1
|
Kashkouli M, Jahanian Sadatmahalleh S, Ziaei S, Kazemnejad A, Saber A, Darvishnia H, Azarbayjani K. Relationship between postpartum depression and plasma vasopressin level at 6-8 weeks postpartum: a cross-sectional study. Sci Rep 2023; 13:3518. [PMID: 36864065 PMCID: PMC9981712 DOI: 10.1038/s41598-022-27223-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 12/28/2022] [Indexed: 03/04/2023] Open
Abstract
Postpartum depression (PPD) is the most important postpartum mood disorder due to its significant effect on both the infant and family health. Arginine vasopressin (AVP) has been suggested as a hormonal agent involved in the development of depression. The purpose of this study was to investigate the relationship between the plasma concentrations of AVP and the score of Edinburgh Postnatal Depression Scale (EPDS). This cross-sectional study was conducted in 2016-2017 in Darehshahr Township, Ilam Province, Iran. In the first phase, 303 pregnant women, who were at 38 weeks, met the inclusion criteria, and were not depressed (according to their EPDS scores) were included in the study. In the 6-8 week postpartum follow-up, using the EPDS, 31 depressed individuals were diagnosed and referred to a psychiatrist for confirmation. The maternal venous blood samples of 24 depressed individuals still meeting the inclusion criteria and 66 randomly selected non-depressed subjects were obtained to measure their AVP plasma concentrations with ELISA assay. There was a significant positive relationship between plasma AVP levels and the EPDS score (P = 0.000, r = 0.658). Also the mean plasma concentration of AVP was significantly higher in the depressed group (41.35 ± 13.75 ng/ml) than in the non-depressed group (26.01 ± 7.83 ng/ml) (P < 0.001). In a multiple logistic regression model for various parameters, increased vasopressin levels were associated with increased odds of PPD (OR = 1.15, 95% CI = 1.07-1.24, P = 0.000). Furthermore, multiparity (OR = 5.45, 95% CI = 1.21-24.43, P = 0.027) and non-exclusive breastfeeding (OR = 13.06, 95% CI = 1.36-125, P = 0.026) were associated with increased odds of PPD. Maternal gender preference (having a baby of desired and desired sex) decreased the odds of PPD (OR = 0.13, 95% CI = 0.02-0.79, P = 0.027 and OR = 0.08, 95% CI = 0.01-0.5, P = 0.007). AVP seems to be a contributor to clinical PPD by affecting the hypothalamic-pituitary-adrenal (HPA) axis activity. Furthermore, primiparous women had significantly lower EPDS scores.
Collapse
Affiliation(s)
- Masoumeh Kashkouli
- grid.412266.50000 0001 1781 3962Department of Reproductive Health and Midwifery, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shahideh Jahanian Sadatmahalleh
- grid.412266.50000 0001 1781 3962Department of Reproductive Health and Midwifery, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeideh Ziaei
- Department of Reproductive Health and Midwifery, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Anoshirvan Kazemnejad
- grid.412266.50000 0001 1781 3962Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ashraf Saber
- Department of Nursing, Faculty of Medical Sciences, Esfarayen, Iran
| | - Hamid Darvishnia
- grid.412462.70000 0000 8810 3346Department of Biology, Payame Noor University (PNU), Tehran, Iran
| | - Khadijeh Azarbayjani
- grid.412266.50000 0001 1781 3962Department of Reproductive Health and Midwifery, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
2
|
Bu G, Lin Y, Liu J, Yu P, Yong T, Yang M, Huang L, Han X, Du X, Kong F, Huang A, Zeng X, Meng F. Evidence for progesterone acting as an inhibitor of stress axis via stimulating pituitary neuropeptide B/W receptor 2 (NPBWR2) expression in chickens. J Steroid Biochem Mol Biol 2023; 226:106218. [PMID: 36368625 DOI: 10.1016/j.jsbmb.2022.106218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022]
Abstract
In vertebrates, the hypothalamus-pituitary-adrenal gland (HPA) axis is the main endocrine pathway regulating the stress response, thus also called the stress axis. It has been well-accepted that the stress axis is tightly controlled by both hypothalamic stimulators and inhibitors [e.g. corticotropin (ACTH)-releasing inhibitory factor (CRIF)]. However, the identity of authentic CRIF remains unclear for decades. Recently, neuropeptide W (NPW) was proved to be the physiological CRIF in chickens. Together with its functional receptor (NPBWR2), they play critical roles in attenuating the activity of the chicken stress axis. Because increasing pieces of evidence suggested that sex steroids could regulate the stress axis, using chicken as a model, we investigated whether the newly identified CRIF and its receptor are under the control of sex steroids in this study. Our results showed that: (1) expression of NPW-NPBWR2 in the hypothalamus-pituitary axis was sexually dimorphic and developmental stage-dependent; (2) progesterone (P4), rather than 17β-estradiol (E2) and dihydrotestosterone (DHT), could dose- and time-dependently upregulate NPBWR2 expression, which was accompanied with the decrease of ACTH synthesis and secretion, in cultured pituitary cells; (3) intraperitoneal injection of P4 could elevate the mRNA level of pituitary NPBWR2; (4) P4-stimulated NPBWR2 expression was relevant to both nPR-mediated genomic action and mPRs-triggered nongenomic route associated with MEK/ERK, PI3K/AKT cascade, and calcium influx. To our knowledge, our results discover a novel route of sex steroids in modulating the stress axis of chickens, which lays a foundation to reveal the complicated interaction network between reproduction and stress axes in chickens.
Collapse
Affiliation(s)
- Guixian Bu
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China.
| | - Ying Lin
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Jianfeng Liu
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Pan Yu
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Tao Yong
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Ming Yang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Linyan Huang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Xingfa Han
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Xiaogang Du
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Fanli Kong
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Anqi Huang
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Xianyin Zeng
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Fengyan Meng
- College of Life Science, Sichuan Agricultural University, Ya'an 625014, PR China.
| |
Collapse
|
3
|
Stewart CA, Finger EC. The supraoptic and paraventricular nuclei in healthy aging and neurodegeneration. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:105-123. [PMID: 34225924 DOI: 10.1016/b978-0-12-820107-7.00007-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The supraoptic (SON) and paraventricular (PVN) nuclei of the hypothalamus undergo structural and functional changes over the course of healthy aging. These nuclei and their connections are also heterogeneously affected by several different neurodegenerative diseases. This chapter reviews the involvement of the SON and PVN, the hypothalamic-pituitary axes, and the peptide hormones produced in both nuclei in healthy aging and in neurodegeneration, with a focus on Alzheimer's disease (AD), frontotemporal dementia (FTD), amyotrophic lateral sclerosis, progressive supranuclear palsy, Parkinson's disease (PD), dementia with Lewy bodies (DLB), multiple system atrophy, and Huntington's disease. Although age-related changes occur in several regions of the hypothalamus, the SON and PVN are relatively preserved during aging and in many neurodegenerative disorders. With aging, these nuclei do undergo some sexually dimorphic changes including changes in size and levels of vasopressin and corticotropin-releasing hormone, likely due to age-related changes in sex hormones. In contrast, oxytocinergic cells and circulating levels of thyrotropin-releasing hormone remain stable. A relative resistance to many forms of neurodegenerative pathology is also observed, in comparison to other hypothalamic and brain regions. Mirroring the pattern observed in aging, pathologic hallmarks of AD, and some subtypes of FTD are observed in the PVN, though to a milder degree than are observed in other brain regions, while the SON is relatively spared. In contrast, the SON appears more vulnerable to alpha-synuclein pathology of DLB and PD. The consequences of these alterations may help to inform several of the physiologic changes observed in aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Chloe A Stewart
- Department of Clinical Neurological Sciences, Lawson Health Research Institute, London, ON, Canada; Graduate Program in Neuroscience, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Elizabeth C Finger
- Department of Clinical Neurological Sciences, Lawson Health Research Institute, London, ON, Canada; Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
4
|
Melief J, Huitinga I, Gold SM. The stress-axis in multiple sclerosis: Clinical, cellular, and molecular aspects. HANDBOOK OF CLINICAL NEUROLOGY 2021; 181:119-126. [PMID: 34238451 DOI: 10.1016/b978-0-12-820683-6.00008-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Altered activity of the hypothalamus-pituitary-adrenal (HPA) stress-axis has been implicated in the pathogenesis and progression of multiple sclerosis (MS) and linked to the development of specific symptoms and comorbidities such as mood disorders, fatigue, or cognitive dysfunction. Overall the HPA-axis is activated or hyperresponsive in MS, though a hyporesponsive HPA-axis has been observed in a subgroup of MS patients that has a more severe course of the disease. Here we provide an overview of the possible causes of HPA-axis activation, sex- and subtype dependent differences, pathological, cellular, and molecular effects, and the clinical correlates of HPA-axis activity in MS.
Collapse
Affiliation(s)
- Jeroen Melief
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands; Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| | - Stefan M Gold
- Department of Psychiatry and Medical Department, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Berlin, Germany; Institute for Neuroimmunology and Multiple Sclerosis, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
5
|
Swaab DF, Wolff SEC, Bao AM. Sexual differentiation of the human hypothalamus: Relationship to gender identity and sexual orientation. HANDBOOK OF CLINICAL NEUROLOGY 2021; 181:427-443. [PMID: 34238476 DOI: 10.1016/b978-0-12-820683-6.00031-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gender identity (an individual's perception of being male or female) and sexual orientation (heterosexuality, homosexuality, or bisexuality) are programmed into our brain during early development. During the intrauterine period in the second half of pregnancy, a testosterone surge masculinizes the fetal male brain. If such a testosterone surge does not occur, this will result in a feminine brain. As sexual differentiation of the brain takes place at a much later stage in development than sexual differentiation of the genitals, these two processes can be influenced independently of each other and can result in gender dysphoria. Nature produces a great variability for all aspects of sexual differentiation of the brain. Mechanisms involved in sexual differentiation of the brain include hormones, genetics, epigenetics, endocrine disruptors, immune response, and self-organization. Furthermore, structural and functional differences in the hypothalamus relating to gender dysphoria and sexual orientation are described in this review. All the genetic, postmortem, and in vivo scanning observations support the neurobiological theory about the origin of gender dysphoria, i.e., it is the sizes of brain structures, the neuron numbers, the molecular composition, functions, and connectivity of brain structures that determine our gender identity or sexual orientation. There is no evidence that one's postnatal social environment plays a crucial role in the development of gender identity or sexual orientation.
