as obesity, type 2 diabetes, aging, chemotherapy, vitamin deficiency, alcohol abuse, and apolipoprotein Eε4 and/or Il-6 gene variants, reduce cellular glucose uptake, and protective factors, such as estrogen, insulin, exercise, mammalian target of rapamycin inhibitors, hydrogen sulfide, and most phytochemicals, increase uptake. Glucose uptake is a fine-tuned process that depends on an abundance of glucose transporters (Gluts) on the cell surface. Gluts are stored in vesicles under the plasma membrane, and protective factors cause these vesicles to fuse with the membrane, resulting in presentation of Gluts on the cell surface. This translocation depends mainly on AKT kinase signaling and can be affected by a range of factors. Reduced AKT kinase signaling results in intracellular glucose deprivation, which causes endoplasmic reticulum stress and iron depletion, leading to activation of HIF-1α, the transcription factor necessary for higher Glut expression. The link between diseases and aging is a topic of growing interest. Here, we show that diseases that affect the same biochemical pathways tend to co-occur, which may explain why osteoporosis and/or diabetes are often associated with AD.
Collapse
|
6
|
Sghaier R, Nury T, Leoni V, Caccia C, Pais De Barros JP, Cherif A, Vejux A, Moreau T, Limem K, Samadi M, Mackrill JJ, Masmoudi AS, Lizard G, Zarrouk A. Dimethyl fumarate and monomethyl fumarate attenuate oxidative stress and mitochondrial alterations leading to oxiapoptophagy in 158N murine oligodendrocytes treated with 7β-hydroxycholesterol. J Steroid Biochem Mol Biol 2019; 194:105432. [PMID: 31344443 DOI: 10.1016/j.jsbmb.2019.105432] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 07/01/2019] [Accepted: 07/19/2019] [Indexed: 01/08/2023]
Abstract
Oxidative stress and mitochondrial dysfunction contribute to the pathogenesis of neurodegenerative diseases and favor lipid peroxidation, leading to increased levels of 7β-hydroxycholesterol (7β-OHC) which induces oxiapoptophagy (OXIdative stress, APOPTOsis, autoPHAGY). The cytoprotective effects of dimethylfumarate (DMF), used in the treatment of relapsing remitting multiple sclerosis and of monomethylfumarate (MMF), its main metabolite, were evaluated on murine oligodendrocytes 158 N exposed to 7β-OHC (50 μM, 24 h) with or without DMF or MMF (25 μM). The activity of 7β-OHC in the presence or absence DMF or MMF was evaluated on several parameters: cell adhesion; plasma membrane integrity measured with propidium iodide (PI), trypan blue and fluoresceine diacetate (FDA) assays; LDH activity; antioxidant enzyme activities (superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx)); generation of lipid peroxidation products (malondialdehyde (MDA), conjugated dienes (CDs)) and protein oxidation products (carbonylated proteins (CPs)); reactive oxygen species (ROS) overproduction conducted with DHE and DHR123. The effect on mitochondria was determined with complementary criteria: measurement of succinate dehydrogenase activity, evaluation of mitochondrial potential (ΔΨm) and mitochondrial superoxide anions (O2●-) production using DiOC6(3) and MitoSOX, respectively; quantification of mitochondrial mass with Mitotracker Red, and of cardiolipins and organic acids. The effects on mitochondrial and peroxisomal ultrastructure were determined by transmission electron microscopy. Intracellular sterol and fatty acid profiles were determined. Apoptosis and autophagy were characterized by staining with Hoechst 33,342, Giemsa and acridine orange, and with antibodies raised against caspase-3 and LC3. DMF and MMF attenuate 7β-OHC-induced cytotoxicity: cell growth inhibition; decreased cell viability; mitochondrial dysfunction (decrease of succinate dehydrogenase activity, loss of ΔΨm, increase of mitochondrial O2●- production, alteration of the tricarboxilic acid (TCA) cycle, and cardiolipins content); oxidative stress induction (ROS overproduction, alteration of GPx, CAT, and SOD activities, increased levels of MDA, CDs, and CPs); changes in fatty acid and cholesterol metabolism; and cell death induction (caspase-3 cleavage, activation of LC3-I in LC3-II). Ultrastructural alterations of mitochondria and peroxisomes were prevented. These results demonstrate that DMF and MMF prevent major dysfunctions associated with neurodegenerative diseases: oxidative stress, mitochondrial dysfunction, apoptosis and autophagy.
