51
|
Russell GM, Lightman SL. Can side effects of steroid treatments be minimized by the temporal aspects of delivery method? Expert Opin Drug Saf 2014; 13:1501-13. [DOI: 10.1517/14740338.2014.965141] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
52
|
Russell GM, Durant C, Ataya A, Papastathi C, Bhake R, Woltersdorf W, Lightman S. Subcutaneous pulsatile glucocorticoid replacement therapy. Clin Endocrinol (Oxf) 2014; 81:289-93. [PMID: 24735400 PMCID: PMC4231230 DOI: 10.1111/cen.12470] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 04/05/2014] [Accepted: 04/11/2014] [Indexed: 11/29/2022]
Abstract
The glucocorticoid hormone cortisol is released in pulses resulting in a complex and dynamic ultradian rhythm of plasma cortisol that underlies the classical circadian rhythm. These oscillating levels are also seen at the level of tissues such as the brain and trigger pulses of gene activation and downstream signalling. Different patterns of glucocorticoid presentation (constant vs pulsatile) result not only in different patterns of gene regulation but also in different neuroendocrine and behavioural responses. Current 'optimal' glucocorticoid replacement therapy results in smooth hormone blood levels and does not replicate physiological pulsatile cortisol secretion. Validation of a novel portable pulsatile continuous subcutaneous delivery system in healthy volunteers under dexamethasone and metyrapone suppression. Pulsatile subcutaneous hydrocortisone more closely replicates physiological circadian and ultradian rhythmicity.
Collapse
Affiliation(s)
- Georgina M Russell
- Henry Wellcome Laboratories for Integrative Neurosciences and Endocrinology, Dorothy Hodgkin Building, University of BristolBristol, UK
| | - Claire Durant
- Henry Wellcome Laboratories for Integrative Neurosciences and Endocrinology, Dorothy Hodgkin Building, University of BristolBristol, UK
| | - Alia Ataya
- School of experimental psychology, University of BristolBristol, UK
| | - Chrysoula Papastathi
- Henry Wellcome Laboratories for Integrative Neurosciences and Endocrinology, Dorothy Hodgkin Building, University of BristolBristol, UK
| | - Ragini Bhake
- Henry Wellcome Laboratories for Integrative Neurosciences and Endocrinology, Dorothy Hodgkin Building, University of BristolBristol, UK
| | | | - Stafford Lightman
- Henry Wellcome Laboratories for Integrative Neurosciences and Endocrinology, Dorothy Hodgkin Building, University of BristolBristol, UK
| |
Collapse
|
53
|
Panagiotakopoulos L, Neigh GN. Development of the HPA axis: where and when do sex differences manifest? Front Neuroendocrinol 2014; 35:285-302. [PMID: 24631756 DOI: 10.1016/j.yfrne.2014.03.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 02/22/2014] [Accepted: 03/04/2014] [Indexed: 01/21/2023]
Abstract
Sex differences in the response to stress contribute to sex differences in somatic, neurological, and psychiatric diseases. Despite a growing literature on the mechanisms that mediate sex differences in the stress response, the ontogeny of these differences has not been comprehensively reviewed. This review focuses on the development of the hypothalamic-pituitary-adrenal (HPA) axis, a key component of the body's response to stress, and examines the critical points of divergence during development between males and females. Insight gained from animal models and clinical studies are presented to fully illustrate the current state of knowledge regarding sex differences in response to stress over development. An appreciation for the developmental timelines of the components of the HPA axis will provide a foundation for future areas of study by highlighting both what is known and calling attention to areas in which sex differences in the development of the HPA axis have been understudied.
Collapse
Affiliation(s)
| | - Gretchen N Neigh
- Emory University, Department of Physiology, United States; Emory University, Department of Psychiatry & Behavioral Sciences, United States.
| |
Collapse
|
54
|
Abstract
Corticosteroids secreted as end product of the hypothalamic-pituitary-adrenal axis act like a double-edged sword in the brain. The hormones coordinate appraisal processes and decision making during the initial phase of a stressful experience and promote subsequently cognitive performance underlying the management of stress adaptation. This action exerted by the steroids on the initiation and termination of the stress response is mediated by 2 related receptor systems: mineralocorticoid receptors (MRs) and glucocorticoid receptors (GRs). The receptor types are unevenly distributed but colocalized in abundance in neurons of the limbic brain to enable these complementary hormone actions. This contribution starts from a historical perspective with the observation that phasic occupancy of GR during ultradian rhythmicity is needed to maintain responsiveness to corticosteroids. Then, during stress, initially MR activation enhances excitability of limbic networks that are engaged in appraisal and emotion regulation. Next, the rising hormone concentration occupies GR, resulting in reallocation of energy to limbic-cortical circuits with a role in behavioral adaptation and memory storage. Upon MR:GR imbalance, dysregulation of the hypothalamic-pituitary-adrenal axis occurs, which can enhance an individual's vulnerability. Imbalance is characteristic for chronic stress experience and depression but also occurs during exposure to synthetic glucocorticoids. Hence, glucocorticoid psychopathology may develop in susceptible individuals because of suppression of ultradian/circadian rhythmicity and depletion of endogenous corticosterone from brain MR. This knowledge generated from testing the balance hypothesis can be translated to a rational glucocorticoid therapy.
Collapse
Affiliation(s)
- E Ron de Kloet
- Department of Medical Pharmacology, Leiden Academic Centre for Drug Research, Leiden University and Department of Endocrinology and Metabolism, Leiden University Medical Center, 2300 RA Leiden, The Netherlands
| |
Collapse
|
55
|
Lightman S, Terry JR. The importance of dynamic signalling for endocrine regulation and drug development: relevance for glucocorticoid hormones. Lancet Diabetes Endocrinol 2014; 2:593-9. [PMID: 24731665 DOI: 10.1016/s2213-8587(13)70182-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Glucocorticoid hormones are heavily prescribed for several indications, including hormone replacement, anti-inflammatory effects, and antineoplastic effects. The pharmaceutical industry has put much effort into the development of novel potent glucocorticoid agonists, whereas there has been little enthusiasm for development of temporal aspects of glucocorticoid drugs. Glucocorticoids are normally secreted in a highly dynamic fashion, not only in the well known 24 h circadian rhythm, but also in an approximately hourly ultradian rhythm. These rhythms are crucial for normal gene regulation and for optimum cognitive function. In this Personal View, we discuss how understanding normal oscillatory patterns of glucocorticoid secretion could help investigators to develop novel glucocorticoid therapeutics that maximise the beneficial effect and diminish unwanted side-effects.