Collapse
Affiliation(s)
- Dick F Swaab
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Samantha E C Wolff
- Department Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Ai-Min Bao
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Corticotropin-Releasing Factor Family: A Stress Hormone-Receptor System's Emerging Role in Mediating Sex-Specific Signaling. Cells 2020; 9:cells9040839. [PMID: 32244319 PMCID: PMC7226788 DOI: 10.3390/cells9040839] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/25/2020] [Accepted: 03/28/2020] [Indexed: 12/14/2022] Open
Abstract
No organ in the body is impervious to the effects of stress, and a coordinated response from all organs is essential to deal with stressors. A dysregulated stress response that fails to bring systems back to homeostasis leads to compromised function and ultimately a diseased state. The components of the corticotropin-releasing factor (CRF) family, an ancient and evolutionarily conserved stress hormone-receptor system, helps both initiate stress responses and bring systems back to homeostasis once the stressors are removed. The mammalian CRF family comprises of four known agonists, CRF and urocortins (UCN1–3), and two known G protein-coupled receptors (GPCRs), CRF1 and CRF2. Evolutionarily, precursors of CRF- and urocortin-like peptides and their receptors were involved in osmoregulation/diuretic functions, in addition to nutrient sensing. Both CRF and UCN1 peptide hormones as well as their receptors appeared after a duplication event nearly 400 million years ago. All four agonists and both CRF receptors show sex-specific changes in expression and/or function, and single nucleotide polymorphisms are associated with a plethora of human diseases. CRF receptors harbor N-terminal cleavable peptide sequences, conferring biased ligand properties. CRF receptors have the ability to heteromerize with each other as well as with other GPCRs. Taken together, CRF receptors and their agonists due to their versatile functional adaptability mediate nuanced responses and are uniquely positioned to orchestrate sex-specific signaling and function in several tissues.
Collapse
|
7
|
Swaab DF, Bao AM. Sex differences in stress-related disorders: Major depressive disorder, bipolar disorder, and posttraumatic stress disorder. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:335-358. [PMID: 33008536 DOI: 10.1016/b978-0-444-64123-6.00023-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Stress-related disorders, such as mood disorders and posttraumatic stress disorder (PTSD), are more common in women than in men. This sex difference is at least partly due to the organizing effect of sex steroids during intrauterine development, while activating or inhibiting effects of circulating sex hormones in the postnatal period and adulthood also play a role. Such effects result in structural and functional changes in neuronal networks, neurotransmitters, and neuropeptides, which make the arousal- and stress-related brain systems more vulnerable to environmental stressful events in women. Certain brainstem nuclei, the amygdala, habenula, prefrontal cortex, and hypothalamus are important hubs in the stress-related neuronal network. Various hypothalamic nuclei play a central role in this sexually dimorphic network. This concerns not only the hypothalamus-pituitary-adrenal axis (HPA-axis), which integrates the neuro-endocrine-immune responses to stress, but also other hypothalamic nuclei and systems that play a key role in the symptoms of mood disorders, such as disordered day-night rhythm, lack of reward feelings, disturbed eating and sex, and disturbed cognitive functions. The present chapter focuses on the structural and functional sex differences that are present in the stress-related brain systems in mood disorders and PTSD, placing the HPA-axis in the center. The individual differences in the vulnerability of the discussed systems, caused by genetic and epigenetic developmental factors warrant further research to develop tailor-made therapeutic strategies.
Collapse
Affiliation(s)
- Dick F Swaab
- Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands; Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Zhejiang, China.
| | - Ai-Min Bao
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Zhejiang, China; Key Laboratory of Mental Disorder Management, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Kovács LÁ, Berta G, Csernus V, Ujvári B, Füredi N, Gaszner B. Corticotropin-Releasing Factor-Producing Cells in the Paraventricular Nucleus of the Hypothalamus and Extended Amygdala Show Age-Dependent FOS and FOSB/DeltaFOSB Immunoreactivity in Acute and Chronic Stress Models in the Rat. Front Aging Neurosci 2019; 11:274. [PMID: 31649527 PMCID: PMC6794369 DOI: 10.3389/fnagi.2019.00274] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/24/2019] [Indexed: 01/01/2023] Open
Abstract
Corticotropin-releasing factor (CRF) immunoreactive (ir) neurons of the paraventricular nucleus of the hypothalamus (PVN) play pivotal role in the coordination of stress response. CRF-producing cells in the central nucleus of amygdala (CeA) and oval division of the bed nucleus of stria terminalis (BNSTov) are also involved in stress adaptation and mood control. Immediate early gene products, subunits of the transcription factor activator protein 1 (AP1) are commonly used as acute (FOS) and/or chronic (FOSB/deltaFOSB) markers for the neuronal activity in stress research. It is well known that the course of aging affects stress adaptation, but little is known about the aging-related stress sensitivity of CRF neurons. To the best of our knowledge, the stress-induced neuronal activity of CRF neurons in the course of aging in acute and chronic stress models was not studied systematically yet. Therefore, the aim of the present study was to quantify the acute restraint stress (ARS) and chronic variable mild stress (CVMS) evoked neuronal activity in CRF cells of the PVN, CeA, and BNSTov using triple-label immunofluorescence throughout the whole lifespan in the rat. We hypothesized that the FOS and FOSB content of CRF cells upon ARS or CVMS decreases with age. Our results showed that the FOS and FOSB response to ARS declined with age in the PVN-CRF cells. BNSTov and CeA CRF cells did not show remarkable stress-induced elevation of these markers neither in ARS, nor in CVMS. Exposure to CVMS resulted in an age-independent significant increase of FOSB/delta FOSB immunosignal in PVN-CRF neurons. Unexpectedly, we detected a remarkable stress-independent FOSB/deltaFOSB signal in CeA- and BNSTov-CRF cells that declined with the course of aging. In summary, PVN-CRF cells show decreasing acute stress sensitivity (i.e., FOS and FOSB immunoreactivity) with the course of aging, while their (FOSB/deltaFOSB) responsivity to chronic challenge is maintained till senescence. Stress exposure does not affect the occurrence of the examined Fos gene products in CeA- and BNSTov-CRF cells remarkably suggesting that their contribution to stress adaptation response does not require AP1-controlled transcriptional changes.
Collapse
Affiliation(s)
- László Á Kovács
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary.,Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, University of Pécs Medical School, Pécs, Hungary
| | - Valér Csernus
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary
| | - Balázs Ujvári
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary.,Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Nóra Füredi
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary.,Centre for Neuroscience, Pécs University, Pécs, Hungary
| | - Balázs Gaszner
- Department of Anatomy, University of Pécs Medical School, Pécs, Hungary.,Centre for Neuroscience, Pécs University, Pécs, Hungary
| |
Collapse
|
9
|
Bao AM, Swaab DF. The human hypothalamus in mood disorders: The HPA axis in the center. IBRO Rep 2018; 6:45-53. [PMID: 31211281 PMCID: PMC6562194 DOI: 10.1016/j.ibror.2018.11.008] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/28/2018] [Indexed: 02/08/2023] Open
Abstract
There are no specific structural neuropathological hallmarks found in the brain of mood disorders. Instead, there are molecular, functional and structural alterations reported in many brain areas. The neurodevelopmental underpinning indicated the presence of various genetic and developmental risk factors. The effect of genetic polymorphisms and developmental sequalae, some of which may start in the womb, result in functional changes in a network mediated by neurotransmitters and neuropeptides, which make the emotion- and stress-related brain systems more vulnerable to stressful events. This network of stress-related neurocircuits consists of, for instance, brainstem nuclei, the amygdala, habenula, prefrontal cortex and hypothalamus. Various nuclei of the hypothalamus form indeed one of the crucial hubs in this network. This structure concerns not only the hypothalamo-pituitary-adrenal (HPA) axis that integrate the neuro-endocrine-immune responses to stress, but also other hypothalamic nuclei and systems that play a key role in the symptoms of depression, such as disordered day-night rhythm, lack of reward feelings, disturbed eating, sex, and disturbed cognitive functions. The present review will focus on the changes in the human hypothalamus in depression, with the HPA axis in the center. We will discuss the inordinate network of neurotransmitters and neuropeptides involved, with the hope to find the most vulnerable neurobiological systems and the possible development of tailor-made treatments for mood disorders in the future.
Collapse
Affiliation(s)
- Ai-Min Bao
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Institute of neuroscience, NHC and CAMS key laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Dick F Swaab
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, Institute of neuroscience, NHC and CAMS key laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.,Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Neurobiological characteristics underlying metabolic differences between males and females. Prog Neurobiol 2018; 176:18-32. [PMID: 30194984 DOI: 10.1016/j.pneurobio.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/22/2018] [Accepted: 09/01/2018] [Indexed: 12/24/2022]
Abstract
The hypothalamus is the main integrating center for metabolic control. Our understanding of how hypothalamic circuits function to control appetite and energy expenditure has increased dramatically in recent years, due to the rapid rise in the incidence of obesity and the search for effective treatments. Increasing evidence indicates that these treatments will most likely differ between males and females. Indeed, sex differences in metabolism have been demonstrated at various levels, including in two of the most studied neuronal populations involved in metabolic control: the anorexigenic proopiomelanocortin neurons and the orexigenic neuropeptide Y/Agouti-related protein neurons. Here we review what is known to date regarding the sex differences in these two neuronal populations, as well as other neuronal populations involved in metabolic control and glial cells.