Collapse
Affiliation(s)
- Randa Sghaier
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France; Univ. Sousse, Laboratory of Biochemistry, Faculty of Medicine, Tunisia; Univ. Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir; Univ. Manouba, Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources (LR11ES31), Higher Institute of Biotechnology, Sidi Thabet, Tunisia
| | - Thomas Nury
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Varese, ASST-Settelaghi, Varese, Italy
| | - Claudio Caccia
- Unit of Medical Genetics and Neurogenetics, IRCCS Carlo Besta, Milano, Italy
| | | | - Ameur Cherif
- Univ. Manouba, Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources (LR11ES31), Higher Institute of Biotechnology, Sidi Thabet, Tunisia
| | - Anne Vejux
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France
| | - Thibault Moreau
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France; Univ. Hospital, Department of Neurology, Dijon, France
| | - Khalifa Limem
- Univ. Sousse, Laboratory of Biochemistry, Faculty of Medicine, Tunisia
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Dept of Chemistry, Univ. Lorraine, Metz Technopôle, Metz, France
| | - John J Mackrill
- Department of Physiology, Biosciences Institute, University College Cork, Cork, Ireland
| | - Ahmed Slaheddine Masmoudi
- Univ. Manouba, Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources (LR11ES31), Higher Institute of Biotechnology, Sidi Thabet, Tunisia
| | - Gérard Lizard
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France.
| | - Amira Zarrouk
- Univ. Sousse, Laboratory of Biochemistry, Faculty of Medicine, Tunisia; Univ. Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir.
| |
Collapse
|
7
|
Hill LA, Vassiliadi DA, Dimopoulou I, Anderson AJ, Boyle LD, Kilgour AHM, Stimson RH, Machado Y, Overall CM, Walker BR, Lewis JG, Hammond GL. Neutrophil elastase-cleaved corticosteroid-binding globulin is absent in human plasma. J Endocrinol 2019; 240:27-39. [PMID: 30452386 PMCID: PMC6347282 DOI: 10.1530/joe-18-0479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 09/28/2018] [Indexed: 12/24/2022]
Abstract
Corticosteroid-binding globulin (CBG) transports glucocorticoids in blood and is a serine protease inhibitor family member. Human CBG has a reactive center loop (RCL) which, when cleaved by neutrophil elastase (NE), disrupts its steroid-binding activity. Measurements of CBG levels are typically based on steroid-binding capacity or immunoassays. Discrepancies in ELISAs using monoclonal antibodies that discriminate between intact vs RCL-cleaved CBG have been interpreted as evidence that CBG with a cleaved RCL and low affinity for cortisol exists in the circulation. We examined the biochemical properties of plasma CBG in samples with discordant ELISA measurements and sought to identify RCL-cleaved CBG in human blood samples. Plasma CBG-binding capacity and ELISA values were consistent in arterial and venous blood draining skeletal muscle, liver and brain, as well as from a tissue (adipose) expected to contain activated neutrophils in obese individuals. Moreover, RCL-cleaved CBG was undetectable in plasma from critically ill patients, irrespective of whether their ELISA measurements were concordant or discordant. We found no evidence of RCL-cleaved CBG in plasma using a heat-dependent polymerization assay, and CBG that resists immunoprecipitation with a monoclonal antibody designed to specifically recognize an intact RCL, bound steroids with a high affinity. In addition, mass spectrometry confirmed the absence of NE-cleaved CBG in plasma in which ELISA values were highly discordant. Human CBG with a NE-cleaved RCL and low affinity for steroids is absent in blood samples, and CBG ELISA discrepancies likely reflect structural differences that alter epitopes recognized by specific monoclonal antibodies.