Collapse
Affiliation(s)
- Stafford Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK.
| | - John R Terry
- College of Engineering, Mathematics, and Physical Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
56
|
|
57
|
Ryan CJ, Peng W, Kheoh T, Welkowsky E, Haqq CM, Chandler DW, Scher HI, Molina A. Androgen dynamics and serum PSA in patients treated with abiraterone acetate. Prostate Cancer Prostatic Dis 2014; 17:192-8. [PMID: 24637537 PMCID: PMC4020277 DOI: 10.1038/pcan.2014.8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/09/2014] [Accepted: 01/26/2014] [Indexed: 11/19/2022]
Abstract
Background: We analyzed the potential of abiraterone acetate (henceforth abiraterone) to reduce androgen levels below lower limits of quantification (LLOQ) and explored the association with changes in PSA decline in metastatic castration-resistant prostate cancer (mCRPC) patients. Methods: COU-AA-301 is a 2:1 randomized, double-blind, placebo-controlled study comparing abiraterone (1000 mg q.d.) plus low-dose prednisone (5 mg b.i.d.) with placebo plus prednisone in mCRPC patients post docetaxel. Serum testosterone, androstenedione and dehydroepiandrosterone sulfate from baseline to week 12 were measured by novel ultrasensitive two-dimensional liquid chromatography coupled to tandem mass spectrometry assays in a subset of subjects in each arm (abiraterone plus prednisone, n=80; prednisone, n=38). The association between PSA response (⩽50% baseline) and undetectable androgens (week 12 androgen level below LLOQ) was analyzed using logistic regression. Results: A significantly greater reduction in serum androgens was observed with abiraterone plus prednisone versus prednisone (all P⩽0.0003), reaching undetectable levels for testosterone (47.2% versus 0%, respectively). A positive association was observed between achieving undetectable serum androgens and PSA decline (testosterone: odds ratio=1.54; 95% confidence interval: 0.546–4.347). Reduction of androgens to undetectable levels did not occur in all patients achieving a PSA response, and a PSA response did not occur in all patients achieving undetectable androgen levels. Conclusions: Abiraterone plus prednisone significantly reduced serum androgens, as measured by ultrasensitive assays and was generally associated with PSA response. However, androgen decline did not uniformly predict PSA decline suggesting ligand-independent or other mechanisms for mCRPC progression.
Collapse
Affiliation(s)
- C J Ryan
- University of California San Francisco, San Francisco, CA, USA
| | - W Peng
- Janssen Research & Development, Los Angeles, CA, USA
| | - T Kheoh
- Janssen Research & Development, Los Angeles, CA, USA
| | - E Welkowsky
- Janssen Research & Development, Los Angeles, CA, USA
| | - C M Haqq
- Janssen Research & Development, Los Angeles, CA, USA
| | | | - H I Scher
- Memorial Sloan Kettering Cancer Center and Weill-Cornell Medical College, New York, NY, USA
| | - A Molina
- Janssen Research & Development, Menlo Park, CA, USA
| |
Collapse
|
58
|
Impact of stress on prefrontal glutamatergic, monoaminergic and cannabinoid systems. Curr Top Behav Neurosci 2014; 18:45-66. [PMID: 25048388 DOI: 10.1007/7854_2014_331] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Stress has been shown to have marked and divergent effects on learning and memory which involves specific brain regions, such as spatial and declarative memory involving the hippocampus, memory of emotional arousing experiences and fear involving the amygdala, and executive functions and fear extinction involving the prefrontal cortex or the PFC. Response to stress involves a coordinated activation of a constellation of physiological systems including the activation of the hypothalamic-pituitary-adrenal (HPA) axis and other modulatory neurotransmitters and signaling systems. This paper presents a concise review of the effects of stress and glucocorticoids on the glutamatergic and monoaminergic (including noradrenergic, dopaminergic, and serotonergic systems) neurotransmitter systems as well as endocannabinoid signaling. Because of the breadth of the scope of this topic, the review is limited to the effects of stress on these brain systems on the prefrontal cortex, and where relevant, the hippocampus and the amygdala.
Collapse
|
59
|
Epigenetics and the regulation of stress vulnerability and resilience. Neuroscience 2013; 264:157-70. [PMID: 24333971 DOI: 10.1016/j.neuroscience.2013.12.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/29/2013] [Accepted: 12/03/2013] [Indexed: 12/13/2022]
Abstract
The human brain has a remarkable capacity to adapt to and learn from a wide range of variations in the environment. However, environmental challenges can also precipitate psychiatric disorders in susceptible individuals. Why any given experience should induce one brain to adapt while another is edged toward psychopathology remains poorly understood. Like all aspects of psychological function, both nature (genetics) and nurture (life experience) sculpt the brain's response to stressful stimuli. Here we review how these two influences intersect at the epigenetic regulation of neuronal gene transcription, and we discuss how the regulation of genomic DNA methylation near key stress-response genes may influence psychological susceptibility or resilience to environmental stressors. Our goal is to offer a perspective on the epigenetics of stress responses that works to bridge the gap between the study of this molecular process in animal models and its potential usefulness for understanding stress vulnerabilities in humans.