Collapse
|
11
|
Zhou JN, Fang H. Transcriptional regulation of corticotropin-releasing hormone gene in stress response. IBRO Rep 2018; 5:137-146. [PMID: 30591954 PMCID: PMC6303479 DOI: 10.1016/j.ibror.2018.08.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 08/22/2018] [Indexed: 01/29/2023] Open
Abstract
As a central player of the hypothalamic-pituitary-adrenal (HPA) axis, the corticotropin -releasing hormone (CRH) neurons in the hypothalamic paraventricular nucleus (PVN) determine the state of HPA axis and play a key role in stress response. Evidence supports that during stress response the transcription and expression of CRH was finely tuned, which involved cis-element-transcriptional factor (TF) interactions and epigenetic mechanisms. Here we reviewed recent progress in CRH transcription regulation from DNA methylation to classic TFs regulation, in which a number of paired receptors were involved. The imbalance of multiple paired receptors in regulating the activity of CRH neurons indicates a possible molecular network mechanisms underlying depression etiology and directs novel therapeutic strategies of depression in the future.
Collapse
Affiliation(s)
- Jiang-Ning Zhou
- Corresponding author at: School of Life Science, University of Science and Technology of China, Hefei, 230027, Anhui, PR China.
| | | |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Neurodevelopmental disorders disproportionately affect males. The mechanisms underlying male vulnerability or female protection are not known and remain understudied. Determining the processes involved is crucial to understanding the etiology and advancing treatment of neurodevelopmental disorders. Here, we review current findings and theories that contribute to male preponderance of neurodevelopmental disorders, with a focus on autism. RECENT FINDINGS Recent work on the biological basis of the male preponderance of autism and other neurodevelopmental disorders includes discussion of a higher genetic burden in females and sex-specific gene mutations or epigenetic changes that differentially confer risk to males or protection to females. Other mechanisms discussed are sex chromosome and sex hormone involvement. Specifically, fetal testosterone is involved in many aspects of development and may interact with neurotransmitter, neuropeptide, or immune pathways to contribute to male vulnerability. Finally, the possibilities of female underdiagnosis and a multi-hit hypothesis are discussed. This review highlights current theories of male bias in developmental disorders. Topics include environmental, genetic, and epigenetic mechanisms; theories of sex chromosomes, hormones, neuroendocrine, and immune function; underdiagnosis of females; and a multi-hit hypothesis.
Collapse
Affiliation(s)
- Sarah L. Ferri
- Department of Molecular Physiology and Biophysics, Iowa Neuroscience Institute, University of Iowa, Pappajohn Biomedical Discovery Building, 169 Newton Road, Iowa City, IA 52242 USA
| | - Ted Abel
- Department of Molecular Physiology and Biophysics, Iowa Neuroscience Institute, University of Iowa, Pappajohn Biomedical Discovery Building, 169 Newton Road, Iowa City, IA 52242 USA
| | - Edward S. Brodkin
- Center for Neurobiology and Behavior, Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Translational Research Laboratory, 125 South 31 Street, Room 2202, Philadelphia, PA 19104-3403 USA
| |
Collapse
|
13
|
The art of matching brain tissue from patients and controls for postmortem research. HANDBOOK OF CLINICAL NEUROLOGY 2018; 150:197-217. [PMID: 29496142 DOI: 10.1016/b978-0-444-63639-3.00015-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The quality of postmortem research depends strongly on a thorough clinical investigation and documentation of the patient's disorder and therapies. In addition, a systematic and professional neuropathologic investigation of both cases and controls is absolutely crucial. In the experience of the Netherlands Brain Bank (NBB), about 20% of clinical neurologic diagnoses, despite being made in first-rate clinics, have to be revised or require an extra diagnosis after a complete and thorough review by the NBB. The neuropathology examination may reveal for instance that the "controls" already have preclinical neurodegenerative alterations. In postmortem studies the patient and control groups must be matched for as many of the known confounding factors as possible. This is necessary to make the groups as similar as possible, except for the topic being investigated. Confounding factors are present before, during, and after death. They are respectively: (1) genetic background, systemic diseases, duration and gravity of illness, medicines and addictive compounds used, age, sex, gender identity, sexual orientation, circadian and seasonal fluctuations, lateralization; (2) agonal state, stress of dying; and (3) postmortem delay, freezing procedures, fixation and storage time. Consequently, a brain bank should have a large number of controls at its disposal for appropriate matching. If matching fails for some confounders, then their influence may be determined by statistical methods such as analysis of variance or regression models.
Collapse
|
14
|
Valentino RJ, Bangasser DA. Sex-biased cellular signaling: molecular basis for sex differences in neuropsychiatric diseases. DIALOGUES IN CLINICAL NEUROSCIENCE 2017. [PMID: 28179810 PMCID: PMC5286724 DOI: 10.31887/dcns.2016.18.4/rvalentino] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The recognition that there are fundamental biological sex differences that extend beyond those that define sexual behavior and reproductive function has inspired the drive toward inclusion of both sexes in research design. This is supported by an underlying clinical rationale that studying both sexes is necessary to elucidate pathophysiology and develop treatments for the entire population. However, at a more basic level, sex differences, like genetic differences, can be exploited to better understand biology. Here, we discuss how sex differences at the molecular level of cell signaling and protein trafficking are amplified to create a state of vulnerability that under the right conditions can result in symptoms of neuropsychiatry disease. Although this dialogue focuses on the specific example of corticotropin-releasing factor, the potential for analogous sex differences in signaling and/or trafficking of receptors for other neuromodulators has broad biological and therapeutic implications.
Collapse
Affiliation(s)
- Rita J Valentino
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia and University of Pennsylvania, USA
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, USA
| |
Collapse
|
15
|
Grimm S, Wirth K, Fan Y, Weigand A, Gärtner M, Feeser M, Dziobek I, Bajbouj M, Aust S. The interaction of corticotropin-releasing hormone receptor gene and early life stress on emotional empathy. Behav Brain Res 2017; 329:180-185. [DOI: 10.1016/j.bbr.2017.04.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/13/2022]
|
16
|
Cooper O, Bonert V, Moser F, Mirocha J, Melmed S. Altered Pituitary Gland Structure and Function in Posttraumatic Stress Disorder. J Endocr Soc 2017; 1:577-587. [PMID: 29264511 PMCID: PMC5686623 DOI: 10.1210/js.2017-00069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 04/10/2017] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Posttraumatic stress disorder (PTSD) is associated with hypothalamus-pituitary-adrenal (HPA) axis response to stressors, but links to neurophysiological and neuroanatomical changes are unclear. The purpose of this study was to determine whether stress-induced cortisol alters negative feedback on pituitary corticotroph function and pituitary volume. DESIGN Prospective controlled study in an outpatient clinic. METHODS Subjects with PTSD and matched control subjects underwent pituitary volume measurement on magnetic resonance imaging, with pituitary function assessed by 24-hour urine free cortisol (UFC), 8:00 am cortisol, and adrenocorticotropic hormone (ACTH) levels, and ACTH levels after 2-day dexamethasone/corticotropin-releasing hormone test. Primary outcome was pituitary volume; secondary outcomes were ACTH area under the curve (AUC) and 24-hour UFC. RESULTS Thirty-nine subjects were screened and 10 subjects with PTSD were matched with 10 healthy control subjects by sex and age. Mean pituitary volume was 729.7 mm3 [standard deviation (SD), 227.3 mm3] in PTSD subjects vs 835.2 mm3 (SD, 302.8 mm3) in control subjects. ACTH AUC was 262.5 pg/mL (SD, 133.3 pg/mL) L in PTSD vs 244.0 pg/mL (SD, 158.3 pg/mL) in control subjects (P = 0.80). In PTSD subjects, UFC levels and pituitary volume inversely correlated with PTSD duration; pituitary volume correlated with ACTH AUC in control subjects (Pearson correlation coefficient, 0.88, P = 0.0009) but not in PTSD subjects. CONCLUSIONS The HPA axis may be downregulated and dysregulated in people with PTSD, as demonstrated by discordant pituitary corticotroph function and pituitary volume vs intact HPA feedback and correlation of pituitary volume with ACTH levels in healthy control subjects. The results suggest a link between pituitary structure and function in PTSD, which may point to endocrine targeted therapeutic approaches.
Collapse
Affiliation(s)
- Odelia Cooper
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Vivien Bonert
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Franklin Moser
- Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - James Mirocha
- Biostatistics Core, Research Institute, Cedars-Sinai Medical Center, Los Angeles, California 90048
| | - Shlomo Melmed
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048
| |
Collapse
|
17
|
Tenk J, Rostás I, Füredi N, Mikó A, Solymár M, Soós S, Gaszner B, Feller D, Székely M, Pétervári E, Balaskó M. Age-related changes in central effects of corticotropin-releasing factor (CRF) suggest a role for this mediator in aging anorexia and cachexia. GeroScience 2017; 39:61-72. [PMID: 28299639 PMCID: PMC5352590 DOI: 10.1007/s11357-017-9962-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 10/20/2016] [Indexed: 11/29/2022] Open
Abstract
Hypothalamic corticotropin-releasing factor (CRF) lays downstream to catabolic melanocortins and at least partly mediates their catabolic effects. Age-related changes in the melanocortin system (weak responsiveness in middle-aged and a strong one in old rats) have been shown to contribute to middle-aged obesity and later to aging anorexia and cachexia of old age groups. We hypothesized that catabolic (anorexigenic and hypermetabolic) CRF effects vary with aging similarly to those of melanocortins. Thus, we aimed to test whether age-related variations of CRF effects may also contribute to middle-aged obesity and aging anorexia leading to weight loss of old age groups. Food intake, body weight, core temperature, heart rate, and activity were recorded in male Wistar rats of young, middle-aged, aging, and old age groups (from 3 to 24 months) during a 7-day intracerebroventricular CRF infusion (0.2 μg/μl/h) in a biotelemetric system. In addition, CRF gene expression was also assessed by quantitative RT-PCR in the paraventricular nucleus (PVN) of intact animals of the same age groups. The infusion suppressed body weight in the young, aging, and old rats, but not in middle-aged animals. Weak anorexigenic and hypermetabolic effects were detected in the young, whereas strong anorexia (without hypermetabolism) developed in the oldest age groups in which post mortem analysis showed also a reduction of retroperitoneal fat mass. CRF gene expression in the PVN increased with aging. Our results support the potential contribution of age-related changes in CRF effects to aging anorexia and cachexia. The role of the peptide in middle-aged obesity cannot be confirmed.