Collapse
Affiliation(s)
- Lesley A Hill
- Departments of Cellular and Physiological Sciences and Obstetrics and Gynaecology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Dimitra A Vassiliadi
- Endocrine Unit, Second Department of Internal Medicine-Research Institute and Diabetes Center, Attiko University Hospital, Athens, Greece
| | - Ioanna Dimopoulou
- Endocrine Unit, Second Department of Internal Medicine-Research Institute and Diabetes Center, Attiko University Hospital, Athens, Greece
| | - Anna J Anderson
- BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Luke D Boyle
- BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Alixe H M Kilgour
- BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Roland H Stimson
- BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Yoan Machado
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Christopher M Overall
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian R Walker
- BHF Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John G Lewis
- Canterbury Health Laboratories, Christchurch, New Zealand
| | - Geoffrey L Hammond
- Departments of Cellular and Physiological Sciences and Obstetrics and Gynaecology, The University of British Columbia, Vancouver, British Columbia, Canada
- Correspondence should be addressed to G L Hammond:
| |
Collapse
|
8
|
Quantification of 11β-hydroxysteroid dehydrogenase 1 kinetics and pharmacodynamic effects of inhibitors in brain using mass spectrometry imaging and stable-isotope tracers in mice. Biochem Pharmacol 2017; 148:88-99. [PMID: 29248595 PMCID: PMC5821700 DOI: 10.1016/j.bcp.2017.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022]
Abstract
11β-Hydroxysteroid dehydrogenase 1 (11β-HSD1; EC 1.1.1.146) generates active glucocorticoid hormones. Small molecule inhibitors have been developed to target 11β-HSD1 for the treatment of dementia; these must enter brain subregions, such as the hippocampus, to be effective. We previously reported mass spectrometry imaging measurement of murine tissue steroids, and deuterated steroid tracer infusion quantification of 11β-HSD1 turnover in humans. Here, these tools are combined to assess tissue pharmacokinetics and pharmacodynamics of an 11β-HSD1 inhibitor that accesses the brain. [9,11,12,12-2H]4-Cortisol was infused (1.75 mg/day) by minipump for 2 days into C57Bl6 mice (male, age 12 weeks, n = 3/group) after which an 11β-HSD1 inhibitor (UE2316) was administered (25 mg/kg oral gavage) and animals culled immediately or 1, 2 and 4 h post-dosing. Mice with global genetic disruption of Hsd11B1 were studied similarly. Turnover of d4-cortisol to d3-cortisone (by loss of the 11-deuterium) and regeneration of d3-cortisol (by 11β-HSD1-mediated reduction) were assessed in plasma, liver and brain using matrix assisted laser desorption ionization coupled to Fourier transform cyclotron resonance mass spectrometry. The tracer d4-cortisol was detected in liver and brain following a two day infusion. Turnover to d3-cortisone and on to d3-cortisol was slower in brain than liver. In contrast, d3-cortisol was not detected in mice lacking 11β-HSD1. UE2316 impaired d3-cortisol generation measured in whole body (assessed in plasma; 53.1% suppression in rate of appearance in d3-cortisol), liver and brain. Differential inhibition in brain regions was observed; active glucocorticoids were suppressed to a greater in extent hippocampus or cortex than in amygdala. These data confirm that the contribution of 11β-HSD1 to the tissue glucocorticoid pool, and the consequences of enzyme inhibition on active glucocorticoid concentrations, are substantial, including in the brain. They further demonstrate the value of mass spectrometry imaging in pharmacokinetic and pharmacodynamic studies.