Collapse
|
60
|
Zannas AS, Binder EB. Gene-environment interactions at theFKBP5locus: sensitive periods, mechanisms and pleiotropism. GENES BRAIN AND BEHAVIOR 2013; 13:25-37. [DOI: 10.1111/gbb.12104] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 11/06/2013] [Accepted: 11/10/2013] [Indexed: 12/13/2022]
Affiliation(s)
- A. S. Zannas
- Max Planck Institute of Psychiatry; Munich Germany
- Department of Psychiatry; Duke University Medical Center; Durham NC USA
| | - E. B. Binder
- Max Planck Institute of Psychiatry; Munich Germany
- Department of Psychiatry and Behavioral Sciences; Emory University Medical School; Atlanta GA USA
| |
Collapse
|
61
|
Park SB, Jung WH, Kang NS, Park JS, Bae GH, Kim HY, Rhee SD, Kang SK, Ahn JH, Jeong HG, Kim KY. Anti-diabetic and anti-inflammatory effect of a novel selective 11β-HSD1 inhibitor in the diet-induced obese mice. Eur J Pharmacol 2013; 721:70-9. [DOI: 10.1016/j.ejphar.2013.09.052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 09/23/2013] [Accepted: 09/26/2013] [Indexed: 11/29/2022]
|
62
|
Juruena MF, Pariante CM, Papadopoulos AS, Poon L, Lightman S, Cleare AJ. The role of mineralocorticoid receptor function in treatment-resistant depression. J Psychopharmacol 2013; 27:1169-1179. [PMID: 23904409 DOI: 10.1177/0269881113499205] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Treatment-resistant depression patients show both reduced glucocorticoid receptor function and a hyperactive hypothalamic-pituitary-adrenal axis. However, few studies have examined the role of the mineralocorticoid receptor. This study aimed to evaluate the functional activity of the mineralocorticoid receptor system in regulating the hypothalamic-pituitary-adrenal axis in well-defined treatment-resistant depression patients. MATERIAL AND METHOD We recruited 24 subjects divided into: (a) treatment-resistant depression; (b) healthy controls. We evaluated: (a) the effect of combined glucocorticoid receptor/mineralocorticoid receptor stimulation with prednisolone; (b) the effect of prednisolone with the mineralocorticoid receptor antagonist spironolactone; and (c) the effect of spironolactone alone. The response of the hypothalamic-pituitary-adrenal axis was measured using salivary cortisol and plasma levels of drugs were also measured. RESULTS Treatment-resistant depression patients had higher cortisol compared with controls after all challenges. In controls, spironolactone increased cortisol compared to placebo. The co-administration of spironolactone with prednisolone in controls decreases the suppressive effects of prednisolone. In contrast, in treatment-resistant depression, spironolactone did not increase cortisol compared to placebo and spironolactone with prednisolone had no effect on the suppressive effects of prednisolone. Patients with treatment-resistant depression had a reduction in the conversation of spironolactone to the active metabolite canrenone. CONCLUSION Our data confirmed that treatment-resistant depression is associated with hypercortisolism and these patients no longer show an hypothalamic-pituitary-adrenal response to the administration of a mineralocorticoid receptor antagonist, suggesting that there is a mineralocorticoid receptor malfunctioning, such as a down regulation, however, pharmacokinetics and pharmacodynamics in these subjects could also have had an effect on the lack of mineralocorticoid receptor response.
Collapse
Affiliation(s)
- Mario F Juruena
- 1King's College London, Institute of Psychiatry, Section of Neurobiology of Mood Disorders, London, UK
| | | | | | | | | | | |
Collapse
|
63
|
Rose'meyer R. A review of the serotonin transporter and prenatal cortisol in the development of autism spectrum disorders. Mol Autism 2013; 4:37. [PMID: 24103554 PMCID: PMC3852299 DOI: 10.1186/2040-2392-4-37] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 09/13/2013] [Indexed: 01/28/2023] Open
Abstract
The diagnosis of autism spectrum disorder (ASD) during early childhood has a profound effect not only on young children but on their families. Aside from the physical and behavioural issues that need to be dealt with, there are significant emotional and financial costs associated with living with someone diagnosed with ASD. Understanding how autism occurs will assist in preparing families to deal with ASD, if not preventing or lessening its occurrence. Serotonin plays a vital role in the development of the brain during the prenatal and postnatal periods, yet very little is known about the serotonergic systems that affect children with ASD. This review seeks to provide an understanding of the biochemistry and physiological actions of serotonin and its termination of action through the serotonin reuptake transporter (SERT). Epidemiological studies investigating prenatal conditions that can increase the risk of ASD describe a number of factors which elevate plasma cortisol levels causing such symptoms during pregnancy such as hypertension, gestational diabetes and depression. Because cortisol plays an important role in driving dysregulation of serotonergic signalling through elevating SERT production in the developing brain, it is also necessary to investigate the physiological functions of cortisol, its action during gestation and metabolic syndromes.
Collapse
Affiliation(s)
- Roselyn Rose'meyer
- School of Medical Sciences, Griffith University, Gold Coast Campus, Parklands Drive, Southport, Queensland 4222, Australia.
| |
Collapse
|
64
|
Stovold R, Meredith SL, Bryant JL, Babur M, Williams KJ, Dean EJ, Dive C, Blackhall FH, White A. Neuroendocrine and epithelial phenotypes in small-cell lung cancer: implications for metastasis and survival in patients. Br J Cancer 2013; 108:1704-11. [PMID: 23519056 PMCID: PMC3668479 DOI: 10.1038/bjc.2013.112] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 02/12/2013] [Accepted: 02/20/2013] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Small-cell lung cancer (SCLC) has a very aggressive clinical course with early metastasis. This study investigated how the distinctive neuroendocrine characteristics contribute to disease progression and invasion in human SCLC. METHODS The neuroendocrine phenotype (pro-opiomelanocortin (POMC)) was quantified by ELISA in blood samples from 43 SCLC patients. The neuroendocrine (POMC, chromogranin A, neuron-specific enolase, NCAM) and epithelial (cytokeratin and E-cadherin) phenotypes were investigated, using ELISA and immunocytochemistry/immunohistochemistry. RESULTS In SCLC patients, 16% had elevated circulating POMC, which was associated with significantly worse survival (P=0.02) and liver metastases (P=0.004). In addition, POMC correlated with epithelial-positive circulating tumour cells (P=0.0002). In a panel of SCLC cell lines, all POMC-secreting cell lines expressed cytokeratin (40% of total). Even after cloning, DMS 79 cells expressed both neuroendocrine and epithelial markers. DMS 79 xenografts secreted POMC into the blood, which mirrored the tumour volume. These xenografts expressed both neuroendocrine and epithelial phenotypes in all tumours, with both phenotypes prevalent in cells invading the surrounding tissue. CONCLUSION Both neuroendocrine and epithelial phenotypes coexist in human SCLC tumours in vitro and in vivo and this persists in invading tumour cells. In patients, POMC secretion predicts poor survival and liver metastases, suggesting a crucial role of the neuroendocrine phenotype.