Collapse
Affiliation(s)
- Judit Tenk
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Ildikó Rostás
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Nóra Füredi
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Alexandra Mikó
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Margit Solymár
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Szilvia Soós
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Balázs Gaszner
- Department of Anatomy, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Diana Feller
- Department of Pharmaceutical Biotechnology, Medical School, University of Pecs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Miklós Székely
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Erika Pétervári
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary
| | - Márta Balaskó
- Institute for Translational Medicine, Medical School, University of Pécs, 12 Szigeti str, Pécs, H-7624, Hungary.
| |
Collapse
|
18
|
Tenk J, Mátrai P, Hegyi P, Rostás I, Garami A, Szabó I, Solymár M, Pétervári E, Czimmer J, Márta K, Mikó A, Füredi N, Párniczky A, Zsiborás C, Balaskó M. In Obesity, HPA Axis Activity Does Not Increase with BMI, but Declines with Aging: A Meta-Analysis of Clinical Studies. PLoS One 2016; 11:e0166842. [PMID: 27870910 PMCID: PMC5117724 DOI: 10.1371/journal.pone.0166842] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 11/05/2016] [Indexed: 12/17/2022] Open
Abstract
Background Obesity is one of the major public health challenges worldwide. It involves numerous endocrine disorders as etiological factors or as complications. Previous studies strongly suggested the involvement of the hypothalamic-pituitary-adrenal (HPA) axis activity in obesity, however, to date, no consistent trend in obesity-associated alterations of the HPA axis has been identified. Aging has been demonstrated to aggravate obesity and to induce abnormalities of the HPA axis. Thus, the question arises whether obesity is correlated with peripheral indicators of HPA function in adult populations. Objectives We aimed to meta-analyze literature data on peripheral cortisol levels as indicators of HPA activity in obesity during aging, in order to identify possible explanations for previous contradictory findings and to suggest new approaches for future clinical studies. Data Sources 3,596 records were identified through searching of PubMed, Embase and Cochrane Library Database. Altogether 26 articles were suitable for analyses. Study Eligibility Criteria Empirical research papers were eligible provided that they reported data of healthy adult individuals, included body mass index (BMI) and measured at least one relevant peripheral cortisol parameter (i.e., either morning blood cortisol or 24-h urinary free cortisol). Statistical Methods We used random effect models in each of the meta-analyses calculating with the DerSimonian and Laird weighting methods. I-squared indicator and Q test were performed to assess heterogeneity. Meta-regression was applied to explore the effect of BMI and age on morning blood and urinary free cortisol levels. To assess publication bias Egger’s test was used. Results Obesity did not show any correlation with the studied peripheral cortisol values. On the other hand, peripheral cortisol levels declined with aging within the obese, but not in the non-obese groups. Conclusions Our analysis demonstrated that obesity or healthy aging does not lead to enhanced HPA axis activity, peripheral cortisol levels rather decline with aging.
Collapse
Affiliation(s)
- Judit Tenk
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Mátrai
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Institute of Bioanalysis, Medical School, University of Pécs, Pécs, Hungary
| | - Péter Hegyi
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Department of Translational Medicine, University of Pécs, Pécs, Hungary
- Hungarian Academy of Sciences - University of Szeged, Momentum Gastroenterology Multidisciplinary Research Group, Szeged, Hungary
| | - Ildikó Rostás
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - András Garami
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Imre Szabó
- Department of Gastroenterology, First Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Margit Solymár
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Erika Pétervári
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - József Czimmer
- Department of Gastroenterology, First Department of Internal Medicine, University of Pécs, Pécs, Hungary
| | - Katalin Márta
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Department of Translational Medicine, University of Pécs, Pécs, Hungary
| | - Alexandra Mikó
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Nóra Füredi
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Andrea Párniczky
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
- Department of Translational Medicine, University of Pécs, Pécs, Hungary
| | - Csaba Zsiborás
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Márta Balaskó
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
19
|
Tenk J, Rostás I, Füredi N, Mikó A, Soós S, Solymár M, Gaszner B, Székely M, Pétervári E, Balaskó M. Acute central effects of corticotropin-releasing factor (CRF) on energy balance: Effects of age and gender. Peptides 2016; 85:63-72. [PMID: 27637621 DOI: 10.1016/j.peptides.2016.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/12/2016] [Accepted: 09/12/2016] [Indexed: 10/21/2022]
Abstract
Previously demonstrated age-related changes in the catabolic melanocortin system that may contribute to middle-aged obesity and aging anorexia, raise the question of the potential involvement of corticotropin-releasing factor (CRF) in these phenomena, as this catabolic hypothalamic mediator acts downstream to melanocortins. Catabolic effects of CRF were shown to be mediated by both CRF1 (hypermetabolism) and CRF2 (anorexia) receptors. To test the potential role of CRF in age-related obesity and aging anorexia, we investigated acute central effects of the peptide on energy balance in male and female rats during the course of aging. Effects of an intracerebroventricular CRF injection on food intake (FI), oxygen-consumption (VO2), core- and tail skin temperatures (Tc and Ts) were studied in male and female Wistar rats of five different age-groups (from 3- to 24-month). Anorexigenic responsiveness was tested during 180-min re-feeding (FeedScale) following 24-h fasting. Thermoregulatory analysis was performed by indirect calorimetry (Oxymax) complemented by thermocouples recording Tc and Ts (indicating heat loss). CRF suppressed FI in 3-month male and female animals. In males, CRF-induced anorexia declined with aging, whereas in females it was maintained in all groups. The peptide increased VO2 and Tc in all male age-groups, while the weaker hypermetabolic response characterizing 3-month females declined rapidly with aging. Thus, age-related alterations in acute central anorexigenic and hypermetabolic effects of CRF show different non-parallel patterns in males and females. Our findings underline the importance of gender differences. They also call the attention to the differential age-related changes in the CRF1 and CRF2 receptor systems.
Collapse
Affiliation(s)
- Judit Tenk
- Institute for Translational Medicine, Medical School, University of Pécs, Hungary
| | - Ildikó Rostás
- Institute for Translational Medicine, Medical School, University of Pécs, Hungary
| | - Nóra Füredi
- Institute for Translational Medicine, Medical School, University of Pécs, Hungary
| | - Alexandra Mikó
- Institute for Translational Medicine, Medical School, University of Pécs, Hungary
| | - Szilvia Soós
- Institute for Translational Medicine, Medical School, University of Pécs, Hungary
| | - Margit Solymár
- Institute for Translational Medicine, Medical School, University of Pécs, Hungary
| | - Balázs Gaszner
- Department of Anatomy, Medical School, University of Pécs, Hungary
| | - Miklós Székely
- Institute for Translational Medicine, Medical School, University of Pécs, Hungary
| | - Erika Pétervári
- Institute for Translational Medicine, Medical School, University of Pécs, Hungary
| | - Márta Balaskó
- Institute for Translational Medicine, Medical School, University of Pécs, Hungary.
| |
Collapse
|
20
|
Stress, sex, and addiction: potential roles of corticotropin-releasing factor, oxytocin, and arginine-vasopressin. Behav Pharmacol 2015; 25:445-57. [PMID: 24949572 DOI: 10.1097/fbp.0000000000000049] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Stress sensitivity and sex are predictive factors for the development of neuropsychiatric disorders. Life stresses are not only risk factors for the development of addiction but also are triggers for relapse to drug use. Therefore, it is imperative to elucidate the molecular mechanisms underlying the interactions between stress and drug abuse, as an understanding of this may help in the development of novel and more effective therapeutic approaches to block the clinical manifestations of drug addiction. The development and clinical course of addiction-related disorders do appear to involve neuroadaptations within neurocircuitries that modulate stress responses and are influenced by several neuropeptides. These include corticotropin-releasing factor, the prototypic member of this class, as well as oxytocin and arginine-vasopressin that play important roles in affiliative behaviors. Interestingly, these peptides function to balance emotional behavior, with sexual dimorphism in the oxytocin/arginine-vasopressin systems, a fact that might play an important role in the differential responses of women and men to stressful stimuli and the specific sex-based prevalence of certain addictive disorders. Thus, this review aims to summarize (i) the contribution of sex differences to the function of dopamine systems, and (ii) the behavioral, neurochemical, and anatomical changes in brain stress systems.
Collapse
|
21
|
Goel N, Workman JL, Lee TT, Innala L, Viau V. Sex differences in the HPA axis. Compr Physiol 2015; 4:1121-55. [PMID: 24944032 DOI: 10.1002/cphy.c130054] [Citation(s) in RCA: 252] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis is a major component of the systems that respond to stress, by coordinating the neuroendocrine and autonomic responses. Tightly controlled regulation of HPA responses is critical for maintaining mental and physical health, as hyper- and hypo-activity have been linked to disease states. A long history of research has revealed sex differences in numerous components of the HPA stress system and its responses, which may partially form the basis for sex disparities in disease development. Despite this, many studies use male subjects exclusively, while fewer reports involve females or provide direct sex comparisons. The purpose of this article is to present sex comparisons in the functional and molecular aspects of the HPA axis, through various phases of activity, including basal, acute stress, and chronic stress conditions. The HPA axis in females initiates more rapidly and produces a greater output of stress hormones. This review focuses on the interactions between the gonadal hormone system and the HPA axis as the key mediators of these sex differences, whereby androgens increase and estrogens decrease HPA activity in adulthood. In addition to the effects of gonadal hormones on the adult response, morphological impacts of hormone exposure during development are also involved in mediating sex differences. Additional systems impinging on the HPA axis that contribute to sex differences include the monoamine neurotransmitters norepinephrine and serotonin. Diverse signals originating from the brain and periphery are integrated to determine the level of HPA axis activity, and these signals are, in many cases, sex-specific.