Collapse
|
9
|
Abstract
The metabolic syndrome describes a clustering of risk factors—visceral obesity, dyslipidaemia, insulin resistance, and salt-sensitive hypertension—that increases mortality related to cardiovascular disease, type 2 diabetes, cancer, and non-alcoholic fatty liver disease. The prevalence of these concurrent comorbidities is ~ 25–30% worldwide, and metabolic syndrome therefore presents a significant global public health burden. Evidence from clinical and preclinical studies indicates that glucocorticoid excess is a key causal feature of metabolic syndrome. This is not increased systemic in circulating cortisol, rather increased bioavailability of active glucocorticoids within tissues. This review examines the role of covert glucocorticoid excess on the hypertension of the metabolic syndrome. Here, the role of the 11β-hydroxysteroid dehydrogenase enzymes, which exert intracrine and paracrine control over glucocorticoid signalling, is examined. 11βHSD1 amplifies glucocorticoid action in cells and contributes to hypertension through direct and indirect effects on the kidney and vasculature. The deactivation of glucocorticoid by 11βHSD2 controls ligand access to glucocorticoid and mineralocorticoid receptors: loss of function promotes salt retention and hypertension. As for hypertension in general, high blood pressure in the metabolic syndrome reflects a complex interaction between multiple systems. The clear association between high dietary salt, glucocorticoid production, and metabolic disorders has major relevance for human health and warrants systematic evaluation.
Collapse
Affiliation(s)
- Matthew A Bailey
- The British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
10
|
Travers S, Martinerie L, Bouvattier C, Boileau P, Lombès M, Pussard E. Multiplexed steroid profiling of gluco- and mineralocorticoids pathways using a liquid chromatography tandem mass spectrometry method. J Steroid Biochem Mol Biol 2017; 165:202-211. [PMID: 27339652 DOI: 10.1016/j.jsbmb.2016.06.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/28/2016] [Accepted: 06/17/2016] [Indexed: 11/24/2022]
Abstract
Serum steroid assays are major tools in the clinical evaluation of adrenal disorders. The main adrenal steroids are routinely measured with immunoassays. However, chromatographic methods are known to offer better specificity. We report a liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay for simultaneous quantification of 15 adrenal steroids targeting the mineralo- and gluco-corticosteroid pathways. Serum steroids combined with deuterated internal standards were extracted using successive protein precipitation and solid phase extraction steps. Cortisol, cortisone, 11-deoxycortisol, 17-hydroxyprogesterone, 21-deoxycortisol, progesterone, 11-deoxycorticosterone, corticosterone, 11-dehydrocorticosterone, 18-hydroxycorticosterone, 18-hydroxy-11-deoxycorticosterone, aldosterone, dehydroepiandrosterone sulfate, testosterone and androstenedione were resolved in fourteen minutes using a BEH C18 column coupled to a methanol-ammonium formate gradient. Detection was performed using multiple reaction monitoring quantitation. Routinely determined steroid levels by immunoassays were compared to those measured by LC-MS/MS. This method was applied to assess steroid profiles in congenital adrenal hyperplasia (CAH) patients with 21-hydroxylase deficiency. Low quantification limits depending on each steroid (ranging from 0.015ng/mL for aldosterone to 20ng/mL for DHEAS) are adapted to the clinical use. Recoveries of steroids range from 64% for 21-deoxycortisol to 101% for cortisol and are fully corrected by internal standards. A good linearity with R>0.989 is obtained for each compound. The inter-day variation coefficients ranged from 4.7% for cortisol to 16.3% for 11-deoxycorticosterone. The immunoassay for cortisol (Immulite 2000, Siemens) showed acceptable agreement with LC-MS/MS (bias +7.2%). However, Bland-Altman plots revealed large negative bias for aldosterone (-33.4%, AldoCT, CisBio international), for 17-hydroxyprogesterone at concentrations below 2ng/mL (-74.1%, OHP-CT MP Biomedical), for androstenedione (-80.3%, RIA D4, Beckman Coulter) and for 11-deoxycortisol (-125.3%, Diasource Immunoassays). Finally, the analysis of samples from 21-hydroxylase defective patients demonstrated the potential usefulness of multiplexed steroid profiling for the diagnosis and/or monitoring of different forms of congenital adrenal hyperplasia. This LC-MS/MS method provides highly sensitive and specific assessments of mineralo- and glucocorticoids pathways from a small volume sample and is therefore a promising potent tool for clinical and experimental endocrine studies.