Collapse
Affiliation(s)
- R Stovold
- Faculty of Life Sciences, Manchester Academic Health Sciences Centre, University of Manchester, 3.016 AV Hill Building, Manchester M13 9PT, UK
- Paterson Institute for Cancer Research, University of Manchester, Manchester M20 4BX, UK
| | - S L Meredith
- Faculty of Life Sciences, Manchester Academic Health Sciences Centre, University of Manchester, 3.016 AV Hill Building, Manchester M13 9PT, UK
| | - J L Bryant
- Faculty of Life Sciences, Manchester Academic Health Sciences Centre, University of Manchester, 3.016 AV Hill Building, Manchester M13 9PT, UK
| | - M Babur
- Faculty of Medical and Human Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Stopford Building, Manchester M13 9PT, UK
| | - K J Williams
- Faculty of Medical and Human Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Stopford Building, Manchester M13 9PT, UK
| | - E J Dean
- Paterson Institute for Cancer Research, University of Manchester, Manchester M20 4BX, UK
| | - C Dive
- Paterson Institute for Cancer Research, University of Manchester, Manchester M20 4BX, UK
| | - F H Blackhall
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - A White
- Faculty of Life Sciences, Manchester Academic Health Sciences Centre, University of Manchester, 3.016 AV Hill Building, Manchester M13 9PT, UK
- Faculty of Medical and Human Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Stopford Building, Manchester M13 9PT, UK
| |
Collapse
|
65
|
Goncharova ND. Stress responsiveness of the hypothalamic-pituitary-adrenal axis: age-related features of the vasopressinergic regulation. Front Endocrinol (Lausanne) 2013; 4:26. [PMID: 23486926 PMCID: PMC3594837 DOI: 10.3389/fendo.2013.00026] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Accepted: 02/22/2013] [Indexed: 12/22/2022] Open
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis plays a key role in adaptation to environmental stresses. Parvicellular neurons of the hypothalamic paraventricular nucleus secrete corticotrophin releasing hormone (CRH) and arginine vasopressin (AVP) into pituitary portal system; CRH and AVP stimulate adrenocorticotropic hormone (ACTH) release through specific G-protein-coupled membrane receptors on pituitary corticotrophs, CRHR1 for CRH and V1b for AVP; the adrenal gland cortex secretes glucocorticoids in response to ACTH. The glucocorticoids activate specific receptors in brain and peripheral tissues thereby triggering the necessary metabolic, immune, neuromodulatory, and behavioral changes to resist stress. While importance of CRH, as a key hypothalamic factor of HPA axis regulation in basal and stress conditions in most species, is generally recognized, role of AVP remains to be clarified. This review focuses on the role of AVP in the regulation of stress responsiveness of the HPA axis with emphasis on the effects of aging on vasopressinergic regulation of HPA axis stress responsiveness. Under most of the known stressors, AVP is necessary for acute ACTH secretion but in a context-specific manner. The current data on the AVP role in regulation of HPA responsiveness to chronic stress in adulthood are rather contradictory. The importance of the vasopressinergic regulation of the HPA stress responsiveness is greatest during fetal development, in neonatal period, and in the lactating adult. Aging associated with increased variability in several parameters of HPA function including basal state, responsiveness to stressors, and special testing. Reports on the possible role of the AVP/V1b receptor system in the increase of HPA axis hyperactivity with aging are contradictory and requires further research. Many contradictory results may be due to age and species differences in the HPA function of rodents and primates.
Collapse
Affiliation(s)
- Nadezhda D. Goncharova
- Research Institute of Medical Primatology of Russian Academy of Medical SciencesSochi, Russia
- Sochi State UniversitySochi, Russia
| |
Collapse
|
66
|
Park JS, Rhee SD, Jung WH, Kang NS, Kim HY, Kang SK, Ahn JH, Kim KY. Anti-diabetic and anti-adipogenic effects of a novel selective 11β-hydroxysteroid dehydrogenase type 1 inhibitor in the diet-induced obese mice. Eur J Pharmacol 2012; 691:19-27. [PMID: 22760069 DOI: 10.1016/j.ejphar.2012.06.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 06/13/2012] [Accepted: 06/13/2012] [Indexed: 01/22/2023]
Abstract
Glucocorticoid excess (Cushing's syndrome) causes metabolic syndrome such as visceral obesity, insulin resistance, diabetes mellitus, dyslipidaemia and hypertension. The selective inhibitors of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) have considerable potential for treating type 2 diabetes mellitus and metabolic syndrome. In the present study, we investigated the anti-diabetic and anti-adipogenic effects of 4-(2-(1,1-dioxido-6-(2,4,6-trichlorophenyl)-1,2,6-thiadiazinan-2-yl)acetamido)adamantane-1-carboxamide (KR-67183), a novel selective 11β-HSD1 inhibitor; we also investigated the underlying molecular mechanisms in the cortisone-induced 3T3-L1 adipogenesis model system and diet-induced obese (DIO) mice. KR-67183 concentration-dependently inhibited 11β-HSD1 activity in human and mouse 11β-HSD1 over-expressed cells and in the ex vivo assay of C57BL/6 mice. In the study with DIO mice, the administration of KR-67183 (20 and 50mg/kg/day, orally for 28 days) improved the glucose tolerance and insulin sensitivity with suppressed 11β-HSD1 activity in the liver and fat. However, KR-67183 showed no change in the adrenal gland weight/body weight ratio and plasma corticosterone concentration in DIO mice. Further, KR-67183 suppressed adipocyte differentiation on cortisone-induced adipogenesis in 3T3-L1 cells is associated with the suppression of the cortisone-induced mRNA levels of FABP4, PPARγ2 and GLUT4, and 11β-HSD1 activity. Taken together, it is suggested that a selective 11β-HSD1 inhibitor, KR-67183, may provide a new therapeutic window in the prevention and treatment without toxicity in type 2 diabetes with obesity.
Collapse
Affiliation(s)
- Ji Seon Park
- Bio-Organic Science Division, Korea Research Institute of Chemical Technology, PO Box 107, Yuseong-gu, Daejeon 305-600, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Walker JJ, Spiga F, Waite E, Zhao Z, Kershaw Y, Terry JR, Lightman SL. The origin of glucocorticoid hormone oscillations. PLoS Biol 2012; 10:e1001341. [PMID: 22679394 PMCID: PMC3367982 DOI: 10.1371/journal.pbio.1001341] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 04/20/2012] [Indexed: 01/26/2023] Open
Abstract
Oscillating levels of adrenal glucocorticoid hormones are essential for optimal gene expression, and for maintaining physiological and behavioural responsiveness to stress. The biological basis for these oscillations is not known, but a neuronal "pulse generator" within the hypothalamus has remained a popular hypothesis. We demonstrate that pulsatile hypothalamic activity is not required for generating ultradian glucocorticoid oscillations. We show that a constant level of corticotrophin-releasing hormone (CRH) can activate a dynamic pituitary-adrenal peripheral network to produce ultradian adrenocorticotrophic hormone and glucocorticoid oscillations with a physiological frequency. This oscillatory response to CRH is dose dependent and becomes disrupted for higher levels of CRH. These data suggest that glucocorticoid oscillations result from a sub-hypothalamic pituitary-adrenal system, which functions as a deterministic peripheral hormone oscillator with a characteristic ultradian frequency. This constitutes a novel mechanism by which the level, rather than the pattern, of CRH determines the dynamics of glucocorticoid hormone secretion.