Collapse
Affiliation(s)
- Nirupa Goel
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
22
|
Grimm S, Gärtner M, Fuge P, Fan Y, Weigand A, Feeser M, Aust S, Heekeren HR, Jacobs A, Heuser I, Bajbouj M. Variation in the corticotropin-releasing hormone receptor 1 (CRHR1) gene modulates age effects on working memory. J Psychiatr Res 2015; 61:57-63. [PMID: 25541005 DOI: 10.1016/j.jpsychires.2014.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/16/2014] [Accepted: 12/03/2014] [Indexed: 01/13/2023]
Abstract
Decline in working memory (WM) functions during aging has been associated with hippocampal dysfunction mediated by age-related changes to the corticotropin-releasing hormone (CRH) system. Recent reports suggest that GG-homozygous individuals of single nucleotide polymorphisms (rs110402 and rs242924) in the CRH receptor 1 (CRHR1) gene show increased stress vulnerability and decreased BOLD responses in WM relevant regions. However, until now, no study investigated the interaction effects of variation in the CRHR1 gene and age on individual differences in WM. Here, young, middle-aged and old subjects (N = 466) were genotyped for rs110402 and rs242924 within the CRHR1 gene and an n-back task was used to investigate the hypothesis that vulnerable genotypes (GG-homozygotes) would show impaired WM functions that might be magnified by increased CRH production with advancing age. Our results show an impact of genotype already in middle-age with significantly better performance in AT-carriers. Working memory performance in AT-carriers did not differ between young and middle-aged subjects, but was significantly impaired in old age. In GG-homozygotes, severe working memory dysfunction occurred already in middle age. Our data indicate that GG-homozygotes of CRHR1 rs110402 and rs242924 represent a genetically driven subtype of early WM impairments due to alterations in hippocampal CRHR1 activation. Early interventions that have proven effective in delaying cognitive decline appear to be particularly important for these subjects at risk for premature memory decline, who are in the prime of their personal and professional lives.
Collapse
Affiliation(s)
- Simone Grimm
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Hospital of Psychiatry, University of Zurich, 8032 Zurich, Switzerland.
| | - Matti Gärtner
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Philipp Fuge
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Yan Fan
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany
| | - Anne Weigand
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Melanie Feeser
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany
| | - Sabine Aust
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Hauke R Heekeren
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany
| | - Arthur Jacobs
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany
| | - Isabella Heuser
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Malek Bajbouj
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| |
Collapse
|
23
|
Stratton MS, Staros M, Budefeld T, Searcy BT, Nash C, Eitel C, Carbone D, Handa RJ, Majdic G, Tobet SA. Embryonic GABA(B) receptor blockade alters cell migration, adult hypothalamic structure, and anxiety- and depression-like behaviors sex specifically in mice. PLoS One 2014; 9:e106015. [PMID: 25162235 PMCID: PMC4146593 DOI: 10.1371/journal.pone.0106015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 07/27/2014] [Indexed: 11/18/2022] Open
Abstract
Neurons of the paraventricular nucleus of the hypothalamus (PVN) regulate the hypothalamic- pituitary-adrenal (HPA) axis and the autonomic nervous system. Females lacking functional GABAB receptors because of a genetic disruption of the R1 subunit have altered cellular characteristics in and around the PVN at birth. The genetic disruption precluded appropriate assessments of physiology or behavior in adulthood. The current study was conducted to test the long term impact of a temporally restricting pharmacological blockade of the GABAB receptor to a 7-day critical period (E11–E17) during embryonic development. Experiments tested the role of GABAB receptor signaling in fetal development of the PVN and later adult capacities for adult stress related behaviors and physiology. In organotypic slices containing fetal PVN, there was a female specific, 52% increase in cell movement speeds with GABAB receptor antagonist treatment that was consistent with a sex-dependent lateral displacement of cells in vivo following 7 days of fetal exposure to GABAB receptor antagonist. Anxiety-like and depression-like behaviors, open-field activity, and HPA mediated responses to restraint stress were measured in adult offspring of mothers treated with GABAB receptor antagonist. Embryonic exposure to GABAB receptor antagonist resulted in reduced HPA axis activation following restraint stress and reduced depression-like behaviors. There was also increased anxiety-like behavior selectively in females and hyperactivity in males. A sex dependent response to disruptions of GABAB receptor signaling was identified for PVN formation and key aspects of physiology and behavior. These changes correspond to sex specific prevalence in similar human disorders, namely anxiety disorders and hyperactivity.
Collapse
Affiliation(s)
- Matthew S. Stratton
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Michelle Staros
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Tomaz Budefeld
- Center for Animal Genomics, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Brian T. Searcy
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Connor Nash
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Chad Eitel
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - David Carbone
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, United States of America
| | - Robert J. Handa
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, Arizona, United States of America
| | - Gregor Majdic
- Center for Animal Genomics, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Stuart A. Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
24
|
Bangasser DA, Valentino RJ. Sex differences in stress-related psychiatric disorders: neurobiological perspectives. Front Neuroendocrinol 2014; 35:303-19. [PMID: 24726661 PMCID: PMC4087049 DOI: 10.1016/j.yfrne.2014.03.008] [Citation(s) in RCA: 476] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 02/26/2014] [Accepted: 03/28/2014] [Indexed: 12/14/2022]
Abstract
Stress is associated with the onset and severity of several psychiatric disorders that occur more frequently in women than men, including posttraumatic stress disorder (PTSD) and depression. Patients with these disorders present with dysregulation of several stress response systems, including the neuroendocrine response to stress, corticolimbic responses to negatively valenced stimuli, and hyperarousal. Thus, sex differences within their underlying circuitry may explain sex biases in disease prevalence. This review describes clinical studies that identify sex differences within the activity of these circuits, as well as preclinical studies that demonstrate cellular and molecular sex differences in stress responses systems. These studies reveal sex differences from the molecular to the systems level that increase endocrine, emotional, and arousal responses to stress in females. Exploring these sex differences is critical because this research can reveal the neurobiological underpinnings of vulnerability to stress-related psychiatric disorders and guide the development of novel pharmacotherapies.
Collapse
Affiliation(s)
- Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States.
| | - Rita J Valentino
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
25
|
Sex differences in the adult HPA axis and affective behaviors are altered by perinatal exposure to a low dose of bisphenol A. Brain Res 2014; 1571:12-24. [DOI: 10.1016/j.brainres.2014.05.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 04/23/2014] [Accepted: 05/10/2014] [Indexed: 01/17/2023]
|
26
|
Rodrigues R, Petersen RB, Perry G. Parallels between major depressive disorder and Alzheimer's disease: role of oxidative stress and genetic vulnerability. Cell Mol Neurobiol 2014; 34:925-49. [PMID: 24927694 DOI: 10.1007/s10571-014-0074-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 05/14/2014] [Indexed: 12/19/2022]
Abstract
The thesis of this review is that oxidative stress is the central factor in major depressive disorder (MDD) and Alzheimer's disease (AD). The major elements involved are inflammatory cytokines, the hypothalamic-pituitary axis, the hypothalamic-pituitary gonadal, and arginine vasopressin systems, which induce glucocorticoid and "oxidopamatergic" cascades when triggered by psychosocial stress, severe life-threatening events, and mental-affective and somatic diseases. In individuals with a genomic vulnerability to depression, these cascades may result in chronic depression-anxiety-stress spectra, resulting in MDD and other known depressive syndromes. In contrast, in subjects with genomic vulnerability to AD, oxidative stress-induced brain damage triggers specific antioxidant defenses, i.e., increased levels of amyloid-β (Aβ) and aggregation of hyper-phosphorylated tau, resulting in paired helical filaments and impaired functions related to the ApoEε4 isoform, leading to complex pathological cascades culminating in AD. Surprisingly, all the AD-associated molecular pathways mentioned in this review have been shown to be similar or analogous to those found in depression, including structural damage, i.e., hippocampal and frontal cortex atrophy. Other interacting molecular signals, i.e., GSK-3β, convergent survival factors (brain-derived neurotrophic factor and heat shock proteins), and transition redox metals are also mentioned to emphasize the vast array of intermediates that could interact via comparable mechanisms in both MDD and AD.
Collapse
Affiliation(s)
- Roberto Rodrigues
- College of Sciences, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA,
| | | | | |
Collapse
|
27
|
Gao SF, Klomp A, Wu JL, Swaab DF, Bao AM. Reduced GAD(65/67) immunoreactivity in the hypothalamic paraventricular nucleus in depression: a postmortem study. J Affect Disord 2013; 149:422-5. [PMID: 23312397 DOI: 10.1016/j.jad.2012.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 12/01/2012] [Accepted: 12/04/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Gamma-aminobutyric acid (GABA) is a major inhibitory neurotransmitter. It diminishes the activity of the hypothalamo-pituitary-adrenal (HPA) axis, which plays an important role in the pathogenesis of depression. The present study aimed at determining GABAergic input in the hypothalamic paraventricular nucleus (PVN) in depression and its correlation with the activity of corticotropin-releasing hormone (CRH) neurons. METHODS The density of glutamic acid decarboxylase (GAD)(65/67)-immunoreactivity (ir) was quantified in the postmortem hypothalamic PVN of 9 major depressive (MDD) and 5 bipolar depressive (BD) patients, together with 12 matched controls, whose CRH-expressing neuron numbers had been determined in a previous study. RESULTS There was a 43% significant reduction of the density of GAD(65/67)-ir in the PVN in MDD (P=0.028) and a 20% non-significant decrease in BD patients. In addition, there was a significant negative correlation between the density of GAD(65/67)-ir and the number of CRH-ir neurons in the PVN in the depression group (Rho=-0.527, P=0.032), but not in the control group. LIMITATIONS The samples were relatively small and the depression group had used antidepressants. CONCLUSION A diminished GABAergic input to the PVN may contribute to the activation of CRH-ir neurons in depression, most prominently in MDD, which provides a rationale for prescribing GABAergic agonists for these patients.