Collapse
Affiliation(s)
- Simon Travers
- Inserm, U1185, Le Kremlin-Bicêtre, F-94276, France; Fac Med Paris-Sud, Univ. Paris-Sud, Université Paris Saclay, UMR-S 1185, Le Kremlin-Bicêtre, F-94276, France; Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital de Bicêtre, Hôpitaux Universitaires Paris Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, F-94275, France
| | - Laetitia Martinerie
- Inserm, U1185, Le Kremlin-Bicêtre, F-94276, France; Service d'Endocrinologie Pédiatrique, Hôpital Robert Debré, Assistance Publique Hôpitaux de Paris, Paris, F-75019, France; PremUp Foundation, Paris, F-75005, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, F-75019, France
| | - Claire Bouvattier
- Fac Med Paris-Sud, Univ. Paris-Sud, Université Paris Saclay, UMR-S 1185, Le Kremlin-Bicêtre, F-94276, France; Département d'Endocrinologie Pédiatrique, Hôpital de Bicêtre, Hôpitaux Universitaires Paris Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, F-94275, France; Centre de référence des maladies rares du développement sexuel, Le Kremlin Bicêtre, F-94275, France
| | - Pascal Boileau
- PremUp Foundation, Paris, F-75005, France; Service de Réanimation Néonatale, CH Poissy St-Germain en-Laye, Poissy, F-78303, France; EA 7285, UFR des Sciences de la Santé, Simone Veil. Université Versailles St-Quentin en Yvelines, Montigny le Bretonneux F-78180, France
| | - Marc Lombès
- Inserm, U1185, Le Kremlin-Bicêtre, F-94276, France; Fac Med Paris-Sud, Univ. Paris-Sud, Université Paris Saclay, UMR-S 1185, Le Kremlin-Bicêtre, F-94276, France; PremUp Foundation, Paris, F-75005, France; Service d'Endocrinologie et Maladies de la Reproduction, Hôpital de Bicêtre, Hôpitaux Universitaires Paris Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, F-94275, France; UMS 32, Institut Biomédical de Bicêtre, Le Kremlin-Bicêtre F-94276, France
| | - Eric Pussard
- Inserm, U1185, Le Kremlin-Bicêtre, F-94276, France; Fac Med Paris-Sud, Univ. Paris-Sud, Université Paris Saclay, UMR-S 1185, Le Kremlin-Bicêtre, F-94276, France; Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital de Bicêtre, Hôpitaux Universitaires Paris Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, F-94275, France; UMS 32, Institut Biomédical de Bicêtre, Le Kremlin-Bicêtre F-94276, France.