Collapse
Affiliation(s)
- Jamie J Walker
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Bristol, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
68
|
The use of cortisol for the objective assessment of stress in animals: Pros and cons. Vet J 2012; 192:137-9. [DOI: 10.1016/j.tvjl.2012.03.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 03/15/2012] [Accepted: 03/18/2012] [Indexed: 11/19/2022]
|
69
|
Perez-Castro C, Renner U, Haedo MR, Stalla GK, Arzt E. Cellular and molecular specificity of pituitary gland physiology. Physiol Rev 2012; 92:1-38. [PMID: 22298650 DOI: 10.1152/physrev.00003.2011] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The anterior pituitary gland has the ability to respond to complex signals derived from central and peripheral systems. Perception of these signals and their integration are mediated by cell interactions and cross-talk of multiple signaling transduction pathways and transcriptional regulatory networks that cooperate for hormone secretion, cell plasticity, and ultimately specific pituitary responses that are essential for an appropriate physiological response. We discuss the physiopathological and molecular mechanisms related to this integrative regulatory system of the anterior pituitary gland and how it contributes to modulate the gland functions and impacts on body homeostasis.
Collapse
Affiliation(s)
- Carolina Perez-Castro
- Laboratorio de Regulación de la Expresión Génica en el Crecimiento, Supervivencia y Diferenciación Celular,Departamento de Química Biológica, Universidad de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
70
|
Rankin J, Walker JJ, Windle R, Lightman SL, Terry JR. Characterizing dynamic interactions between ultradian glucocorticoid rhythmicity and acute stress using the phase response curve. PLoS One 2012; 7:e30978. [PMID: 22363526 PMCID: PMC3283588 DOI: 10.1371/journal.pone.0030978] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 12/30/2011] [Indexed: 11/29/2022] Open
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis is a dynamic oscillatory hormone signalling system that regulates the pulsatile secretion of glucocorticoids from the adrenal glands. In addition to regulation of basal levels of glucocorticoids, the HPA axis provides a rapid hormonal response to stress that is vitally important for homeostasis. Recently it has become clear that glucocorticoid pulses encode an important biological signal that regulates receptor signalling both in the central nervous system and in peripheral tissues. It is therefore important to understand how stressful stimuli disrupt the pulsatile dynamics of this system. Using a computational model that incorporates the crucial feed-forward and feedback components of the axis, we provide novel insight into experimental observations that the size of the stress-induced hormonal response is critically dependent on the timing of the stress. Further, we employ the theory of Phase Response Curves to show that an acute stressor acts as a phase-resetting mechanism for the ultradian rhythm of glucocorticoid secretion. Using our model, we demonstrate that the magnitude of an acute stress is a critical factor in determining whether the system resets via a Type 1 or Type 0 mechanism. By fitting our model to our in vivo stress-response data, we show that the glucocorticoid response to an acute noise stress in rats is governed by a Type 0 phase-resetting curve. Our results provide additional evidence for the concept of a deterministic sub-hypothalamic oscillator regulating the ultradian glucocorticoid rhythm, which constitutes a highly responsive peripheral hormone system that interacts dynamically with hypothalamic inputs to regulate the overall hormonal response to stress.
Collapse
Affiliation(s)
- James Rankin
- Bristol Centre for Applied Nonlinear Mathematics, University of Bristol, Bristol, United Kingdom
- INRIA Sophia-Antipolis, Sophia Antipolis, France
| | - Jamie J. Walker
- Bristol Centre for Applied Nonlinear Mathematics, University of Bristol, Bristol, United Kingdom
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, University of Bristol, Bristol, United Kingdom
| | - Richard Windle
- School of Nursing, Midwifery and Physiotherapy, University of Nottingham, Nottingham, United Kingdom
| | - Stafford L. Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, University of Bristol, Bristol, United Kingdom
| | - John R. Terry
- Henry Wellcome Laboratories for Integrative Neuroscience & Endocrinology, University of Bristol, Bristol, United Kingdom
- Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, United Kingdom
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, United Kingdom
- * E-mail:
| |
Collapse
|
71
|
Conway-Campbell BL, Pooley JR, Hager GL, Lightman SL. Molecular dynamics of ultradian glucocorticoid receptor action. Mol Cell Endocrinol 2012; 348:383-93. [PMID: 21872640 DOI: 10.1016/j.mce.2011.08.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 07/19/2011] [Accepted: 08/12/2011] [Indexed: 01/08/2023]
Abstract
In recent years it has become evident that glucocorticoid receptor (GR) action in the nucleus is highly dynamic, characterized by a rapid exchange at the chromatin template. This stochastic mode of GR action couples perfectly with a deterministic pulsatile availability of endogenous ligand in vivo. The endogenous glucocorticoid hormone (cortisol in man and corticosterone in rodent) is secreted from the adrenal gland with an ultradian rhythm made up of pulses at approximately hourly intervals. These two components - the rapidly fluctuating ligand and the rapidly exchanging receptor - appear to have evolved to establish and maintain a system that is exquisitely responsive to the physiological demands of the organism. In this review, we discuss recent and innovative work that questions the idea of steady state, static hormone receptor responses, and replaces them with new concepts of stochastic mechanisms and oscillatory activity essential for optimal function in molecular and cellular systems.
Collapse
Affiliation(s)
- Becky L Conway-Campbell
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol BS1 3NY, UK.
| | | | | | | |
Collapse
|
72
|
Andrews MH, Wood SA, Windle RJ, Lightman SL, Ingram CD. Acute glucocorticoid administration rapidly suppresses basal and stress-induced hypothalamo-pituitary-adrenal axis activity. Endocrinology 2012; 153:200-11. [PMID: 22087024 PMCID: PMC3279736 DOI: 10.1210/en.2011-1434] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hypothalamo-pituitary-adrenal (HPA) axis activity is subject to negative feedback control by glucocorticoids. Although the rapid component of this feedback is widely considered to contribute to regulation of dynamic HPA activity, few in vivo data exist on the temporal and pharmacological characteristics of this phenomenon. Thus, frequent automated blood sampling was undertaken in rats to determine the effects of acute glucocorticoid administration on basal and stress-induced corticosterone secretion. The glucocorticoid agonist methylprednisolone (5-2000 μg) or dexamethasone (5-500 μg) injected iv at the peak of the diurnal rhythm caused dose-dependent suppression of basal corticosterone secretion, which was attenuated by the glucocorticoid receptor antagonist RU38486. With 50 μg methylprednisolone, the onset of this suppression occurred at 40 min and remained significant for 120 min. However, although higher doses led to a greater and more sustained suppression of endogenous corticosterone, the response was delayed by the emergence of an initial stimulatory response that imposed a finite minimum delay. A corticosterone response to injection of CRH (1 μg, iv) during the period of maximal suppression indicated a suprapituitary site for the inhibitory effect glucocorticoid activation. This mechanism was supported by glucocorticoid injection immediately before a psychological stress (30 min, white noise); methylprednisolone caused dose-dependent attenuation of stress-induced corticosterone release and expression of the activity marker c-fos mRNA in the paraventricular nucleus but did not block the pituitary response to CRH. Thus, in rats, glucocorticoid receptor activation rapidly suppresses basal and stress-induced HPA activity that operates, at least in part, through a central mechanism of action.