Collapse
Affiliation(s)
- Shang-Feng Gao
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | | | | | | | | |
Collapse
|
28
|
Wang Q, Van Heerikhuize J, Aronica E, Kawata M, Seress L, Joels M, Swaab DF, Lucassen PJ. Glucocorticoid receptor protein expression in human hippocampus; stability with age. Neurobiol Aging 2013; 34:1662-73. [PMID: 23290588 DOI: 10.1016/j.neurobiolaging.2012.11.019] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 11/18/2012] [Accepted: 11/25/2012] [Indexed: 01/22/2023]
Abstract
The glucocorticoid receptor (GR) exerts numerous functions in the body and brain. In the brain, it has been implicated, amongst others, in feedback regulation of the hypothalamic-pituitary-adrenal axis, with potential deficits during aging and in depression. GRs are abundantly expressed in the hippocampus of rodent, except for the Ammon's horn (CA) 3 subregion. In rhesus monkey however, GR protein was largely absent from all hippocampal subregions, which prompted us to investigate its distribution in human hippocampus. After validation of antibody specificity, we investigated GRα protein distribution in the postmortem hippocampus of 26 human control subjects (1-98 years of age) and quantified changes with age and sex. In contrast to monkey, abundant GR-immunoreactivity was present in nuclei of almost all neurons of the hippocampal CA subfields and dentate gyrus (DG), although neurons of the CA3 subregion displayed lower levels of immunoreactivity. Colocalization with glial fibrillary acidic protein confirmed that GR was additionally expressed in approximately 50% of the astrocytes in the CA regions, with lower levels of colocalization (approximately 20%) in the DG. With increased age, GR expression remained stable in the CA regions in both sexes, whereas a significant negative correlation was found with age only in the DG of females. Thus, in contrast to the very low levels previously reported in monkey, GR protein is prominently expressed in human hippocampus, indicating that this region can form an important target for corticosteroid effects in human.
Collapse
Affiliation(s)
- Qian Wang
- Swammerdam Institute for Life Sciences (SILS)-Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
29
|
|
30
|
Volumetric parcellation methodology of the human hypothalamus in neuroimaging: normative data and sex differences. Neuroimage 2012; 69:1-10. [PMID: 23247186 DOI: 10.1016/j.neuroimage.2012.12.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 11/30/2012] [Accepted: 12/01/2012] [Indexed: 11/22/2022] Open
Abstract
There is increasing evidence regarding the importance of the hypothalamus for understanding sex differences in relation to neurological, psychiatric, endocrine and sleep disorders. Although different in histology, physiology, connections and function, multiple hypothalamic nuclei subserve non-voluntary functions and are nodal points for the purpose of maintaining homeostasis of the organism. Thus, given the critical importance of hypothalamic nuclei and their key multiple roles in regulating basic functions, it is important to develop the ability to conduct in vivo human studies of anatomic structure, volume, connectivity, and function of hypothalamic regions represented at the level of its nuclei. The goals of the present study were to develop a novel method of semi-automated volumetric parcellation for the human hypothalamus that could be used to investigate clinical conditions using MRI and to demonstrate its applicability. The proposed new method subdivides the hypothalamus into five parcels based on visible anatomic landmarks associated with specific nuclear groupings and was confirmed using two ex vivo hypothalami that were imaged in a 7 T (7 T) scanner and processed histologically. Imaging results were compared with histology from the same brain. Further, the method was applied to 44 healthy adults (26 men; 18 women, comparable on age, handedness, ethnicity, SES) to derive normative volumes and assess sex differences in hypothalamic regions using 1.5 T MRI. Men compared to women had a significantly larger total hypothalamus, relative to cerebrum size, similar for both hemispheres, a difference that was primarily driven by the tuberal region, with the sex effect size being largest in the superior tuberal region and, to a lesser extent, inferior tuberal region. Given the critical role of hypothalamic nuclei in multiple chronic diseases and the importance of sex differences, we argue that the use of the novel methodology presented here will allow for critical investigations of these disorders and further delineation of potential treatments, particularly sex-specific approaches to gene and drug discoveries that involve hypothalamic nuclei.
Collapse
|
31
|
Gao SF, Qi XR, Zhao J, Balesar R, Bao AM, Swaab DF. Decreased NOS1 expression in the anterior cingulate cortex in depression. ACTA ACUST UNITED AC 2012; 23:2956-64. [PMID: 22989585 DOI: 10.1093/cercor/bhs285] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Decreased function of the anterior cingulate cortex (ACC) is crucially involved in the pathogenesis of depression. A key role of nitric oxide (NO) has also been proposed. We aimed to determine the NO content in the cerebrospinal fluid (CSF) and the expression of NO synthase (NOS) isoforms, that is, NOS1, NOS2, and NOS3 in the ACC in depression. In depressive patients, CSF-NOx levels (the levels of the NO metabolites nitrite and nitrate) were significantly decreased (P = 0.007), indicating a more general decrease of NO production in this disorder. This agreed with a trend toward lower NOS1-mRNA levels (P = 0.083) and a significant decrease of NOS1-immunoreactivity (ir) (P = 0.043) in ACC. In controls, there was a significant positive correlation between ACC-NOS1-ir cell densities and their CSF-NOx levels. Furthermore, both localization of NOS1 in pyramidal neurons that are known to be glutamatergic and co-localization between NOS1 and GABAergic neurons were observed in human ACC. The diminished ACC-NOS1 expression and decreased CSF-NOx levels may be involved in the alterations of ACC activity in depression, possibly by affecting glutamatergic and GABAergic neurotransmission.
Collapse
Affiliation(s)
- Shang-Feng Gao
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, PR China and
| | | | | | | | | | | |
Collapse
|
32
|
Bangasser DA, Valentino RJ. Sex differences in molecular and cellular substrates of stress. Cell Mol Neurobiol 2012; 32:709-23. [PMID: 22488525 DOI: 10.1007/s10571-012-9824-4] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 02/22/2012] [Indexed: 12/20/2022]
Abstract
Women are twice as likely as men to suffer from stress-related psychiatric disorders, like unipolar depression and post-traumatic stress disorder. Although the underlying neural mechanisms are not well characterized, the pivotal role of stress in the onset and severity of these diseases has led to the idea that sex differences in stress responses account for this sex bias. Corticotropin-releasing factor (CRF) orchestrates stress responses by acting both as a neurohormone to initiate the hypothalamic-pituitary-adrenal (HPA) axis and as a neuromodulator in the brain. One target of CRF modulation is the locus coeruleus (LC)-norepinephrine system, which coordinates arousal components of the stress response. Hypersecretion of CRF and dysregulation of targets downstream from CRF, such as the HPA axis and LC-norepinephrine system, are characteristic features of many stress-related psychiatric diseases, suggesting a causal role for CRF and its targets in the development of these disorders. This review will describe sex differences in CRF and the LC-norepinephrine system that can increase stress sensitivity in females, making them vulnerable to stress-related disorders. Evidence for gonadal hormone regulation of hypothalamic CRF is discussed as an effect that can lead to increased HPA axis activity in females. Sex differences in the structure of LC neurons that create the potential for hyperarousal in response to emotional stimuli are described. Finally, sex differences at the molecular level of the CRF(1) receptor that make the LC-norepinephrine system more reactive in females are reviewed. The implications of these sex differences for the treatment of stress-related psychiatric disorders also will be discussed.
Collapse
Affiliation(s)
- Debra A Bangasser
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | |
Collapse
|
33
|
van Wamelen DJ, Aziz NA, Anink JJ, Roos RAC, Swaab DF. Paraventricular nucleus neuropeptide expression in Huntington's disease patients. Brain Pathol 2012; 22:654-61. [PMID: 22257050 DOI: 10.1111/j.1750-3639.2012.00565.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Neuroendocrine, metabolic and autonomic nervous system dysfunctions are prevalent among patients with Huntington's disease (HD) and may underlie symptoms such as depression, weight loss and autonomic failure. Using post-mortem paraffin-embedded tissue, we assessed the integrity of the major neuropeptide populations in the paraventricular nucleus (PVN)-the hypothalamic neuroendocrine and autonomic integration center-in HD patients. The number corticotropin-releasing hormone, cocaine- and amphetamine-regulated transcript, arginine vasopressin and oxytocin immunoreactive (ir) neurons did not differ between HD patients and control subjects. However, the significant positive correlation between arginine vasopressin and oxytocin ir neurons in control subjects (P = 0.036) was absent in patients. Corticotropin-releasing hormone mRNA levels were 68% higher in HD patients (P = 0.046). Thyrotropin-releasing hormone mRNA levels did not differ between HD patients and control subjects, although a negative correlation with disease duration was present in the former (P = 0.036). These findings indicate that the PVN is largely unaffected in HD patients. However, our findings suggest that hypothalamic-pituitary-thyroid axis activity may alter during the course of the disease and that autonomic nervous system dysfunction might partly arise from an imbalance between arginine vasopressin and oxytocin neurons in the PVN.
Collapse
Affiliation(s)
- Daniel J van Wamelen
- Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam ZO, The Netherlands.
| | | | | | | | | |
Collapse
|
34
|
Bao AM, Ruhé HG, Gao SF, Swaab DF. Neurotransmitters and neuropeptides in depression. HANDBOOK OF CLINICAL NEUROLOGY 2012; 106:107-36. [PMID: 22608619 DOI: 10.1016/b978-0-444-52002-9.00008-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- A-M Bao
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.
| | | | | | | |
Collapse
|
35
|
Abstract
The underlying assumption in popular and scientific publications on sex differences in the brain is that human brains can take one of two forms “male” or “female,” and that the differences between these two forms underlie differences between men and women in personality, cognition, emotion, and behavior. Documented sex differences in brain structure are typically taken to support this dimorphic view of the brain. However, neuroanatomical data reveal that sex interacts with other factors in utero and throughout life to determine the structure of the brain, and that because these interactions are complex, the result is a multi-morphic, rather than a dimorphic, brain. More specifically, here I argue that human brains are composed of an ever-changing heterogeneous mosaic of “male” and “female” brain characteristics (rather than being all “male” or all “female”) that cannot be aligned on a continuum between a “male brain” and a “female brain.” I further suggest that sex differences in the direction of change in the brain mosaic following specific environmental events lead to sex differences in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Daphna Joel
- Department of Psychology, Tel-Aviv University Tel-Aviv, Israel
| |
Collapse
|
36
|
Hippocampal GR expression is increased in elderly depressed females. Neuropharmacology 2011; 62:527-33. [PMID: 21945289 DOI: 10.1016/j.neuropharm.2011.09.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 09/11/2011] [Accepted: 09/12/2011] [Indexed: 12/25/2022]
Abstract
Hyperactivity of the Hypthalamus-Pituitary-Adrenal (HPA)-axis is common in major depression and evident from e.g., a frequently exaggerated response to combined application of dexamethasone and CRH in this disorder. HPA-axis activity and hence the secretion of glucocorticoids (GC), the endpoint of the HPA-axis, depends to some extent on GC binding to glucocorticoid receptors (GR) that are abundantly expressed in the hippocampus. To assess whether differences in hippocampal GR expression occur in association with depression, we investigated GR-alpha protein immunoreactivity (ir) in postmortem hippocampal tissue of an elderly cohort of 9 well-characterized depressed patients and 9 control subjects that were pair-wise matched for age, sex, CSF-pH and postmortem delay. Abundant nuclear GR-ir was observed in neurons of the hippocampal Ammon's horn (CA) and dentate gyrus (DG) subregions. GR-ir in the DG correlated positively with age in the depressed but not the control group. Although no significant differences were found in GR-ir between the depressed and control groups, a significant increase in GR-ir was present in depressed females compared to depressed males. Whether this sex difference in hippocampal GR-ir in depression relates to the increased incidence of depression in females awaits further study. This article is part of a Special Issue entitled 'Anxiety and Depression'.