| |
Collapse
|
11
|
Mass spectrometry and its evolving role in assessing tissue specific steroid metabolism. Biochem Soc Trans 2016; 44:645-51. [PMID: 27068983 DOI: 10.1042/bst20150234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Indexed: 12/21/2022]
Abstract
Glucocorticoid hormones play vital roles in regulating diverse biological processes in health and disease. Tissue levels are regulated by enzymes which activate and inactivate hormones. The enzyme, 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1), in particular, has been identified as a potential drug target; inhibiting this enzyme attenuates glucocorticoid action by lowering local levels of active hormone. A variety of mass spectrometric approaches have been developed to characterize this enzymein vivo Endogenous glucocorticoids and their metabolites can be profiled in urine by GC-MS and circulating steroids are now more commonly quantified by liquid chromatography tandem mass spectrometry. Tracer dilution studies have allowed rates of generation of glucocorticoids by the enzyme to be distinguished from hormone generated directly by the adrenal glands and, in combination with arterio-venous (AV) sampling, rates of production by specific tissues have been quantified. This has allowed the contribution of liver, adipose, muscle and brain to cortisol production in metabolic disease and hence prioritized drug targets. Most recently MS imaging in combination with on-tissue derivatization has been developed to profile oxo-steroids in tissue sections, allowing molecular maps to be generated across complex tissues, where regional functions are important. The review provides a synopsis of how measurement of steroids by MS has evolved with technological advances and this has provided insight into the dynamic turnover of glucocorticoidsin vivo, highlighting the milestones that have advanced the field and identifying the remaining challenges for researchers, in terms of analytical chemistry and endocrine physiology and biochemistry.
Collapse
|
12
|
Werumeus Buning J, van Faassen M, Brummelman P, Dullaart RPF, van den Berg G, van der Klauw MM, Kerstens MN, Stegeman CA, Muller Kobold AC, Kema IP, Wolffenbuttel BHR, van Beek AP. Effects of Hydrocortisone on the Regulation of Blood Pressure: Results From a Randomized Controlled Trial. J Clin Endocrinol Metab 2016; 101:3691-3699. [PMID: 27490921 DOI: 10.1210/jc.2016-2216] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CONTEXT Cardiovascular risk is increased in patients with secondary adrenal insufficiency, which may be ascribed to an unfavorable metabolic profile consequent to a relatively high hydrocortisone replacement dose. OBJECTIVE We determined the effects of a higher versus a lower glucocorticoid replacement dose on blood pressure (BP), the renin-angiotensin-aldosterone system, 11β-hydroxysteroid dehydrogenase enzyme activity and circulating (nor)metanephrines. DESIGN, SETTING, AND PATIENTS Forty-seven patients with secondary adrenal insufficiency from the University Medical Center Groningen participated in this randomized double-blind crossover study. INTERVENTIONS Patients randomly received 0.2-0.3 mg hydrocortisone/kg body weight followed by 0.4-0.6 mg hydrocortisone/kg body weight, or vice versa, each during 10 weeks. MAIN OUTCOME MEASURE(S) BP and regulating hormones were measured. RESULTS The higher hydrocortisone dose resulted in an increase in systolic BP of 5 (12) mm Hg (P = .011), diastolic BP of 2 (9) mm Hg (P = .050), and a median [interquartile range] drop in plasma potassium of -0.1 [-0.3; 0.1] nmol/liter (P = .048). The higher hydrocortisone dose led to decreases in serum aldosterone of -28 [-101; 9] pmol/liter (P = .020) and plasma renin of -1.3 [-4.5; 1.2 ] pg/mL (P = .051), and increased the ratio of plasma and urinary cortisol to cortisone (including their metabolites) (P < .001 for all). Furthermore, on the higher dose, plasma and urinary normetanephrine decreased by -0.101 [-0.242; 0.029] nmol/liter (P < .001) and -1.48 [-4.06; 0.29] μmol/mol creatinine (P < .001) respectively. CONCLUSIONS A higher dose of hydrocortisone increased systolic and diastolic BP and was accompanied by changes in the renin-angiotensin-aldosterone system, 11β-hydroxysteroid dehydrogenase enzyme activity, and circulating normetanephrine. This demonstrates that hydrocortisone dose even within the physiological range affects several pathways involved in BP regulation.