Collapse
Affiliation(s)
- Marcus H Andrews
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol BS1 3NY, United Kingdom
| | | | | | | | | |
Collapse
|
73
|
Hill MN, Tasker JG. Endocannabinoid signaling, glucocorticoid-mediated negative feedback, and regulation of the hypothalamic-pituitary-adrenal axis. Neuroscience 2011; 204:5-16. [PMID: 22214537 DOI: 10.1016/j.neuroscience.2011.12.030] [Citation(s) in RCA: 235] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 12/13/2011] [Accepted: 12/15/2011] [Indexed: 12/20/2022]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis regulates the outflow of glucocorticoid hormones under basal conditions and in response to stress. Within the last decade, a large body of evidence has mounted indicating that the endocannabinoid system is involved in the central regulation of the stress response; however, the specific role endocannabinoid signaling plays in phases of HPA axis regulation, and the neural sites of action mediating this regulation, were not mapped out until recently. This review aims to collapse the current state of knowledge regarding the role of the endocannabinoid system in the regulation of the HPA axis to put together a working model of how and where endocannabinoids act within the brain to regulate outflow of the HPA axis. Specifically, we discuss the role of the endocannabinoid system in the regulation of the HPA axis under basal conditions, activation in response to acute stress, and glucocorticoid-mediated negative feedback. Interestingly, there appears to be some anatomical specificity to the role of the endocannabinoid system in each phase of HPA axis regulation, as well as distinct roles of both anandamide and 2-arachidonoylglycerol in these phases. Overall, the current level of information indicates that endocannabinoid signaling acts to suppress HPA axis activity through concerted actions within the prefrontal cortex, amygdala, and hypothalamus.
Collapse
Affiliation(s)
- M N Hill
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | | |
Collapse
|
74
|
Osterlund CD, Jarvis E, Chadayammuri A, Unnithan R, Weiser MJ, Spencer RL. Tonic, but not phasic corticosterone, constrains stress activatedextracellular-regulated-kinase 1/ 2 immunoreactivity within the hypothalamic paraventricular nucleus. J Neuroendocrinol 2011; 23:1241-51. [PMID: 21929693 PMCID: PMC3220802 DOI: 10.1111/j.1365-2826.2011.02220.x] [Citation(s) in RCA: 14] [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] [Indexed: 11/30/2022]
Abstract
The negative-feedback actions of corticosterone (CORT) depend on both phasic and tonic CORT secretion patterns to regulate hypothalamic-pituitary-adrenal (HPA) axis activity. How these two different CORT secretion pattens influence specific intracellular signal transduction pathway activity within the cellular elements of the HPA axis has not been determined. For example, it is unknown whether CORT has suppressive actions over signal transduction events within medial parvocellular paraventricular nucleus (PVN) corticotrophin-releasing hormone (CRH) neurones, nor whether these suppressive actions are responsible for alterations in PVN transcriptional processes and neurohormone secretion associated with stress. The extracellular-regulated kinase (ERK) is a stress activated intracellular signalling molecule that is potentially subject to glucocorticoid negative-feedback regulation. We tested the ability of CORT to modulate levels of the active (phosphorylated) form of ERK (pERK1/2) in the PVN of rats. Acute psychological stress (restraint) produced a rapid increase in the number of PVN pERK1/2 immunopositive cells within CRH neurones. Absence of tonic CORT via adrenalectomy (ADX) produced no change in basal pERK1/2 cell counts but augmented the increased pERK1/2 cell counts elicited by acute restraint. Treatment of ADX rats with CORT in the drinking water normalised this enhanced pERK1/2 response to stress. By contrast, treatment of ADX rats with a phasic increase in CORT 1 h before restraint had no effect on pERK1/2 cell counts, despite substantially suppressing stress-induced PVN crh gene expression and adrenonocorticotrophic hormone secretion. This tonic CORT inhibition of stress-induced activation of ERK1/2 may involve both alteration of the activity of stress-dependent neural inputs to PVN CRH neurones and alteration within those neurones of stress-dependent intracellular signalling mechanisms associated with ERK activation.
Collapse
Affiliation(s)
- C D Osterlund
- Department of Psychology and Neuroscience, University of Colorado, UCB 345, Boulder, CO 80309, USA.
| | | | | | | | | | | |
Collapse
|
75
|
Scheff JD, Calvano SE, Lowry SF, Androulakis IP. Transcriptional implications of ultradian glucocorticoid secretion in homeostasis and in the acute stress response. Physiol Genomics 2011; 44:121-9. [PMID: 22128089 DOI: 10.1152/physiolgenomics.00128.2011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Endogenous glucocorticoids are secreted by the hypothalamic-pituitary-adrenal (HPA) axis in response to a wide range of stressors. Glucocorticoids exert significant downstream effects, including the regulation of many inflammatory genes. The HPA axis functions such that glucocorticoids are released in a pulsatile manner, producing ultradian rhythms in plasma glucocorticoid levels. It is becoming increasingly evident that this ultradian pulsatility is important in maintaining proper homeostatic regulation and responsiveness to stress. This is particularly interesting from a clinical perspective given that pathological dysfunctions of the HPA axis produce altered ultradian patterns. Modeling this system facilitates the understanding of how glucocorticoid pulsatility arises, how it can be lost, and the transcriptional implications of ultradian rhythms. To approach these questions, we developed a mathematical model that integrates the cyclic production of glucocorticoids by the HPA axis and their downstream effects by integrating existing models of the HPA axis and glucocorticoid pharmacodynamics. This combined model allowed us to evaluate the implications of pulsatility in homeostasis as well as in response to acute stress. The presence of ultradian rhythms allows the system to maintain a lower response to homeostatic levels of glucocorticoids, but diminished feedback within the HPA axis leads to a loss of glucocorticoid rhythmicity. Furthermore, the loss of HPA pulsatility in homeostasis correlates with a decrease in the peak output in response to an acute stressor. These results are important in understanding how cyclic glucocorticoid secretion helps maintain the responsiveness of the HPA axis.