Collapse
|
37
|
Aiello TP, Whitaker-Azmitia PM. Sexual differentiation and the neuroendocrine hypothesis of autism. Anat Rec (Hoboken) 2011; 294:1663-70. [PMID: 21901838 DOI: 10.1002/ar.21251] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 05/25/2010] [Indexed: 11/09/2022]
Abstract
The phenotypic expression of autism spectrum disorders varies widely in severity and characteristics and it is, therefore, likely that a number of etiological factors are involved. However, one finding which has been found consistently is that there is a greater incidence of autism in boys than girls. Recently, attention has been given to the extreme male hypothesis-that is that autism behaviors are an extreme form of typical male behaviors, including lack of empathy and language deficits but an increase in so-called systemizing behaviors, such as attention to detail and collecting. This points to the possibility that an alteration during sexual differentiation of the brain may occur in autism. During sexual differentiation of the brain, two brain regions are highly sexually dimorphic-the amygdala and the hypothalamus. Both of these regions are also implicated in the neuroendocrine hypothesis of autism, wherein a balance between oxytocin and cortisol may contribute to the disorder. We are thus proposing that the extreme male hypothesis and the neuroendocrine hypothesis are in fact compatible in that sexual differentiation of the brain towards an extreme male phenotype would result in the neuroendocrine changes proposed in autism. We have preliminary data, treating developing rat pups with the differentiating hormone 17-β estradiol during a critical time and showing changes in social behaviors and oxytocin, to support this hypothesis. Further studies should be undertaken to confirm the role of extremes of normal sexual differentiation in producing the neuroendocrine changes associated with autism.
Collapse
|
38
|
Bao AM, Swaab DF. Sexual differentiation of the human brain: relation to gender identity, sexual orientation and neuropsychiatric disorders. Front Neuroendocrinol 2011; 32:214-26. [PMID: 21334362 DOI: 10.1016/j.yfrne.2011.02.007] [Citation(s) in RCA: 206] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Revised: 02/04/2011] [Accepted: 02/14/2011] [Indexed: 11/28/2022]
Abstract
During the intrauterine period a testosterone surge masculinizes the fetal brain, whereas the absence of such a surge results in a feminine brain. As sexual differentiation of the brain takes place at a much later stage in development than sexual differentiation of the genitals, these two processes can be influenced independently of each other. Sex differences in cognition, gender identity (an individual's perception of their own sexual identity), sexual orientation (heterosexuality, homosexuality or bisexuality), and the risks of developing neuropsychiatric disorders are programmed into our brain during early development. There is no evidence that one's postnatal social environment plays a crucial role in gender identity or sexual orientation. We discuss the relationships between structural and functional sex differences of various brain areas and the way they change along with any changes in the supply of sex hormones on the one hand and sex differences in behavior in health and disease on the other.
Collapse
Affiliation(s)
- Ai-Min Bao
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China.
| | | |
Collapse
|
39
|
(Re-)activation of neurons in aging and dementia: Lessons from the hypothalamus. Exp Gerontol 2011; 46:178-84. [DOI: 10.1016/j.exger.2010.08.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 08/25/2010] [Accepted: 08/27/2010] [Indexed: 01/22/2023]
|
40
|
Bao AM, Swaab DF. Sex differences in the brain, behavior, and neuropsychiatric disorders. Neuroscientist 2011; 16:550-65. [PMID: 20889965 DOI: 10.1177/1073858410377005] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sex differences in the brain are reflected in behavior and in the risk for neuropsychiatric disorders. The fetal brain develops in the male direction due to a direct effect of testosterone on the developing neurons, or in the female direction due to the absence of such a testosterone surge. Because sexual differentiation of the genitals takes place earlier in intrauterine life than sexual differentiation of the brain, these two processes can be influenced independently of each other. Gender identity (the conviction of belonging to the male or female gender), sexual orientation (heterosexuality, homosexuality, or bisexuality), pedophilia, sex differences in cognition, and the risks for neuropsychiatric disorders are programmed into our brains during early development. There is no proof that postnatal social environment has any crucial effect on gender identity or sexual orientation. Structural and functional sex differences in brain areas, together with changes in sex hormone levels and their receptors in development and adulthood, are closely related to sex differences in behavior and neuropsychiatric disorders. Knowing that such a relationship exists may help bring about sex-specific therapeutic strategies.
Collapse
Affiliation(s)
- Ai-Min Bao
- Department of Neurobiology, Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China.
| | | |
Collapse
|
41
|
Pompili M, Serafini G, Innamorati M, Möller-Leimkühler AM, Giupponi G, Girardi P, Tatarelli R, Lester D. The hypothalamic-pituitary-adrenal axis and serotonin abnormalities: a selective overview for the implications of suicide prevention. Eur Arch Psychiatry Clin Neurosci 2010; 260:583-600. [PMID: 20174927 DOI: 10.1007/s00406-010-0108-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2009] [Accepted: 02/05/2010] [Indexed: 12/11/2022]
Abstract
Suicidal behavior and mood disorders are one of the world's largest public health problems. The biological vulnerability for these problems includes genetic factors involved in the regulation of the serotonergic system and stress system. The hypothalamic-pituitary-adrenal (HPA) axis is a neuroendocrine system that regulates the body's response to stress and has complex interactions with brain serotonergic, noradrenergic and dopaminergic systems. Corticotropin-releasing hormone and vasopressin act synergistically to stimulate the secretion of ACTH that stimulates the biosynthesis of corticosteroids such as cortisol from cholesterol. Cortisol is a major stress hormone and has effects on many tissues, including on mineralocorticoid receptors and glucocorticoid receptors in the brain. Glucocorticoids produce behavioral changes, and one important target of glucocorticoids is the hypothalamus, which is a major controlling center of the HPA axis. Stress plays a major role in the various pathophysiological processes associated with mood disorders and suicidal behavior. Serotonergic dysfunction is a well-established substrate for mood disorders and suicidal behavior. Corticosteroids may play an important role in the relationship between stress, mood changes and perhaps suicidal behavior by interacting with 5-HT1A receptors. Abnormalities in the HPA axis in response to increased levels of stress are found to be associated with a dysregulation in the serotonergic system, both in subjects with mood disorders and those who engage in suicidal behavior. HPA over-activity may be a good predictor of mood disorders and perhaps suicidal behavior via abnormalities in the serotonergic system.
Collapse
Affiliation(s)
- Maurizio Pompili
- Department of Psychiatry, Sant'Andrea Hospital, Via di Grottarossa 1035, 00189, Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
McClellan KM, Stratton MS, Tobet SA. Roles for gamma-aminobutyric acid in the development of the paraventricular nucleus of the hypothalamus. J Comp Neurol 2010; 518:2710-28. [PMID: 20506472 PMCID: PMC2879086 DOI: 10.1002/cne.22360] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The development of the hypothalamic paraventricular nucleus (PVN) involves several factors that work together to establish a cell group that regulates neuroendocrine functions and behaviors. Several molecular markers were noted within the developing PVN, including estrogen receptors (ER), neuronal nitric oxide synthase (nNOS), and brain-derived neurotrophic factor (BDNF). By contrast, immunoreactive gamma-aminobutyric acid (GABA) was found in cells and fibers surrounding the PVN. Two animal models were used to test the hypothesis that GABA works through GABA(A) and GABA(B) receptors to influence the development of the PVN. Treatment with bicuculline to decrease GABA(A) receptor signaling from embryonic day (E) 10 to E17 resulted in fewer cells containing immunoreactive (ir) ERalpha in the region of the PVN vs. control. GABA(B)R1 receptor subunit knockout mice were used to examine the PVN at P0 without GABA(B) signaling. In female but not male GABA(B)R1 subunit knockout mice, the positions of cells containing ir ERalpha shifted from medial to lateral compared with wild-type controls, whereas the total number of ir ERalpha-containing cells was unchanged. In E17 knockout mice, ir nNOS cells and fibers were spread over a greater area. There was also a significant decrease in ir BDNF in the knockout mice in a region-dependent manner. Changes in cell position and protein expression subsequent to disruption of GABA signaling may be due, in part, to changes in nNOS and BDNF signaling. Based on the current study, the PVN can be added as another site where GABA exerts morphogenetic actions in development.