Collapse
Affiliation(s)
- Jorien Werumeus Buning
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martijn van Faassen
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Pauline Brummelman
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerrit van den Berg
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Melanie M van der Klauw
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Michiel N Kerstens
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Coen A Stegeman
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anneke C Muller Kobold
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ido P Kema
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - André P van Beek
- Department of Endocrinology (J.W.B., P.B., R.P.F.D., G.v.d.B., M.M.v.d.K., M.N.K., A.P.v.B.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Laboratory Medicine (M.v.F., A.C.M.K., I.P.K.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Nephrology (C.A.S.), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Lemche E, Chaban OS, Lemche AV. Neuroendocrinological and Epigenetic Mechanisms Subserving Autonomic Imbalance and HPA Dysfunction in the Metabolic Syndrome. Front Neurosci 2016; 10:142. [PMID: 27147943 PMCID: PMC4830841 DOI: 10.3389/fnins.2016.00142] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/21/2016] [Indexed: 12/18/2022] Open
Abstract
Impact of environmental stress upon pathophysiology of the metabolic syndrome (MetS) has been substantiated by epidemiological, psychophysiological, and endocrinological studies. This review discusses recent advances in the understanding of causative roles of nutritional factors, sympathomedullo-adrenal (SMA) and hypothalamic-pituitary adrenocortical (HPA) axes, and adipose tissue chronic low-grade inflammation processes in MetS. Disturbances in the neuroendocrine systems for leptin, melanocortin, and neuropeptide Y (NPY)/agouti-related protein systems have been found resulting directly in MetS-like conditions. The review identifies candidate risk genes from factors shown critical for the functioning of each of these neuroendocrine signaling cascades. In its meta-analytic part, recent studies in epigenetic modification (histone methylation, acetylation, phosphorylation, ubiquitination) and posttranscriptional gene regulation by microRNAs are evaluated. Several studies suggest modification mechanisms of early life stress (ELS) and diet-induced obesity (DIO) programming in the hypothalamic regions with populations of POMC-expressing neurons. Epigenetic modifications were found in cortisol (here HSD11B1 expression), melanocortin, leptin, NPY, and adiponectin genes. With respect to adiposity genes, epigenetic modifications were documented for fat mass gene cluster APOA1/C3/A4/A5, and the lipolysis gene LIPE. With regard to inflammatory, immune and subcellular metabolism, PPARG, NKBF1, TNFA, TCF7C2, and those genes expressing cytochrome P450 family enzymes involved in steroidogenesis and in hepatic lipoproteins were documented for epigenetic modifications.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London London, UK
| | - Oleg S Chaban
- Section of Psychosomatic Medicine, Bogomolets National Medical University Kiev, Ukraine
| | - Alexandra V Lemche
- Department of Medical Science, Institute of Clinical Research Berlin, Germany
| |
Collapse
|
14
|
Skarlandtová H, Bičíková M, Neužil P, Mlček M, Hrachovina V, Svoboda T, Medová E, Kudlička J, Dohnalová A, Havránek Š, Kazihnítková H, Máčová L, Vařejková E, Kittnar O. The Cortisol to Cortisone Ratio during Cardiac Catheterisation in Sows. Prague Med Rep 2015; 116:279-89. [DOI: 10.14712/23362936.2015.67] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
A possible effect of mini-invasive heart intervention on a response of hypothalamo-pituitary-adrenal stress axis and conversion of cortisone to cortisol were studied. We have analysed two stress markers levels (cortisol, cortisone) and cortisol/cortisone ratio in 25 sows using minimally invasive heart catheterisation as the stress factor. The values of studied parameters were assessed in four periods of the experiment: (1) the baseline level on the day before intervention, (2) after the introduction of anaesthesia, (3) after conducting tissue stimulation or ablation, and (4) after the end of the catheterisation. For statistical analyses we used the non-parametric Friedman test for four dependent samples (including all four stages of the operation) or three dependent samples (influence of operation only, baseline level was excluded). Statistically significant differences in both Friedman tests were found for cortisol and for cortisone. We have found the highest level of cortisol/cortisone ratio in unstressed conditions, then it decreased to the minimal level at the end of the intervention. We have concluded that cortisol levels are blunted by the influence of anaesthesia after its administration, and therefore decrease back to the baseline at the end of the operation.
Collapse
|