Collapse
Affiliation(s)
- Jeremy D Scheff
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| | | | | | | |
Collapse
|
76
|
Mason BL, Thomas SA, Lightman SL, Pariante CM. Desipramine treatment has minimal effects on the brain accumulation of glucocorticoids in P-gp-deficient and wild-type mice. Psychoneuroendocrinology 2011; 36:1351-60. [PMID: 21481537 PMCID: PMC3179986 DOI: 10.1016/j.psyneuen.2011.03.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 03/15/2011] [Accepted: 03/16/2011] [Indexed: 12/18/2022]
Abstract
Hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis in patients with depression can be reduced by antidepressants, which are thought to improve endogenous glucocorticoid-mediated negative feedback. A proportion of peripherally released glucocorticoids need to enter brain tissue, protected by the blood-brain barrier (BBB), in order to achieve this negative feedback effect at the level of the central nervous systems (CNS). The multidrug resistance transporter P-glycoprotein (P-gp) has been shown to actively transport glucocorticoid hormones and has been implicated in the regulation of glucocorticoid access to the CNS. Using an in situ brain/choroid plexus perfusion method, we tested the hypothesis that the antidepressant desipramine increases glucocorticoid accumulation in the mouse brain by inhibiting P-gp, following either chronic treatment (8 days, 20 mg/kg/day, IP) or acute administration (20 min brain perfusion in the presence of either 0.9 μM or 10 μM desipramine). Contrary to our hypothesis, chronic treatment with desipramine did not affect the accumulation of [³H]dexamethasone in any sample compared to saline-treated mice. Acute desipramine had limited and variable effects on glucocorticoid accumulation in the CNS, with accumulation of [³H]dexamethasone increased in the cerebellum, accumulation of [³H]cortisol reduced in the frontal cortex, hypothalamus, and cerebellum, and accumulation of [³H]corticosterone (the endogenous glucocorticoid in rodents) not affected. Overall, under the conditions tested, these results do not support the hypothesis that treatment with desipramine can inhibit P-gp at the BBB and subsequently increase the accumulation of glucocorticoids in the brain.
Collapse
Affiliation(s)
- Brittany L Mason
- Institute of Pharmaceutical Science, King's College London, London, UK.
| | | | | | | |
Collapse
|
77
|
Abstract
Stress activation of the hypothalamic-pituitary-adrenal (HPA) axis culminates in increased circulating corticosteroid concentrations. Stress-induced corticosteroids exert diverse actions in multiple target tissues over a broad range of timescales, ranging from rapid actions, which are induced within seconds to minutes and gene transcription independent, to slow actions, which are delayed, long lasting, and transcription dependent. Rapid corticosteroid actions in the brain include, among others, a fast negative feedback mechanism responsible for shutting down the activated HPA axis centrally. We provide a brief review of the cellular mechanisms responsible for rapid corticosteroid actions in different brain structures of the rat, including the hypothalamus, hippocampus, amygdala, and in the anterior pituitary. We propose a model for the direct feedback inhibition of the HPA axis by glucocorticoids in the hypothalamus. According to this model, glucocorticoids activate membrane glucocorticoid receptors to induce endocannabinoid synthesis in the hypothalamic paraventricular nucleus (PVN) and retrograde cannabinoid type I receptor-mediated suppression of the excitatory synaptic drive to PVN neuroendocrine cells. Rapid corticosteroid actions in the hippocampus, amygdala, and pituitary are mediated by diverse cellular mechanisms and may also contribute to the rapid negative feedback regulation of the HPA neuroendocrine axis as well as to the stress regulation of emotional and spatial memory formation.
Collapse
Affiliation(s)
- Jeffrey G Tasker
- Department of Cell and Molecular Biology and Neuroscience Program, Tulane University, New Orleans, LA 70118, USA.
| | | |
Collapse
|
78
|
Aguilera G. HPA axis responsiveness to stress: implications for healthy aging. Exp Gerontol 2011; 46:90-5. [PMID: 20833240 PMCID: PMC3026863 DOI: 10.1016/j.exger.2010.08.023] [Citation(s) in RCA: 174] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 08/19/2010] [Accepted: 08/27/2010] [Indexed: 02/06/2023]
Abstract
The major neuroendocrine response mediating stress adaptation is activation of the hypothalamic pituitary adrenal axis, with stimulation of corticotropin releasing hormone (CRH) and vasopressin (VP) from parvocellular neurons of the hypothalamic paraventricular nucleus, leading to stimulation of pituitary ACTH secretion and increases in glucocorticoid secretion from the adrenal cortex. Basal production and transient increases during stress of glucocorticoids and its hypothalamic regulators are essential for neuronal plasticity and normal brain function. While activation of the HPA axis is essential for survival during stress, chronic exposure to stress hormones can predispose to psychological, metabolic and immune alterations. Thus, prompt termination of the stress response is essential to prevent negative effects of inappropriate levels of CRH and glucocorticoids. This review addresses the regulation of HPA axis activity with emphasis on the mechanisms of termination of CRH transcription, which is a critical step in this process. In addition, the actions by which glucocorticoids, CRH and VP can affect the aging process will be discussed.
Collapse
Affiliation(s)
- Greti Aguilera
- Section on Endocrine Physiology, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shiver Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
79
|
Marković VM, Čupić Ž, Vukojević V, Kolar-Anić L. Predictive modeling of the hypothalamic-pituitary-adrenal (HPA) axis response to acute and chronic stress. Endocr J 2011; 58:889-904. [PMID: 21852742 DOI: 10.1507/endocrj.ej11-0037] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Detailed dynamics of the hypothalamic-pituitary-adrenal (HPA) axis is complex, depending on the individual metabolic load of an organism, its current status (healthy/ill, circadian phase (day/night), ultradian phase) and environmental impact. Therefore, it is difficult to compare the HPA axis activity between different individuals or draw unequivocal conclusions about the overall status of the HPA axis in an individual using single time-point measurements of cortisol levels. The aim of this study is to identify parameters that enable us to compare different dynamic states of the HPA axis and use them to investigate self-regulation mechanisms in the HPA axis under acute and chronic stress. In this regard, a four-dimensional stoichiometric model of the HPA axis was used. Acute stress was modeled by inducing an abrupt change in cortisol level during the course of numerical integration, whereas chronic stress was modeled by changing the mean stationary state concentrations of CRH. Effects of acute stress intensity, duration and time of onset with respect to the ultradian amplitude, ultradian phase and the circadian phase of the perturbed oscillation were studied in detail. Bifurcation analysis was used to predict the response of the HPA axis to chronic stress. Model predictions were compared with experimental findings reported in the literature and relevance for pharmacotherapy with glucocorticoids was discussed.