Collapse
Affiliation(s)
- Kristy M. McClellan
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523
- School of Science, Buena Vista University, Storm Lake, IA 50588
| | - Matthew S. Stratton
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523
| | - Stuart A. Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523
| |
Collapse
|
43
|
Gan EH, Quinton R. Physiological Significance of the Rhythmic Secretion of Hypothalamic and Pituitary Hormones. PROGRESS IN BRAIN RESEARCH 2010; 181:111-26. [DOI: 10.1016/s0079-6123(08)81007-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
44
|
Bao AM, Swaab DF. Corticotropin-Releasing Hormone and Arginine Vasopressin in Depression. HORMONES OF THE LIMBIC SYSTEM 2010; 82:339-65. [DOI: 10.1016/s0083-6729(10)82018-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Savic I, Garcia-Falgueras A, Swaab DF. Sexual differentiation of the human brain in relation to gender identity and sexual orientation. PROGRESS IN BRAIN RESEARCH 2010; 186:41-62. [DOI: 10.1016/b978-0-444-53630-3.00004-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Veldhuis JD, Roelfsema F, Iranmanesh A, Carroll BJ, Keenan DM, Pincus SM. Basal, pulsatile, entropic (patterned), and spiky (staccato-like) properties of ACTH secretion: impact of age, gender, and body mass index. J Clin Endocrinol Metab 2009; 94:4045-52. [PMID: 19755477 PMCID: PMC2758736 DOI: 10.1210/jc.2009-1143] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Age, gender, and BMI determine ultradian modes of LH and GH secretion, viz., pulsatile, basal, pattern-defined regularity [approximate entropy (ApEn)] and spikiness (sharp, brief excursions). Whether the same determinants apply to ACTH secretion is not known. SETTING The study was conducted at a tertiary medical center. SUBJECTS We studied normal women (n = 22) and men (n = 26) [ages, 23-77 yr; body mass index (BMI), 21-32 kg/m(2)]. METHODS Volunteers underwent 10-min blood sampling to create 24-h ACTH concentration profiles. OUTCOMES Dynamic measures of ACTH secretion were studied. RESULTS Mean ACTH concentrations (R(2) = 0.15; P = 0.006) and both pulsatile (R(2) = 0.12; P = 0.018) and basal (nonpulsatile) (R(2) = 0.16; P = 0.005) ACTH secretion correlated directly with BMI (n = 48). Men had greater basal (P = 0.047), pulsatile (P = 0.031), and total (P = 0.010) 24-h ACTH secretion than women, including when total secretion was normalized for BMI (P = 0.019). In men, both ACTH-cortisol feedforward and cortisol-ACTH feedback asynchrony (cross-ApEn) increased with age (R(2) = 0.20 and 0.22; P = 0.021 and 0.018). ACTH spikiness rose with age (P = 0.046), principally in women. Irregularity of cortisol secretion (ApEn) increased with age (n = 48; P = 0.010), especially in men. In both sexes, percentage pulsatile ACTH secretion predicted 24-h mean cortisol concentrations (R(2) = 0.14; P = 0.009). CONCLUSION Valid comparisons of ultradian ACTH dynamics will require cohorts matched for age, gender, and BMI, conditions hitherto not satisfied in most physiological studies of this axis.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Endocrine Research Unit, Mayo Medical School, Mayo School of Graduate Medical Education, Center for Translational Science Activities, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Sex Hormones and Cognitive Functioning of Women. Arh Hig Rada Toksikol 2009; 60:363-74. [DOI: 10.2478/10004-1254-60-2009-1911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spolni hormoni i kognitivno funkcioniranje ženaU radu se raspravlja o organizirajućim i aktivirajućim učincima spolnih hormona te o njihovu utjecaju na kognitivno funkcioniranje. Dosadašnja su istraživanja pokazala spolne razlike u nekim specifičnim kognitivnim sposobnostima. Žene su u prosjeku bolje u verbalnoj fluentnosti, perceptivnoj brzini i točnosti, kao i finijoj motorici, dok su muškarci u prosjeku bolji u prostornim i matematičkim sposobnostima. Ove razlike u kognitivnom funkcioniranju dovode se u vezu s izlaganjem mozga fetusa različitim razinama spolnih hormona tijekom prenatalnog života. Studije na skupinama rođenim s genskim poremećajima, kao što su sindrom neosjetljivosti na androgene, kongenitalna adrenalna hiperplazija i Turnerov sindrom također upućuju na organizirajuće učinke spolnih hormona na kognitivno funkcioniranje.Nadalje, dosadašnja istraživanja pokazuju da povišene razine ženskih spolnih hormona u kasnoj folikularnoj i/ili lutealnoj fazi menstrualnog ciklusa potenciraju tipičan ženski kognitivni obrazac funkcioniranja, koji karakterizira veća učinkovitost u zadacima koje u prosjeku bolje rješavaju žene. Niske pak razine ovih hormona, koje karakteriziraju menstrualnu fazu ciklusa, potenciraju tipičan muški obrazac funkcioniranja, koji uključuje bolju učinkovitost u zadacima koje u prosjeku bolje rješavaju muškarci.U radu se također raspravlja o metodološkim razlikama u dosadašnjim istraživanjima organizirajućih i aktivirajućih učinaka spolnih hormona na kognitivno funkcioniranje, kao i o smjernicama za buduća istraživanja.
Collapse
|
48
|
Sweetser S, Camilleri M, Linker Nord SJ, Burton DD, Castenada L, Croop R, Tong G, Dockens R, Zinsmeister AR. Do corticotropin releasing factor-1 receptors influence colonic transit and bowel function in women with irritable bowel syndrome? Am J Physiol Gastrointest Liver Physiol 2009; 296:G1299-306. [PMID: 19342506 PMCID: PMC2697942 DOI: 10.1152/ajpgi.00011.2009] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Corticotropin releasing factor (CRF), a mediator of stress response, alters gastrointestinal (GI) functions. Stress-related changes in colonic motility are blocked by selective CRF(1) receptor antagonists. Our aim was to assess whether modulation of central and peripheral CRF(1) receptors affects colonic transit and bowel function in female patients with diarrhea-predominant irritable bowel syndrome (D-IBS). This randomized, double-blind, placebo-controlled, 2-wk study evaluated the effects of oral pexacerfont (BMS-562086), a selective CRF(1) receptor antagonist, 25 and 100 mg qd, on GI and colonic transit of solids [by validated scintigraphy with primary end point colonic geometric center (GC) at 24 h] and bowel function (by validated daily diaries) in 39 women with D-IBS. The 100-mg dose was comparable to a dose that inhibited colonic motility in stressed rats. Treatment effects were compared by analysis of covariance with baseline colonic transit as covariate. The study had 80% power (alpha = 0.05) to detect clinically meaningful (26%) differences in colonic transit. Thirty-nine of 55 patients fulfilled eligibility criteria (9 screen failures, 5 baseline GC24 outside prespecified range). At baseline, three treatment groups had comparable age, body mass index, and GC 24 h. Significant effects of pexacerfont relative to placebo were not detected on colonic GC24 (P = 0.53), gastric emptying, orocecal transit, ascending colon emptying half-time, and stool frequency, consistency, and ease of passage. No safety issues were identified. We conclude that in women with D-IBS, pexacerfont, 25 or 100 mg qd, does not significantly alter colonic or other regional transit or bowel function. The role of central and peripheral CRF(1) receptors in bowel function in D-IBS requires further study.
Collapse
Affiliation(s)
- Seth Sweetser
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and Department of Health Sciences Research, Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, Minnesota; and Discovery Medicine and Clinical Pharmacology Neurosciences, Bristol-Myers Squibb, Princeton, New Jersey
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and Department of Health Sciences Research, Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, Minnesota; and Discovery Medicine and Clinical Pharmacology Neurosciences, Bristol-Myers Squibb, Princeton, New Jersey
| | - Sara J. Linker Nord
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and Department of Health Sciences Research, Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, Minnesota; and Discovery Medicine and Clinical Pharmacology Neurosciences, Bristol-Myers Squibb, Princeton, New Jersey
| | - Duane D. Burton
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and Department of Health Sciences Research, Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, Minnesota; and Discovery Medicine and Clinical Pharmacology Neurosciences, Bristol-Myers Squibb, Princeton, New Jersey
| | - Lorna Castenada
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and Department of Health Sciences Research, Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, Minnesota; and Discovery Medicine and Clinical Pharmacology Neurosciences, Bristol-Myers Squibb, Princeton, New Jersey
| | - Robert Croop
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and Department of Health Sciences Research, Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, Minnesota; and Discovery Medicine and Clinical Pharmacology Neurosciences, Bristol-Myers Squibb, Princeton, New Jersey
| | - Gary Tong
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and Department of Health Sciences Research, Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, Minnesota; and Discovery Medicine and Clinical Pharmacology Neurosciences, Bristol-Myers Squibb, Princeton, New Jersey
| | - Randy Dockens
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and Department of Health Sciences Research, Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, Minnesota; and Discovery Medicine and Clinical Pharmacology Neurosciences, Bristol-Myers Squibb, Princeton, New Jersey
| | - Alan R. Zinsmeister
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) and Department of Health Sciences Research, Division of Biostatistics, College of Medicine, Mayo Clinic, Rochester, Minnesota; and Discovery Medicine and Clinical Pharmacology Neurosciences, Bristol-Myers Squibb, Princeton, New Jersey
| |
Collapse
|
49
|
Tobet S, Knoll JG, Hartshorn C, Aurand E, Stratton M, Kumar P, Searcy B, McClellan K. Brain sex differences and hormone influences: a moving experience? J Neuroendocrinol 2009; 21:387-92. [PMID: 19207813 PMCID: PMC2669491 DOI: 10.1111/j.1365-2826.2009.01834.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Sex differences in the nervous system come in many forms. Although a majority of sexually dimorphic characteristics in the brain have been described in older animals, mechanisms that determine sexually differentiated brain characteristics often operate during critical perinatal periods. Both genetic and hormonal factors likely contribute to physiological mechanisms in development to generate the ontogeny of sexual dimorphisms in brain. Relevant mechanisms may include neurogenesis, cell migration, cell differentiation, cell death, axon guidance and synaptogenesis. On a molecular level, there are several ways to categorize factors that drive brain development. These range from the actions of transcription factors in cell nuclei that regulate the expression of genes that control cell development and differentiation, to effector molecules that directly contribute to signalling from one cell to another. In addition, several peptides or proteins in these and other categories might be referred to as 'biomarkers' of sexual differentiation with undetermined functions in development or adulthood. Although a majority of sex differences are revealed as a direct consequence of hormone actions, some may only be revealed after genetic or environmental disruption. Sex differences in cell positions in the developing hypothalamus, and steroid hormone influences on cell movements in vitro, suggest that cell migration may be one target for early molecular actions that impact brain development and sexual differentiation.
Collapse
Affiliation(s)
- S Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Bibliography. Current world literature. Adrenal cortex. Curr Opin Endocrinol Diabetes Obes 2008; 15:284-299. [PMID: 18438178 DOI: 10.1097/med.0b013e3283040e80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|