Collapse
Affiliation(s)
- Vladimir M Marković
- Faculty of Physical Chemistry, University of Belgrade, 11000 Belgrade, Serbia
| | | | | | | |
Collapse
|
80
|
Walker JJ, Terry JR, Tsaneva-Atanasova K, Armstrong SP, McArdle CA, Lightman SL. Encoding and decoding mechanisms of pulsatile hormone secretion. J Neuroendocrinol 2010; 22:1226-38. [PMID: 21054582 DOI: 10.1111/j.1365-2826.2010.02087.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Ultradian pulsatile hormone secretion underlies the activity of most neuroendocrine systems, including the hypothalamic-pituitary adrenal (HPA) and gonadal (HPG) axes, and this pulsatile mode of signalling permits the encoding of information through both amplitude and frequency modulation. In the HPA axis, glucocorticoid pulse amplitude increases in anticipation of waking, and, in the HPG axis, changing gonadotrophin-releasing hormone pulse frequency is the primary means by which the body alters its reproductive status during development (i.e. puberty). The prevalence of hormone pulsatility raises two crucial questions: how are ultradian pulses encoded (or generated) by these systems, and how are these pulses decoded (or interpreted) at their target sites? We have looked at mechanisms within the HPA axis responsible for encoding the pulsatile mode of glucocorticoid signalling that we observe in vivo. We review evidence regarding the 'hypothalamic pulse generator' hypothesis, and describe an alternative model for pulse generation, which involves steroid feedback-dependent endogenous rhythmic activity throughout the HPA axis. We consider the decoding of hormone pulsatility by taking the HPG axis as a model system and focussing on molecular mechanisms of frequency decoding by pituitary gonadotrophs.
Collapse
Affiliation(s)
- J J Walker
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, UK.
| | | | | | | | | | | |
Collapse
|
81
|
Sarabdjitsingh RA, Conway-Campbell BL, Leggett JD, Waite EJ, Meijer OC, de Kloet ER, Lightman SL. Stress responsiveness varies over the ultradian glucocorticoid cycle in a brain-region-specific manner. Endocrinology 2010; 151:5369-79. [PMID: 20861228 DOI: 10.1210/en.2010-0832] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Glucocorticoid hormones are released in rapid hourly hormone bursts by the adrenal gland. These ultradian oscillations are fundamental to hypothalamic-pituitary-adrenal activity and transcriptional regulation of glucocorticoid responsive genes. The physiological relevance of glucocorticoid pulsatility is however unknown. Using a novel automated infusion system, we artificially created different patterns (modulating pulse amplitude) of corticosterone (cort). Identical amounts of cort either in constant or in hourly pulses were infused into adrenalectomized rats. At the end of the infusion period, either during rising or falling concentrations of a cort pulse, animals were exposed to 99 dB noise stress (10 min). Pulsatile cort infusion led to a differential stress response, dependent on the phase of the pulse during which the stress was applied. Although constant administration of cort resulted in a blunted ACTH response to the stressor, a brisker response occurred during the rising phase of plasma cort than during the falling phase. This phase-dependent effect was also seen in the behavioral response to the stressor, which was again greater during the rising phase of each cort pulse. Within the brain itself, we found differential C-fos activation responses to noise stress in the pituitary, paraventricular nucleus, amygdala, and hippocampus. This effect was both glucocorticoid pulse amplitude and phase dependent, suggesting that different stress circuits are differentially responsive to the pattern of glucocorticoid exposure. Our data suggest that the oscillatory changes in plasma glucocorticoid levels are critical for the maintenance of normal physiological reactivity to a stressor and in addition modulate emotionality and exploratory behavior.
Collapse
Affiliation(s)
- Ratna A Sarabdjitsingh
- Division of Medical Pharmacology, Leiden/Amsterdam Centre for Drug Research/Leiden University Medical Centre, University of Leiden, Einsteinweg 55, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
82
|
Lightman SL, Conway-Campbell BL. The crucial role of pulsatile activity of the HPA axis for continuous dynamic equilibration. Nat Rev Neurosci 2010; 11:710-8. [PMID: 20842176 DOI: 10.1038/nrn2914] [Citation(s) in RCA: 255] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The classical concept of hypothalamus-pituitary-adrenal (HPA) homeostasis comprises a feedback system within which circulating levels of glucocorticoid hormones maintain the brain and body in an optimal steady state. However, studies involving new techniques for investigating the real-time dynamics of both glucocorticoid hormones and glucocorticoid receptor function paint a different picture--namely, of continuous dynamic equilibration throughout this neuroendocrine system. This dynamic state is dictated by feedforward and feedback regulatory loops and by stochastic interactions at the level of DNA binding. We propose that this continuous oscillatory activity is crucial for optimal responsiveness of glucocorticoid-sensitive neural processes.
Collapse
Affiliation(s)
- Stafford L Lightman
- University of Bristol, Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| | | |
Collapse
|
83
|
Harno E, White A. Will treating diabetes with 11β-HSD1 inhibitors affect the HPA axis? Trends Endocrinol Metab 2010; 21:619-27. [PMID: 20594868 DOI: 10.1016/j.tem.2010.06.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 06/01/2010] [Accepted: 06/03/2010] [Indexed: 11/28/2022]
Abstract
Inhibitors of 11β-HSD1 are in clinical trials for the treatment of type 2 diabetes. These compounds act by decreasing the cortisol generated in liver and adipose tissue, and therefore reducing tissue-specific gluconeogenesis and fatty acid metabolism. However, there is concern that reduction in tissue-regenerated cortisol might decrease feedback to the hypothalamic-pituitary-adrenal (HPA) axis, resulting in upregulation of cortisol from the adrenal gland. This review considers evidence from 11β-HSD1 knockout and transgenic mice, inhibitor studies and results from clinical trials evaluating HPA axis biomarkers. It is clear that analysis of the HPA axis is not sufficiently detailed, and there is a need to understand the subtle changes in the axis associated with pulsatility, diurnal rhythm and stress.
Collapse
Affiliation(s)
- Erika Harno
- Faculties of Life Sciences and Medical and Human Sciences, Manchester, Academic Health Sciences Centre, University of Manchester, UK
| | | |
Collapse
|